US20120114712A1 - Umbilical cord biomaterial for medical use - Google Patents

Umbilical cord biomaterial for medical use Download PDF

Info

Publication number
US20120114712A1
US20120114712A1 US13/333,172 US201113333172A US2012114712A1 US 20120114712 A1 US20120114712 A1 US 20120114712A1 US 201113333172 A US201113333172 A US 201113333172A US 2012114712 A1 US2012114712 A1 US 2012114712A1
Authority
US
United States
Prior art keywords
biomaterial
umbilical cord
membrane
stem cell
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/333,172
Inventor
Qing Liu
Henry Rendon Barragan
George Matcham
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Celularity Inc
Original Assignee
Anthrogenesis Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Anthrogenesis Corp filed Critical Anthrogenesis Corp
Priority to US13/333,172 priority Critical patent/US20120114712A1/en
Publication of US20120114712A1 publication Critical patent/US20120114712A1/en
Assigned to HLI CELLULAR THERAPEUTICS, LLC, A DELAWARE LIMITED LIABILITY COMPANY reassignment HLI CELLULAR THERAPEUTICS, LLC, A DELAWARE LIMITED LIABILITY COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANTHROGENESIS CORPORATION, A NEW JERSEY CORPORATION, D/B/A CELGENE CELLULAR THERAPEUTICS
Assigned to CELULARITY BIOSOURCING, LLC reassignment CELULARITY BIOSOURCING, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: HLI CELLULAR THERAPEUTICS, LLC
Assigned to Celularity, Inc. reassignment Celularity, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CELULARITY BIOSOURCING, LLC
Abandoned legal-status Critical Current

Links

Images

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
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body 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
    • A61L27/3604Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix characterised by the human or animal origin of the biological material, e.g. hair, fascia, fish scales, silk, shellac, pericardium, pleura, renal tissue, amniotic membrane, parenchymal tissue, fetal tissue, muscle tissue, fat tissue, enamel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/3683Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix subjected to a specific treatment prior to implantation, e.g. decellularising, demineralising, grinding, cellular disruption/non-collagenous protein removal, anti-calcification, crosslinking, supercritical fluid extraction, enzyme treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • A61L27/3834Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • 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
    • A61L2430/06Materials or treatment for tissue regeneration for cartilage reconstruction, e.g. meniscus

Definitions

  • the present invention generally relates to biomaterials derived from the umbilical cord membrane, compositions comprising the umbilical cord membrane, and methods of treatment using the compositions.
  • the present invention provides biomaterials derived from umbilical cord.
  • the biomaterial comprises an umbilical cord membrane.
  • the present invention provides methods of making the umbilical cord biomaterial, and of using the biomaterial, e.g., to repair organs and tissues.
  • the present invention provides a biomaterial comprising an isolated mammalian umbilical cord membrane.
  • the biomaterial comprises at least one umbilical vessel (e.g., an umbilical artery or umbilical vein).
  • the biomaterial comprises Wharton's jelly.
  • the biomaterial comprises Wharton's jelly but lacks umbilical vessels.
  • the biomaterial comprises the umbilical cord membrane, Wharton's jelly, and all three umbilical vessels.
  • the umbilical cord biomaterial is not, however, an umbilical cord that has not been processed in any manner.
  • the biomaterial comprises less water by weight than native umbilical cord membrane in vivo, e.g., the biomaterial comprises 40%, 30%, 20%, 10% or less water by weight. In other more specific embodiments, the biomaterial comprises at least 60%, at least 70%, or at least 80% water by weight. In another specific embodiment, the biomaterial is decellularized prior to use. In another specific embodiment, the biomaterial is not decellularized prior to use. In another specific embodiment, the umbilical cord membrane of the biomaterial is cut or slit longitudinally. In another specific embodiment, the biomaterial is substantially flat. In another specific embodiment, the biomaterial is substantially tubular. In another specific embodiment, the biomaterial comprises artificially crosslinked proteins.
  • the umbilical cord biomaterial comprises an exogenous bioactive molecule, that is, a bioactive molecule not obtained from the umbilical cord used to make the biomaterial.
  • said bioactive molecule is a cytokine or growth factor.
  • said bioactive molecule is an extracellular matrix protein.
  • said extracellular matrix protein is collagen, fibronectin, elastin, vitronectin, or hyaluronic acid.
  • said bioactive molecule is hyaluronic acid.
  • said hyaluronic acid is crosslinked to said umbilical cord membrane.
  • said biomaterial comprises an exogenous polymer.
  • said exogenous polymer is a synthetic biodegradable polymer or an anionic polymer.
  • said synthetic biodegradable polymer is a polyhydroxyalkanoate.
  • said anionic polymer is dextran sulfate or pentosan polysulfate.
  • the bioactive molecule is an antibiotic, a hormone, a growth factor, an anti-tumor agent, an anti-fungal agent, an anti-viral agent, a pain medication, an anti-histamine, an anti-inflammatory agent, an anti-infective agent, a wound healing agent, a wound sealant, a cellular attractant or a scaffolding reagent.
  • the bioactive molecule is a small molecule, e.g., a small organic molecule, e.g., a drug.
  • the umbilical cord biomaterial comprises a hydrogel composition.
  • said hydrogel composition comprises a polyvinyl alcohol, a polyethylene glycol, a hyaluronic acid, a dextran, or a derivative or analog thereof.
  • the umbilical cord biomaterial can also comprise, e.g., be seeded with, one or more types of stem and/or progenitor cells.
  • the biomaterial comprises an exogenous stem cell, that is, a stem cell not native to the umbilical cord from which the biomaterial is derived, or from an individual different from the umbilical cord donor.
  • the stem cell is a placental stem cell, a mesenchymal stem cell, an embryonic stem cell, an adult stem cell, or a somatic stem cell.
  • the somatic stem cell is a neural stem cell, a hepatic stem cell, a pancreatic stem cell, an endothelial stem cell, a cardiac stem cell, a stromal cell, or a muscle stem cell.
  • the umbilical cord biomaterial comprises an exogenous adult (e.g., fully-differentiated or committed progenitor) cell.
  • the invention provides laminates comprising the umbilical cord biomaterial.
  • Such laminates can comprise layers of umbilical cord biomaterial, or one or more layers of umbilical cord biomaterial layered with one or more layers of another material.
  • the layers can be substantially aligned, or can be offset, e.g., overlapping, to form, e.g., a laminate of biomaterial longer and/or wider than an umbilical cord.
  • the invention provides a laminate comprising a plurality (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) of layers, wherein at least one of the layers comprises an umbilical cord biomaterial.
  • said laminate comprises one or more layers of an amniotic membrane material.
  • said amniotic membrane material is a dried amniotic membrane laminated to said umbilical cord membrane.
  • said amniotic membrane material has been sonicated or otherwise disrupted.
  • at least some of the proteins in at least one of the layers of the laminate are artificially crosslinked, either to other proteins in the same layer, or to proteins in one or more adjoining layers.
  • the invention further provides laminates comprising the umbilical cord biomaterial and other useful compounds.
  • the invention provides a laminate comprising umbilical cord biomaterial and an exogenous stem cell or an exogenous adult cell.
  • the laminate comprises a hydrogel composition.
  • said hydrogel composition comprises a polyvinyl alcohol, a polyethylene glycol, a hyaluronic acid, a dextran, or a derivative or analog thereof.
  • the invention also provides a method of producing a biomaterial comprising isolating and decellularizing a biomaterial comprising an umbilical cord membrane.
  • the biomaterial comprises Wharton's jelly, or one or more umbilical cord vessels.
  • the method comprises isolating said umbilical cord membrane from one or more umbilical cord vessels.
  • the method may further comprise drying said biomaterial to less than 20% water by weight.
  • said biomaterial is dried at about 26° C. to about 65° C., or at about 35° C. to about 50° C.
  • the composition can, for example, be dried with a hygroscopic compound.
  • the biomaterial is freeze dried, or dried using vacuum.
  • Said decellularizing may, for example, comprise contacting the umbilical cord membrane with a detergent solution, for example, a solution comprising 0.01-2.0% deoxycholic acid.
  • the detergent may be a nonionic detergent, an anionic detergent, or a combination thereof.
  • said detergent is Triton X-100 or sodium dodecyl sulfate, or a combination thereof.
  • the invention further provides methods of making a laminate comprising an umbilical cord membrane biomaterial.
  • a method can comprise crosslinking at least some proteins in at least one layer of said laminate.
  • the method of making the laminate comprises layering a plurality of umbilical cord membranes in contact with each other to form a laminate.
  • one or more of said membranes comprise less than 20% water by weight prior to said layering.
  • said laminate is dried to less than 20% water by weight after said layering.
  • one or more of said membranes comprise at least 60%, at least 70% or at least 80% water by weight.
  • said laminate comprises at least 60%, at least 70%, or at least 80% water by weight.
  • the invention also provides for the use of the umbilical cord membrane biomaterial to deliver one or more therapeutic agents to a subject.
  • the invention provides a method of delivering a therapeutic agent to a subject comprising contacting the subject with an umbilical cord membrane biomaterial, or laminate thereof, wherein said composition or laminate comprises a therapeutic agent.
  • said subject is a human.
  • said therapeutic agent is an antibiotic, an anti-cancer agent, an anti-bacterial agent, an anti-viral agent, a vaccine, an anesthetic, an analgesic, an anti-asthmatic agent, an anti-inflammatory agent, an anti-depressant, an anti-diabetic agent, an anti-psychotic, a central nervous system stimulant, a hormone, an immunosuppressant, a muscle relaxant, or a prostaglandin.
  • the invention further provides a method of using an umbilical cord biomaterial, e.g., one that comprises an isolated umbilical cord membrane, in the repair of a tympanic membrane deformity, comprising contacting said tympanic membrane with such a biomaterial.
  • the deformity is a perforation, which may be, e.g., a central perforation or a marginal perforation.
  • the perforation may be caused by, e.g., trauma, or as part of a surgical procedure.
  • said contacting is sufficient to occlude the perforation.
  • said perforation has not healed spontaneously within two months of developing the perforation.
  • said deformity is an atelectatic tympanic membrane, a deformity relating to a choleastoma, a retraction pocket or a deformity resulting from a tympanosclerosis.
  • the invention also provides for the repair of other deformities using the umbilical cord membrane biomaterial.
  • the invention provides a method of repairing a nasal septum having a perforation, comprising contacting said septum with a biomaterial comprising an umbilical cord biomaterial, e.g., one comprising an isolated mammalian umbilical cord membrane.
  • said method comprises contacting cartilage within said septum with said biomaterial.
  • exogenous bioactive compound means a molecule introduced into the biomaterial that has a detectable effect on one or more biological systems. Examples of bioactive compounds are listed, without limitation, in Section 5.1.2, below.
  • substantially flat in reference to umbilical cord biomaterial, means that the majority of the umbilical cord biomaterial is planar, but can comprise differences in thickness, whether naturally-occurring or artificially-induced, e.g., natural variations in membrane thickness, ridges, patterns, raised areas, warps, and the like, induced, e.g., during drying on a mesh.
  • substantially tubular in reference to umbilical cord biomaterial, means that the majority of the umbilical cord biomaterial is tubular (i.e., circular, ovoid, or irregular in cross-section, and comprising the material defining an interior or lumen).
  • the substantially tubular biomaterial can be a closed tube or partially open tube.
  • umbilical cord biomaterial means any biomaterial comprising an isolated umbilical cord outer membrane, particularly a biomaterial manufactured or derived from umbilical cord, and includes, e.g., dried whole umbilical cord, dried umbilical cord membrane (with or without vessels), umbilical cord membrane laminated with a second material, etc.
  • the term does not, however, encompass an umbilical cord that has not been treated or manipulated in any manner, that is, an umbilical cord that has not been modified from an in vivo state.
  • FIGS. 1A and 1B depict the effect of radiation dose on water uptake [ ⁇ , (Ww ⁇ Wd)/Wd*100] and equilibrium water content [ ⁇ , (Ww ⁇ Wd)/Ww*100] for human umbilical cord biomaterial incubated for 10 (A) and 20 (B) days in 1% D-cell solution.
  • FIGS. 2A and 2B depict a comparison of the change in thickness ( ⁇ ) and water uptake ( ⁇ ) during rehydration of gamma sterilized human umbilical cord biomaterial incubated for 10 (A) and 20 (B) days in 1% D-cell solution. Error bars indicate standard deviation.
  • FIG. 3 depicts a comparison of the denaturation temperature of rehydrated human umbilical cord membrane that had previously been incubated in 1% D-cell solution for 10 or 20 days. There is essentially no difference between the different incubation times and there is a linear decrease in denaturation temperature with increasing radiation dose.
  • FIG. 4 depicts a schematic of tensile testing of membrane samples based on American Society for Testing and Materials (ASTM) standard D1708.
  • a dog bone shaped sample 1 is mounted in a rectangular support of vellum paper.
  • the membrane and the support are incubated in PBS for 1 hour at 37° C.
  • the support keeps the membrane flat and eases loading into the mechanical tester.
  • the tester comprises and upper grip 2 and a lower grip 3 . Once the support and membrane have been secured in the grips of the mechanical tester with glue 4 , the supporting struts 5 are cut and the sample can be tested with no interference from the vellum support.
  • FIG. 5A depicts a suture pull-out assay apparatus with an lower grip 10 holding vellum paper 11 to which a 1 ⁇ 2 cm section of umbilical cord biomaterial 12 is glued, and an upper grip 13 attached to a suture 14 that passes through the umbilical cord biomaterial. Force was applied upwards through the suture at approximately 12.7 mm/min.
  • FIG. 5B depicts results of a comparison of the human umbilical cord biomaterial (HUC) and dried human amniotic membrane pull-out resistance in Newtons (N).
  • HUC human umbilical cord biomaterial
  • N dried human amniotic membrane pull-out resistance in Newtons
  • the umbilical cord biomaterial of the invention is derived from a mammalian umbilical cord or part thereof that comprises an umbilical cord membrane (that is, the outer membrane of the umbilical cord).
  • the umbilical cord is a substantially tubular organ, typically 10-15 cm in length, that connects the fetus to the placenta and houses the umbilical vessels.
  • the umbilical cord comprises an outer membrane that wraps around two umbilical arteries and one umbilical vein, which are contained within a ground substance known as Wharton's jelly.
  • the main components of Wharton's jelly are proteoglycans. Wharton's jelly also contains large, stellate fibroblasts and macrophages.
  • the umbilical cord membrane biomaterial of the invention typically comprises only the umbilical cord membrane, but can also comprise Wharton's jelly and/or one or more of the umbilical vessels.
  • the umbilical cord biomaterial is an umbilical cord membrane substantially isolated from the remaining umbilical cord components (e.g., Wharton's jelly and umbilical vessels).
  • the umbilical cord biomaterial comprises an umbilical cord membrane and Wharton's jelly (that is, the ground material in which the umbilical cord vessels are contained in the intact umbilical cord) that are isolated from the remaining umbilical cord components (e.g., umbilical cord vessels).
  • the umbilical cord membrane biomaterial comprises the membrane, Wharton's jelly and one or more umbilical cord vessels.
  • the umbilical cord biomaterial comprises an isolated umbilical cord (e.g., comprising Wharton's jelly and vessels, Wharton's jelly only, or vessels only) that has been flattened into a sheet or strip.
  • the umbilical cord membrane biomaterial can be a substantially tubular structure from which the contents (Wharton's jelly and vessels) have been removed.
  • the biomaterial can also comprise an umbilical cord membrane that has been slit or cut for part or all of the length of the umbilical cord to expose the contents of the umbilical cord.
  • the biomaterial comprising umbilical cord membrane and/or Wharton's jelly and/or vessels
  • the biomaterial can be decellularized.
  • the biomaterial comprises umbilical cord membrane-associated cells or Wharton's jelly-associated cells that have been killed.
  • the biomaterial comprises umbilical cord membrane-associated cells or Wharton's jelly-associated cells that have been maintained in a living state.
  • the umbilical cord biomaterial of the invention can be derived from the umbilical cord of any mammal, for example, from equine, bovine, porcine or catarrhine sources, but is most preferably derived from human umbilical cord.
  • the umbilical cord biomaterial is preferably dry or substantially dry.
  • the umbilical cord biomaterial is substantially dry, i.e., is 20% or less water by weight.
  • the umbilical cord biomaterial can be substantially flat.
  • the dry biomaterial may, in another embodiment, substantially retain the shape of the native umbilical cord, that is, the dry membrane may be substantially tubular.
  • the umbilical cord biomaterial can also be shaped to assume different conformations, e.g., can be curved, cut, molded, or the like, to fit to a part of the body.
  • the umbilical cord biomaterial has not been protease-treated, heat-denatured or artificially (e.g., chemically or radiologically) crosslinked.
  • the umbilical cord biomaterial comprises artificially crosslinked proteins, e.g., chemically or radiologically crosslinked collagen.
  • the umbilical cord biomaterial contains substantially no structural proteins that are artificially crosslinked.
  • the umbilical cord biomaterial is not fixed.
  • a preferred umbilical cord biomaterial is produced by the methods disclosed herein (see Section 5.1.4, below, and Examples 1 and 2).
  • a single layer of the acellular, dried umbilical cord biomaterial is approximately 50 microns to 250 microns in thickness, typically approximately 90 microns to 220 microns in thickness. In other specific embodiments, a single layer of the umbilical cord biomaterial is approximately 75-200 microns, 100-200 microns, 100-220 microns, 120-220 microns, or 150-250 microns in thickness in the dry state. In another embodiment, the average thickness of the umbilical cord biomaterial is about 157 microns (e.g., ⁇ 20%). In another specific embodiment, the pull out strength of the dried umbilical cord biomaterial is approximately 1.5 Newtons (N), compared to a pull out strength of a dried amniotic membrane material at approximately 0.4N.
  • N Newtons
  • the umbilical cord biomaterial is sided, that is, the umbilical cord biomaterial comprises an epithelial side (from the interior of the umbilical cord), and an outer, mesothelial side (from the exterior of the umbilical cord).
  • the umbilical cord biomaterial is non-immunogenic.
  • the umbilical cord biomaterial comprises particular cytokines, i.e., interleukin (IL)-1b, IL-2, IL-3, IL-6, IL-7, IL-12, IL-15, IFN- ⁇ , MIP-1b, and/or MCP-1.
  • cytokines i.e., interleukin (IL)-1b, IL-2, IL-3, IL-6, IL-7, IL-12, IL-15, IFN- ⁇ , MIP-1b, and/or MCP-1.
  • the umbilical cord biomaterial is translucent. In other embodiments, the umbilical cord biomaterial is opaque, or is colored or dyed, e.g., permanently colored or dyed, using a medically-acceptable dyeing or coloring agent. Such an agent may be adsorbed onto the biomaterial, or the biomaterial may be impregnated or coated with such an agent. In this embodiment, any known non-toxic, non-irritating coloring agent or dye may be used.
  • the umbilical cord biomaterial comprises of collagen (types I, III and IV; typically about 75%-80%% of the matrix of the biomaterial), fibronectin, elastin, and may further comprise glycosaminoglycans, (GAGs, e.g., hyaluronic acid) and/or proteoglycans.
  • GAGs glycosaminoglycans
  • laminin is not present, or is present in trace amounts (i.e., less than 0.1% of the dry weight of the biomaterial).
  • the umbilical cord biomaterial comprises collagen types I, III, IV, V, VI and VII.
  • the umbilical cord biomaterial can comprise non-structural components, such as, for example, one or more growth factors, e.g., platelet-derived growth factors (PDGFs), vascular-endothelial growth factor (VEGF), fibroblast growth factor (FGF), transforming growth factor- ⁇ 1, and the like.
  • the umbilical cord biomaterial comprises growth factors such as FGF, b-FGF, EGF, IGF-1, PDGF and TGF- ⁇ .
  • the composition of the umbilical cord biomaterial may thus be ideally suited to encourage the migration of fibroblasts and macrophages, and thus, e.g., the promotion of wound healing.
  • the invention provides an umbilical cord biomaterial wherein at least 50% of the dry weight of the biomaterial is collagen I. In various more specific embodiments, at least 55%, 60%, 65% or 70% of the dry weight of the biomaterial is collagen I. In another specific embodiment, the invention provides an umbilical cord biomaterial wherein at most 5% of the dry weight of the biomaterial is collagen III. In various more specific embodiments, at most 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4.0%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3.0% or 2.9% of the dry weight of the biomaterial is collagen III.
  • the invention provides an umbilical cord biomaterial wherein at least 4% of the dry weight of the biomaterial is collagen IV. In various more specific embodiments, at least 5%, 6%, 7%, 8%, 9%, 10% or 11% of the dry weight of said biomaterial is collagen IV. In another specific embodiment, the invention provides an umbilical cord biomaterial wherein at most 4% of the dry weight of the biomaterial is elastin. In various more specific embodiments, at most 3.8%, 3.6%, 3.4%, 3.2%, 3.0%, 2.8%, 2.6%, 2.4%, 2.2%, 2.0%, or 1.8% of the dry weight of the biomaterial is elastin. In another specific embodiment, at least 4% of the dry weight of the biomaterial is glycosaminoglycan. In various more specific embodiments, at least 4.1%, 4.2%, 4.3%, 4.4%, 4.5% or 4.6% of the dry weight of said biomaterial is glycosaminoglycan.
  • the umbilical cord biomaterial may be used in a single-layered format, for example, as a single-layer sheet or an un-laminated membrane.
  • the umbilical cord biomaterial may be used in a double-layer or multiple-layer format, e.g., the umbilical cord biomaterial may be laminated. Lamination can provide greater stiffness and durability, for example, during the healing process.
  • the umbilical cord biomaterial may be, for example, laminated as described below (see Section 5.1.7).
  • the umbilical cord biomaterial may further comprise collagen from a non-umbilical cord source.
  • one or more layers of umbilical cord biomaterial may comprise, e.g., be coated or impregnated with, or layered with, purified extracted collagen.
  • Such collagen may be obtained, for example, from commercial sources, or may be produced according to known methods, such as those disclosed in U.S. Pat. Nos. 4,420,339, 5,814,328, and 5,436,135, the disclosures of which are hereby incorporated by reference.
  • Such collagen can also be obtained from a placental source, including a placenta obtained from the same donor as the umbilical cord biomaterial.
  • the umbilical cord biomaterial can comprise one or more compounds or substances that are not present in the umbilical cord material from which the biomaterial is derived. Moreover, the umbilical cord biomaterial can comprise non-naturally-occurring amounts of one or more compounds or substances that are normally present in the umbilical cord from which the biomaterial is derived.
  • the umbilical cord biomaterial can comprise, e.g., can be impregnated with, a bioactive compound, such as those listed in Section 5.1.2, below.
  • bioactive compounds include, but are not limited to, small organic molecules (e.g., drugs), antibiotics (such as, for example, Clindamycin, Minocycline, Doxycycline, Gentamycin), hormones, growth factors, anti-tumor agents, anti-fungal agents, anti-viral agents, pain medications, anti-histamines, anti-inflammatory agents, anti-infectives including but not limited to silver (such as silver salts, including but not limited to silver nitrate and silver sulfadiazine), elemental silver, antibiotics, bactericidal enzymes (such as lysozyme), wound healing agents (such as cytokines including but not limited to PDGF, TGF; thymosin), hyaluronic acid as a wound healing agent, wound sealants (such as fibrin with or without thrombin), cellular attractant and scaffolding reagents (such as added fibronectin) and the like.
  • drugs such as, for example, Clindamycin, Minocycline, D
  • the umbilical cord biomaterial may be impregnated with at least one growth factor, for example, fibroblast growth factor, epithelial growth factor, etc.
  • the biomaterial may also be impregnated with small organic molecules such as specific inhibitors of particular biochemical processes e.g., membrane receptor inhibitors, kinase inhibitors, growth inhibitors, anticancer drugs, antibiotics, etc.
  • Impregnating the umbilical cord biomaterial with a bioactive compound may be accomplished, e.g., by immersing the biomaterial in a solution of the bioactive compound of the desired concentration for a time sufficient to allow the biomaterial to absorb and to equilibrate with the solution.
  • the biomaterial so impregnated is a dried biomaterial, and the solution partially or fully re-hydrates the biomaterial, compared to an umbilical cord or umbilical cord membrane in vivo.
  • the biomaterial is impregnated prior to drying the biomaterial, e.g., to substantial dryness.
  • the umbilical cord biomaterial may be combined with a hydrogel to form a composite.
  • a hydrogel composition known to one skilled in the art is encompassed within the invention, e.g., any of the hydrogel compositions disclosed in the following reviews: Graham, 1998, Med. Device Technol. 9 (1): 18-22; Peppas et al., 2000, Eur. J. Pharm. Biopharm. 50 (1): 27-46; Nguyen et al., 2002, Biomaterials, 23 (22): 4307-14; Henincl et al., 2002, Adv. Drug Deliv. Rev 54 (1): 13-36; Skelhome et al., 2002, Med. Device. Technol.
  • the hydrogel composition is applied on the umbilical cord biomaterial, that is, is disposed on the surface of the biomaterial.
  • the hydrogel composition for example, may be sprayed onto the umbilical cord biomaterial or coated onto the surface of the biomaterial, or the biomaterial may, for example, be soaked, bathed or saturated with the hydrogel composition.
  • the hydrogel is sandwiched between two or more layers of umbilical cord biomaterial.
  • the hydrogel is sandwiched between two layers of umbilical cord biomaterial, wherein the edges of the two layers of biomaterial are sealed so as to substantially or completely contain the hydrogel.
  • hydrogels useful in the methods and compositions of the invention can be made from any water-interactive, or water soluble polymer known in the art, including but not limited to, polyvinylalcohol (PVA), polyhydroxyehthyl methacrylate, polyethylene glycol, polyvinyl pyrrolidone, hyaluronic acid, alginate, collagen, gelatin, dextran or derivatives and analogs thereof.
  • PVA polyvinylalcohol
  • polyethylene glycol polyethylene glycol
  • polyvinyl pyrrolidone polyvinyl pyrrolidone
  • hyaluronic acid alginate
  • collagen collagen
  • gelatin dextran or derivatives and analogs thereof.
  • a composition comprises an umbilical cord biomaterial of the invention, one or more bioactive compounds and a hydrogel.
  • a composition comprises an umbilical cord biomaterial and a hydrogel composition that comprises one or more bioactive compounds.
  • a composition comprises an umbilical cord biomaterial comprising one or more bioactive compounds and a hydrogel composition comprising one or more bioactive compounds.
  • the bioactive compounds can be, for example, one or more compounds as described in Section 5.1.2, below.
  • the umbilical cord biomaterial of the invention can comprise (e.g., be impregnated with or coated with) one or more bioactive or medicinal compounds, such as small organic molecules (e.g., drugs), antibiotics, antiviral agents, antimicrobial agents, anti-inflammatory agents, antiproliferative agents, cytokines, enzyme or protein inhibitors, antihistamines, and the like.
  • bioactive or medicinal compounds such as small organic molecules (e.g., drugs), antibiotics, antiviral agents, antimicrobial agents, anti-inflammatory agents, antiproliferative agents, cytokines, enzyme or protein inhibitors, antihistamines, and the like.
  • the umbilical cord biomaterial may be coated or impregnated with antibiotics (such as Clindamycin, Minocycline, Doxycycline, Gentamycin), hormones, growth factors, anti-tumor agents, anti-fungal agents, anti-viral agents, pain medications (including XYLOCAINE®, Lidocaine, Procaine, Novocaine, etc.), antihistamines (e.g., diphenhydramine, BENADRYL®, etc.), anti-inflammatory agents, anti-infectives including but not limited to silver (such as silver salts, including but not limited to silver nitrate and silver sulfadiazine), elemental silver, antibiotics, bactericidal enzymes (such as lysozome), wound healing agents (such as cytokines including but not limited to PDGF (e.g., REGRANEX®), TGF; thymosin), hyaluronic acid as a wound healing agent, wound sealants (such as fibrin with or without
  • the umbilical cord biomaterial, or composites comprising umbilical cord biomaterial may comprise any of the compounds listed herein, without limitation, individually or in any combination. Any of the biologically active compounds listed herein may be formulated by known methods for immediate release or extended release. Additionally, the umbilical cord biomaterial may comprise two or more biologically active compounds in different manners; e.g., the biomaterial may be impregnated with one biologically active compound and coated with another. In another embodiment, the umbilical cord biomaterial comprises one biologically active compound formulated for extended release, and a second biologically active compound formulated for immediate release.
  • the umbilical cord biomaterial may comprise, e.g., be impregnated or coated with, a physiologically-available form of one or more nutrients required for wound healing.
  • the nutrient is formulated for extended release.
  • the umbilical cord biomaterial, or composite comprising umbilical cord biomaterial may comprise an antibiotic.
  • the antibiotic is a macrolide (e.g., tobramycin (TOBI®)), a cephalosporin (e.g., cephalexin (KEFLEX®)), cephradine (VELOSEF®)), cefuroxime (CEFTIN®, cefprozil (CEFZIL®), cefaclor (CECLOR®), cefixime (SUPRAX® or cefadroxil (DURICEF®), a clarithromycin (e.g., clarithromycin (Biaxin)), an erythromycin (e.g., erythromycin (EMYCIN®)), a penicillin (e.g., penicillin V (V-CILLINK® or PEN VEEK®)) or a quinolone (e.g., ofloxacin (FLOXIN®), ciprofloxacin (CIPRO®) or
  • the umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial may comprise, e.g., be coated or impregnated with, an antifungal agent.
  • Suitable antifungal agents include but are not limited to amphotericin B, itraconazole, ketoconazole, fluconazole, intrathecal, flucytosine, miconazole, butoconazole, clotrimazole, nystatin, terconazole, tioconazole, ciclopirox, econazole, haloprogrin, naftifine, terbinafine, undecylenate, and griseofuldin.
  • the umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial may comprise, e.g., be coated or impregnated with, an anti-inflammatory agent.
  • anti-inflammatory agents include, but are not limited to, non-steroidal anti-inflammatory drugs such as salicylic acid, acetylsalicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, acetaminophen, indomethacin, sulindac, etodolac, mefenamic acid, meclofenamate sodium, tolmetin, ketorolac, dichlofenac, ibuprofen, naproxen, naproxen sodium, fenoprofen, ketoprofen, flurbinprofen, oxaprozin, piroxicam, meloxicam, ampiroxicam, droxicam, pivoxicam, tenoxicam, na
  • the umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial may comprise, e.g., be coated or impregnated with, an antiviral agent.
  • antiviral agents include, but are not limited to, nucleoside analogs, such as zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin, as well as foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, and the alpha-interferons.
  • the umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial comprises, e.g., may be coated or impregnated with, a cytokine receptor modulator.
  • cytokine receptor modulators include, but are not limited to, soluble cytokine receptors (e.g., the extracellular domain of a TNF- ⁇ receptor or a fragment thereof, the extracellular domain of an IL-10 receptor or a fragment thereof, and the extracellular domain of an IL-6 receptor or a fragment thereof), cytokines or fragments thereof (e.g., interleukin (IL)-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-15, TNF- ⁇ , TNF- ⁇ , interferon (IFN)- ⁇ , IFN- ⁇ , IFN- ⁇ , and GM-CSF), anti-cytokine receptor antibodies (e.g., anti-IFN receptor antibodies,
  • a cytokine receptor modulator is IL-4, IL-10, or a fragment thereof.
  • a cytokine receptor modulator is an anti-IL-1 antibody, anti-IL-6 antibody, anti-IL-12 receptor antibody, or anti-TNF- ⁇ antibody.
  • a cytokine receptor modulator is the extracellular domain of a TNF- ⁇ receptor or a fragment thereof. In certain embodiments, a cytokine receptor modulator is not a TNF- ⁇ antagonist.
  • proteins, polypeptides or peptides (including antibodies) that are utilized as immunomodulatory agents are derived from the same species as the recipient of the proteins, polypeptides or peptides so as to reduce the likelihood of an immune response to those proteins, polypeptides or peptides.
  • the proteins, polypeptides, or peptides that are utilized as immunomodulatory agents are human or humanized.
  • the umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial may also comprise, e.g., be coated or impregnated with. a cytokine.
  • cytokines include, but are not limited to, colony stimulating factor 1 (CSF-1), interleukin-2 (IL-2), interleukin-3 (IL-3), interleukin-4 (IL-4), interleukin-5 (IL-5), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-9 (IL-9), interleukin-10 (IL-10), interleukin-12 (IL-12), interleukin 15 (IL-15), interleukin 18 (IL-18), insulin-like growth factor 1 (IGF-1), platelet derived growth factor (PDGF), erythropoietin (Epo), epidermal growth factor (EGF), fibroblast growth factor (FGF) (basic or acidic), granulocyte macrophage stimulating factor (GM-CSF), granulocyte colony stimulating factor (G
  • the umbilical cord biomaterial may also comprise, e.g., be coated or impregnated with, a hormone.
  • hormones include, but are not limited to, luteinizing hormone releasing hormone (LHRH), growth hormone (GH), growth hormone releasing hormone, ACTH, somatostatin, somatotropin, somatomedin, parathyroid hormone, hypothalamic releasing factors, insulin, glucagon, enkephalins, vasopressin, calcitonin, heparin, low molecular weight heparins, heparinoids, synthetic and natural opioids, insulin thyroid stimulating hormones, and endorphins.
  • ⁇ -interferons include, but are not limited to, interferon ⁇ 1-a and interferon ⁇ 1-b.
  • the umbilical cord biomaterial, or composite comprising umbilical cord biomaterial may also comprise, e.g., be coated or impregnated with, an alkylating agent.
  • alkylating agents include, but are not limited to nitrogen mustards, ethylenimines, methylmelamines, alkyl sulfonates, nitrosoureas, triazenes, mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil, hexamethylmelaine, thiotepa, busulfan, carmustine, streptozocin, dacarbazine and temozolomide.
  • the umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial may also comprise, e.g., be coated or impregnated with, an immunomodulatory agent, including but not limited to methothrexate, leflunomide, cyclophosphamide, cyclosporine A, macrolide antibiotics (e.g., FK506 (tacrolimus)), methylprednisolone (MP), corticosteroids, steroids, mycophenolate mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar, malononitriloamindes (e.g., leflunamide), T cell receptor modulators, and cytokine receptor modulators, peptide mimetics, and antibodies (e.g., human, humanized, chimeric, monoclonal, polyclonal, Fvs, ScFvs, Fab or F(ab) 2 fragments or epitope binding fragments
  • immunomodulatory agents include, but are not limited to, methothrexate, leflunomide, cyclophosphamide, cytoxan, Immuran, cyclosporine A, minocycline, azathioprine, antibiotics (e.g., FK506 (tacrolimus)), methylprednisolone (MP), corticosteroids, steroids, mycophenolate mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar, malononitriloamindes (e.g., leflunamide), T cell receptor modulators, and cytokine receptor modulators.
  • antibiotics e.g., FK506 (tacrolimus)
  • MP methylprednisolone
  • corticosteroids methylprednisolone
  • steroids e.g., methylprednisolone
  • mycophenolate mofetil e.g., rap
  • T cell receptor modulators include, but are not limited to, anti-T cell receptor antibodies (e.g., anti-CD4 antibodies (e.g., cM-T412 (Boehringer), IDEC-CE9.Is (IDEC and SKB), mAb 4162W94, Orthoclone and OKTcdr4a (Janssen-Cilag)), anti-CD3 antibodies (e.g., Nuvion (Product Design Labs), OKT3 (Johnson & Johnson), or Rituxan (IDEC)), anti-CD5 antibodies (e.g., an anti-CD5 ricin-linked immunoconjugate), anti-CD7 antibodies (e.g., CHH-380 (Novartis)), anti-CD8 antibodies, anti-CD40 ligand monoclonal antibodies (e.g., IDEC-131 (IDEC)), anti-CD52 antibodies (e.g., CAMPATH 1H (Ilex)), anti-CD2 antibodies, anti-CD1 1a antibodies (e.g.
  • a T cell receptor modulator is a CD2 antagonist. In other embodiments, a T cell receptor modulator is not a CD2 antagonist. In another specific embodiment, a T cell receptor modulator is a CD2 binding molecule, preferably MEDI-507. In other embodiments, a T cell receptor modulator is not a CD2 binding molecule.
  • the umbilical cord biomaterial, or composite comprising umbilical cord biomaterial may also comprise, e.g., be coated or impregnated with a class of immunomodulatory compounds known as IMIDS®.
  • IMID® and “IMIDS®” (Celgene Corporation) encompasses small organic molecules that markedly inhibit TNF- ⁇ , LPS induced monocyte IL-1 ⁇ and IL-12, and partially inhibit IL-6 production. Specific immunomodulatory compounds are discussed below.
  • immunomodulatory compounds include, but are not limited to, cyano and carboxy derivatives of substituted styrenes such as those disclosed in U.S. Pat. No. 5,929,117; 1-oxo-2-(2,6-dioxo-3-fluoropiperidin-3-yl)isoindolines and 1,3-dioxo-2-(2,6-dioxo-3-fluoropiperidine-3-yl)isoindolines such as those described in U.S. Pat. Nos. 5,874,448 and 5,955,476; the tetra substituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolines described in U.S.
  • immunomodulatory compounds include, but are not limited to, 1-oxo- and 1,3dioxo-2-(2,6-dioxopiperidin-3-yl)isoindolines substituted with amino in the benzo ring as described in U.S. Pat. No. 5,635,517 which is incorporated herein by reference. These compounds have the structure I:
  • immunomodulatory compounds include, but are not limited to:
  • one of X and Y is C ⁇ O and the other of X and Y is C ⁇ O or CH 2 ;
  • each of R 1 , R 2 , R 3 , and R 4 independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R 1 , R 2 , R 3 , and R 4 is —NHR 5 and the remaining of R 1 , R 2 , R 3 , and R 4 are hydrogen;
  • R 5 is hydrogen or alkyl of 1 to 8 carbon atoms
  • R 6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzyl, or halo
  • R 6 is other than hydrogen if X and Y are C ⁇ O and (i) each of R 1 , R 2 , R 3 , and R 4 is fluoro or (ii) one of R 1 , R 2 , R 3 , or R 4 is amino.
  • R 1 is hydrogen or methyl.
  • the invention encompasses the use of enantiomerically pure forms (e.g., optically pure (R) or (S) enantiomers) of these compounds.
  • one of X and Y is C ⁇ O and the other is CH 2 or C ⁇ O;
  • R 1 is H, (C 1 -C 8 )alkyl, (C 3 -C 7 )cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl, C(O)R 3 , C(S)R 3 , C(O)OR 4 , (C 1 -C 8 )alkyl-N(R 6 ) 2 , (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , C(O)NHR 3 , C(S)NHR 3 , C(O)NR 3 R 3′ , C(S)NR 3 R 3′
  • R 2 is H, F, benzyl, (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, or (C 2 -C 8 )alkynyl;
  • R 3 and R 3′ are independently (C 1 -C 8 )alkyl, (C 3 -C 7 )cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl, (C 0 -C 8 )alkyl-N(R 6 ) 2 , (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , (C 1 -C 8 )alkyl-O(CO)R 5 , or C(O)OR 5 ;
  • R 4 is (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, (C 1 -C 4 )alkyl-OR 5 , benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, or (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl;
  • R 5 is (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, or (C 2 -C 5 )heteroaryl;
  • each occurrence of R 6 is independently H, (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, (C 2 -C 5 )heteroaryl, or (C 0 -C 8 )alkyl-C(O)O—R 5 or the R 6 groups can join to form a heterocycloalkyl group;
  • n 0 or 1
  • R 1 is (C 3 -C 7 )cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl, C(O)R 3 , C(O)OR 4 , (C 1 -C 8 )alkyl-N(R 6 ) 2 , (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , C(S)NHR 3 , or (C 1 -C 8 )alkyl-O(CO)R 5 ;
  • R 2 is H or (C 1 -C 8 )alkyl
  • R 3 is (C 1 -C 8 )alkyl, (C 3 -C 7 )cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl, (C 5 -C 8 )alkyl-N(R 6 ) 2 ; (C 0 -C 8 )alkyl-NH—C(O)O—R 5 ; (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , (C 1 -C 8 )alkyl-O(CO)R 5 , or C(O)OR 5 ; and the other variables have the same definition
  • R 2 is H or (C 1 -C 4 )alkyl.
  • R 1 is (C 1 -C 8 )alkyl or benzyl.
  • R 1 is H, (C 1 -C 8 )alkyl, benzyl, CH 2 OCH 3 , CH 2 CH 2 OCH 3 , or
  • R 1 is
  • R 7 is independently H, (C 1 -C 8 )alkyl, (C 3 -C 7 )cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, halogen, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl, (C 0 -C 8 )alkyl-N(R 6 ) 2 , (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , (C 1 -C 8 )alkyl-O(CO)R 5 , or C(O)OR 5 , or adjacent occurrences of R 7 can be taken together to form
  • R 1 is C(O)R 3 .
  • R 3 is (C 0 -C 4 )alkyl-(C 2 -C 5 )heteroaryl, (C 1 -C 8 )alkyl, aryl, or (C 0 -C 4 )alkyl-OR 5 .
  • heteroaryl is pyridyl, furyl, or thienyl.
  • R 1 is C(O)OR 4 .
  • the H of C(O)NHC(O) can be replaced with (C 1 -C 4 )alkyl, aryl, or benzyl.
  • compounds in this class include, but are not limited to: [2-(2,6-dioxo-piperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-4-ylmethyl]-amide; (2-(2,6-dioxo-piperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-4-ylmethyl)-carbamic acid tert-butyl ester; 4-(aminomethyl)-2-(2,6-dioxo(3-piperidyl))-isoindoline-1,3-dione; N-(2-(2,6-dioxo-piperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-4-ylmethyl)-acetamide; N- ⁇ (2-(2,6-dioxo(3
  • one of X and Y is C ⁇ O and the other is CH 2 or C ⁇ O;
  • R is H or CH 2 OCOR′
  • each of R 1 , R 2 , R 3 , or R 4 independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R 1 , R 2 , R 3 , or R 4 is nitro or —NHR 5 and the remaining of R 1 , R 2 , R 3 , or R 4 are hydrogen;
  • R 5 is hydrogen or alkyl of 1 to 8 carbons
  • R 6 hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro;
  • R′ is R 7 —CHR 10 —N(R 8 R 9 );
  • R 7 is m-phenylene or p-phenylene or —(C n H 2n )— in which n has a value of 0 to 4;
  • each of R 8 and R 9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R 8 and R 9 taken together are tetramethylene, pentamethylene, hexamethylene, or —CH 2 CH 2 X 1 CH 2 CH 2 — in which X 1 is —O—, —S—, or —NH—;
  • R 10 is hydrogen, alkyl of to 8 carbon atoms, or phenyl
  • one of X and Y is C ⁇ O and the other of X and Y is C ⁇ O or CH 2 ;
  • each of R 1 , R 2 , R 3 , or R 4 independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R 1 , R 2 , R 3 , and R 4 is —NHR 5 and the remaining of R 1 , R 2 , R 3 , and R 4 are hydrogen;
  • R 5 is hydrogen or alkyl of 1 to 8 carbon atoms
  • R 6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro;
  • R 7 is m-phenylene or p-phenylene or —(C n H 2n )— in which n has a value of 0 to 4;
  • each of R 8 and R 9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R 8 and R 9 taken together are tetramethylene, pentamethylene, hexamethylene, or —CH 2 CH 2 X 1 CH 2 CH 2 — in which X 1 is —O—, —S—, or —NH—;
  • R 10 is hydrogen, alkyl of to 8 carbon atoms, or phenyl.
  • one of X and Y is C ⁇ O and the other of X and Y is C ⁇ O or CH 2 ;
  • each of R 1 , R 2 , R 3 , and R 4 is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R 1 , R 2 , R 3 , and R 4 is nitro or protected amino and the remaining of R 1 , R 2 , R 3 , and R 4 are hydrogen; and
  • R 6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro.
  • one of X and Y is C ⁇ O and the other of X and Y is C ⁇ O or CH 2 ;
  • each of R 1 , R 2 , R 3 , and R 4 independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R 1 , R 2 , R 3 , and R 4 is —NHR 5 and the remaining of R 1 , R 2 , R 3 , and R 4 are hydrogen;
  • R 5 is hydrogen, alkyl of 1 to 8 carbon atoms, or CO—R 7 —CH(R 10 )NR 8 R 9 in which each of R 7 , R 8 , R 9 , and R 10 is as herein defined;
  • R 6 is alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro.
  • one of X and Y is C ⁇ O and the other of X and Y is C ⁇ O or CH 2 ;
  • R 6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzyl, chloro, or fluoro;
  • R 7 is m-phenylene, p-phenylene or —(C n H 2n )— in which n has a value of 0 to 4;
  • each of R 8 and R 9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R 8 and R 9 taken together are tetramethylene, pentamethylene, hexamethylene, or —CH 2 CH 2 X 1 CH 2 CH 2 — in which X 1 is —O—, —S— or —NH—; and
  • R 10 is hydrogen, alkyl of 1 to 8 carbon atoms, or phenyl.
  • Preferred immunomodulatory compounds are 4-(amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-1,3-dione and 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione.
  • the compounds can be obtained via standard, synthetic methods (see e.g., U.S. Pat. No. 5,635,517, incorporated herein by reference).
  • the compounds are available from Celgene Corporation, Warren, N.J.
  • 4-(Amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-1,3-dione has the following chemical structure:
  • the compound 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione has the following chemical structure:
  • specific immunomodulatory compounds encompass polymorphic forms of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione such as Form A, B, C, D, E, F, G and H, disclosed in U.S. provisional application no. 60/499,723 filed on Sep. 4, 2003, and U.S. non-provisional application Ser. No. 10/934,863, filed Sep. 3, 2004, both of which are incorporated herein by reference.
  • Form A of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is an unsolvated, crystalline material that can be obtained from non-aqueous solvent systems.
  • Form A has an X-ray powder diffraction pattern comprising significant peaks at approximately 8, 14.5, 16, 17.5, 20.5, 24 and 26 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 270° C.
  • Form A is weakly or not hygroscopic and appears to be the most thermodynamically stable anhydrous polymorph of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidine-2,6-dione discovered thus far.
  • Form B of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a hemihydrated, crystalline material that can be obtained from various solvent systems, including, but not limited to, hexane, toluene, and water.
  • Form B has an X-ray powder diffraction pattern comprising significant peaks at approximately 16, 18, 22 and 27 degrees 2 ⁇ , and has endotherms from DSC curve of about 146 and 268° C., which are identified dehydration and melting by hot stage microscopy experiments. Interconversion studies show that Form B converts to Form E in aqueous solvent systems, and converts to other forms in acetone and other anhydrous systems.
  • Form C of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a hemisolvated crystalline material that can be obtained from solvents such as, but not limited to, acetone.
  • Form C has an X-ray powder diffraction pattern comprising significant peaks at approximately 15.5 and 25 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 269° C.
  • Form C is not hygroscopic below about 85% RH, but can convert to Form B at higher relative humidities.
  • Form D of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a crystalline, solvated polymorph prepared from a mixture of acetonitrile and water.
  • Form D has an X-ray powder diffraction pattern comprising significant peaks at approximately 27 and 28 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 270° C.
  • Form D is either weakly or not hygroscopic, but will typically convert to Form B when stressed at higher relative humidities.
  • Form E of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a dihydrated, crystalline material that can be obtained by slurrying 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione in water and by a slow evaporation of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione in a solvent system with a ratio of about 9:1 acetone:water.
  • Form E has an X-ray powder diffraction pattern comprising significant peaks at approximately 20, 24.5 and 29 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 269° C.
  • Form E can convert to Form C in an acetone solvent system and to Form G in a THF solvent system. In aqueous solvent systems, Form E appears to be the most stable form.
  • Desolvation experiments performed on Form E show that upon heating at about 125° C. for about five minutes, Form E can convert to Form B. Upon heating at 175° C. for about five minutes, Form B can convert to Form F.
  • Form F of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is an unsolvated, crystalline material that can be obtained from the dehydration of Form E.
  • Form F has an X-ray powder diffraction pattern comprising significant peaks at approximately 19, 19.5 and 25 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 269° C.
  • Form G of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is an unsolvated, crystalline material that can be obtained from slurrying forms B and E in a solvent such as, but not limited to, tetrahydrofuran (THF).
  • Form G has an X-ray powder diffraction pattern comprising significant peaks at approximately 21, 23 and 24.5 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 267° C.
  • Form H of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a partially hydrated (about 0.25 moles) crystalline material that can be obtained by exposing Form E to 0% relative humidity.
  • Form H has an X-ray powder diffraction pattern comprising significant peaks at approximately 15, 26 and 31 degrees 2 ⁇ , and has a differential scanning calorimetry melting temperature maximum of about 269° C.
  • immunomodulatory compounds include, but are not limited to, 1-oxo-2-(2,6-dioxo-3-fluoropiperidin-3yl)isoindolines and 1,3-dioxo-2-(2,6-dioxo-3-fluoropiperidine-3-yl)isoindolines such as those described in U.S. Pat. Nos. 5,874,448 and 5,955,476, each of which is incorporated herein by reference. Representative compounds are of formula:
  • each of R 1 , R 2 , R 3 , and R 4 is hydrogen, halo, alkyl of 1 to 4 carbon atoms; alkoxy of 1 to 4 carbon atoms, or amino.
  • immunomodulatory compounds include, but are not limited to, the tetra substituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolines described in U.S. Pat. No. 5,798,368, which is incorporated herein by reference.
  • Representative compounds are of formula:
  • each of R 1 , R 2 , R 3 , and R 4 independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms.
  • immunomodulatory compounds include, but are not limited to, 1-oxo and 1,3-dioxo-2-(2,6-dioxopiperidin-3-yl)isoindolines disclosed in U.S. Pat. No. 6,403,613, which is incorporated herein by reference.
  • Representative compounds are of formula:
  • Y is oxygen or H 2 ,
  • a first of R 1 and R 2 is halo, alkyl, alkoxy, alkylamino, dialkylamino, cyano, or carbamoyl
  • the second of R 1 and R 2 independently of the first, is hydrogen, halo, alkyl, alkoxy, alkylamino, dialkylamino, cyano, or carbamoyl
  • R 3 is hydrogen, alkyl, or benzyl.
  • R 1 and R 2 are halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl,
  • the second of R 1 and R 2 independently of the first, is hydrogen, halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, alkylamino in which alkyl is of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl, and
  • R 3 is hydrogen, alkyl of from 1 to 4 carbon atoms, or benzyl. Specific examples include, but are not limited to, 1-oxo-2-(2,6-dioxopiperidin-3-yl)-4-methylisoindoline.
  • R 1 and R 2 are halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl,
  • the second of R 1 and R 2 independently of the first, is hydrogen, halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, alkylamino in which alkyl is of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl, and
  • R 3 is hydrogen, alkyl of from 1 to 4 carbon atoms, or benzyl.
  • immunomodulatory compounds include, but are not limited to, 1-oxo and 1,3-dioxoisoindolines substituted in the 4- or 5-position of the indoline ring described in U.S. Pat. No. 6,380,239 and co-pending U.S. application Ser. No. 10/900,270, filed Jul. 28, 2004, which are incorporated herein by reference.
  • Representative compounds are of formula:
  • the carbon atom designated C* constitutes a center of chirality (when n is not zero and R 1 is not the same as R 2 ); one of X 1 and X 2 is amino, nitro, alkyl of one to six carbons, or NH—Z, and the other of X 1 or X 2 is hydrogen; each of R 1 and R 2 independent of the other, is hydroxy or NH—Z; R 3 is hydrogen, alkyl of one to six carbons, halo, or haloalkyl; Z is hydrogen, aryl, alkyl of one to six carbons, formyl, or acyl of one to six carbons; and n has a value of 0, 1, or 2; provided that if X 1 is amino, and n is 1 or 2, then R 1 and R 2 are not both hydroxy; and the salts thereof.
  • the carbon atom designated C* constitutes a center of chirality when n is not zero and R 1 is not R 2 ;
  • one of X 1 and X 2 is amino, nitro, alkyl of one to six carbons, or NH—Z, and the other of X 1 or X 2 is hydrogen; each of R 1 and R 2 independent of the other, is hydroxy or NH—Z;
  • R 3 is alkyl of one to six carbons, halo, or hydrogen;
  • Z is hydrogen, aryl or an alkyl or acyl of one to six carbons; and
  • n has a value of 0, 1, or 2.
  • the carbon atom designated C* constitutes a center of chirality when n is not zero and R 1 is not R 2 ;
  • one of X 1 and X 2 is amino, nitro, alkyl of one to six carbons, or NH—Z, and the other of X 1 or X 2 is hydrogen; each of R 1 and R 2 independent of the other, is hydroxy or NH—Z;
  • R 3 is alkyl of one to six carbons, halo, or hydrogen;
  • Z is hydrogen, aryl, or an alkyl or acyl of one to six carbons; and
  • n has a value of 0, 1, or 2; and the salts thereof.
  • X 1 and X 2 are nitro, or NH—Z, and the other of X 1 or X 2 is hydrogen;
  • each of R 1 and R 2 is hydroxy or NH—Z;
  • R 3 is alkyl of one to six carbons, halo, or hydrogen
  • Z is hydrogen, phenyl, an acyl of one to six carbons, or an alkyl of one to six carbons;
  • n has a value of 0, 1, or 2;
  • X 1 and X 2 are alkyl of one to six carbons;
  • each of R 1 and R 2 is hydroxy or NH—Z;
  • R 3 is alkyl of one to six carbons, halo, or hydrogen
  • Z is hydrogen, phenyl, an acyl of one to six carbons, or an alkyl of one to six carbons;
  • n has a value of 0, 1, or 2;
  • immunomodulatory compounds include, but are not limited to, isoindoline-1-one and isoindoline-1,3-dione substituted in the 2-position with 2,6-dioxo-3-hydroxypiperidin-5-yl described in U.S. Pat. No. 6,458,810, which is incorporated herein by reference.
  • Representative compounds are of formula:
  • X is —C(O)— or —CH 2 —;
  • R 1 is alkyl of 1 to 8 carbon atoms or —NHR 3 ;
  • R 2 is hydrogen, alkyl of 1 to 8 carbon atoms, or halogen
  • R 3 is hydrogen
  • alkyl of 1 to 8 carbon atoms unsubstituted or substituted with alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms,
  • phenyl unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms,
  • R 4 is hydrogen
  • alkyl of 1 to 8 carbon atoms unsubstituted or substituted with alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms,
  • phenyl unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms, or
  • benzyl unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms.
  • immunomodulatory compounds disclosed herein can either be commercially purchased or prepared according to the methods described in the patents or patent publications disclosed herein. Further, optically pure compounds can be asymmetrically synthesized or resolved using known resolving agents or chiral columns as well as other standard synthetic organic chemistry techniques.
  • the term “pharmaceutically acceptable salt” encompasses non-toxic acid and base addition salts of the compound to which the term refers.
  • Acceptable non-toxic acid addition salts include those derived from organic and inorganic acids or bases know in the art, which include, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulphonic acid, acetic acid, tartaric acid, lactic acid, succinic acid, citric acid, malic acid, maleic acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, embolic acid, enanthic acid, and the like.
  • bases that can be used to prepare pharmaceutically acceptable base addition salts of such acidic compounds are those that form non-toxic base addition salts, i.e., salts containing pharmacologically acceptable cations such as, but not limited to, alkali metal or alkaline earth metal salts and the calcium, magnesium, sodium or potassium salts in particular.
  • Suitable organic bases include, but are not limited to, N,N-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumaine (N-methylglucamine), lysine, and procaine.
  • solvate means a compound of the present invention or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • prodrug means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide the compound.
  • prodrugs include, but are not limited to, derivatives of immunomodulatory compounds of the invention that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs include derivatives of immunomodulatory compounds of the invention that comprise —NO, —NO 2 , —ONO, or —ONO 2 moieties.
  • Prodrugs can typically be prepared using well-known methods, such as those described in 1 Burger's Medicinal Chemistry and Drug Discovery, 172-178, 949-982 (Manfred E. Wolff ed., 5th ed. 1995), and Design of Prodrugs (H. Bundgaard ed., Elselvier, New York 1985).
  • biohydrolyzable amide As used herein and unless otherwise indicated, the terms “biohydrolyzable amide,” “biohydrolyzable ester,” “biohydrolyzable carbamate,” “biohydrolyzable carbonate,” “biohydrolyzable ureide,” “biohydrolyzable phosphate” mean an amide, ester, carbamate, carbonate, ureide, or phosphate, respectively, of a compound that either: 1) does not interfere with the biological activity of the compound but can confer upon that compound advantageous properties in vivo, such as uptake, duration of action, or onset of action; or 2) is biologically inactive but is converted in vivo to the biologically active compound.
  • biohydrolyzable esters include, but are not limited to, lower alkyl esters, lower acyloxyalkyl esters (such as acetoxylmethyl, acetoxyethyl, aminocarbonyloxymethyl, pivaloyloxymethyl, and pivaloyloxyethyl esters), lactonyl esters (such as phthalidyl and thiophthalidyl esters), lower alkoxyacyloxyalkyl esters (such as methoxycarbonyl-oxymethyl, ethoxycarbonyloxyethyl and isopropoxycarbonyloxyethyl esters), alkoxyalkyl esters, choline esters, and acylamino alkyl esters (such as acetamidomethyl esters).
  • lower alkyl esters such as acetoxylmethyl, acetoxyethyl, aminocarbonyloxymethyl, pivaloyloxymethyl, and pivaloyloxyethyl est
  • biohydrolyzable amides include, but are not limited to, lower alkyl amides, ⁇ -amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides.
  • biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, amino acids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.
  • stereoisomer encompasses all enantiomerically/stereomerically pure and enantiomerically/stereomerically enriched compounds of this invention.
  • stereomerically pure or “enantiomerically pure” means that a compound comprises one stereoisomer and is substantially free of its counter stereoisomer or enantiomer.
  • a compound is stereomerically or enantiomerically pure when the compound contains 80%, 90%, or 95% or more of one stereoisomer and 20%, 10%, or 5% or less of the counter stereoisomer.
  • a compound of the invention is considered optically active or stereomerically/enantiomerically pure (i.e., substantially the R-form or substantially the S-form) with respect to a chiral center when the compound is about 80% ee (enantiomeric excess) or greater, preferably, equal to or greater than 90% ee with respect to a particular chiral center, and more preferably 95% ee with respect to a particular chiral center.
  • Various immunomodulatory compounds of the invention contain one or more chiral centers, and can exist as racemic mixtures of enantiomers or mixtures of diastereomers. This invention encompasses the use of stereomerically pure forms of such compounds, as well as the use of mixtures of those forms.
  • mixtures comprising equal or unequal amounts of the enantiomers of a particular immunomodulatory compounds of the invention may be used in methods and compositions of the invention.
  • These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral columns or chiral resolving agents. See, e.g., Jacques, J., et al., Enantiomers, Racemates and Resolutions (Wiley-Interscience, New York, 1981); Wilen, S. H., et al., Tetrahedron 33:2725 (1977); Eliel, E. L., Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, S. H., Tables of Resolving Agents and Optical Resolutions p. 268 (E. L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, Ind., 1972).
  • the amount of the bioactive compound coating or impregnating the umbilical cord biomaterial may vary, and will preferably depend upon the particular bioactive compound to be delivered, and the effect desired.
  • the bioactive compound is an anti-inflammatory agent
  • the amount of the anti-inflammatory agent on or contained by the umbilical cord biomaterial is an amount sufficient to measurably reduce one or more symptoms or indicia of inflammation in a tissue contacted by, or proximal to, e.g., an umbilical cord biomaterial implant.
  • the umbilical cord biomaterial of the invention may comprise, e.g., be coated or impregnated with, at least 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 100, 1250, 1500, 2000, 2500, 300, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000 or at least 1000000 nanograms
  • the umbilical cord biomaterial of the invention may be coated with, or impregnated with, no more than 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 100, 1250, 1500, 2000, 2500, 300, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000 or at least 1000000 nanograms of a bioactive compound
  • the umbilical cord biomaterial may be formed into any shape or conformation that will facilitate its use in the methods of the invention.
  • the umbilical cord biomaterial can be formed into any shape or conformation that will facilitate, e.g., the occlusion of a tympanic membrane perforation, particularly in the context of a tympanoplasty or myringoplasty; repair of a joint, ligament or tendon; etc.
  • the umbilical cord biomaterial may, for example, be provided as an extended membrane, e.g., the entire membrane from a single umbilical cord.
  • the umbilical cord biomaterial can also be provided as square, rectangular, circular or oval shaped pieces, or may be cut to conform generally to the shape of, e.g., a tympanic membrane, tendon, or other bodily structure.
  • the umbilical cord biomaterial can be tubular.
  • umbilical cord biomaterial may be provided as pieces measuring approximately 1 ⁇ 1 cm, 1.5 ⁇ 1.5 cm, 2 ⁇ 2 cm, 2.5 ⁇ 2.5 cm, 1 ⁇ 1.5 cm, 1 ⁇ 2 cm, 1 ⁇ 2.5 cm, 1 ⁇ 3 cm, 1 ⁇ 3.5 cm, 1 ⁇ 4 cm, 1 ⁇ 4.5 cm, 1 ⁇ 5 cm, 1 ⁇ 5.5 cm, 1 ⁇ 6 cm, 1 ⁇ 6.5 cm, 1 ⁇ 7 cm, 1.5 ⁇ 2 cm, 1.5 ⁇ 2.5 cm, 1.5 ⁇ 3 cm, 1.5 ⁇ 3.5 cm, 1.5 ⁇ 4 cm, 1.5 ⁇ 4.5 cm, 1.5 ⁇ 5 cm, 1.5 ⁇ 5.5 cm, 1.5 ⁇ 6 cm, 1.5 ⁇ 6.5 cm, 1.5 ⁇ 7 cm, 2 ⁇ 2.5 cm, 2 ⁇ 3 cm, 2 ⁇ 3.5 cm, 2 ⁇ 4 cm, 2 ⁇ 4.5 cm, 2 ⁇ 5 cm, 2 ⁇ 5.5 cm, 2 ⁇ 6 cm, 2 ⁇ 6.5 cm, 2 ⁇ 7 cm, 2.5 ⁇ 2.5 cm, 2.5 ⁇ 3 cm, 2.5 ⁇ 3.5 cm, 2.5 ⁇ 4 cm, 2.5 ⁇ 4.5 cm, 2.5 ⁇ 5 cm, 2.5 ⁇ 5.5 cm, 2.5 ⁇ 6 cm, 2.5 ⁇ 6.5 cm, 2.5 ⁇ 7 cm, 3 ⁇
  • the biomaterial is about 2.5 cm ⁇ 2.5 cm to 3.5 cm ⁇ 3.5 cm, or, more preferably, about 3 ⁇ 3 cm 2 . Longer and/or wider pieces can be formed by laminating two or more smaller pieces, as described elsewhere herein. Further, the biomaterial may be provided as a sheet from which an end user may cut two or more pieces, or may be provided as a roll or strip.
  • the biomaterial may be provided to the end user either dry, or pre-wetted in a suitable physiologically-compatible, medically-useful liquid, such as a saline solution.
  • the solution comprises one or more bioactive compounds, as described in Section 5.1.2, above.
  • said bioactive compound is disposed onto or within the umbilical cord biomaterial such that the majority of the bioactive compound contacts the tympanic membrane at some point during the time the umbilical cord biomaterial contacts the tympanic membrane.
  • the umbilical cord biomaterial of the invention can be made in a number of ways.
  • the biomaterial is preferably produced by any means that preserves the biochemical and structural characteristics of the membrane's components—chiefly collagen, elastin, laminin, and fibronectin. That is, the biomaterial can be made so as to preserve, or substantially preserve, the native structure of the protein components of the biomaterial.
  • the biomaterial may also be altered, e.g., the proteins of the biomaterial can be crosslinked, so as to improve the strength (e.g., tensile strength) of the biomaterial.
  • the biomaterial can be completely, or substantially completely, decellularized prior to use, that is, can comprise only, or substantially only, an umbilical cord outer membrane, or can be made to retain other components of the umbilical cord (e.g., Wharton's jelly, umbilical vessel(s), umbilical cord cells, and the like).
  • an umbilical cord is separated from a placenta obtained by normal birth. The umbilical cord is then cleaned and disinfected, and optionally stored for further processing, e.g., decellularization and/or drying.
  • the umbilical cord is separated from the placenta as soon as possible after delivery of the newborn.
  • the umbilical cord may be used immediately, or may be stored for 2-5 days from the time of delivery prior to any further treatment.
  • the expectant mother is screened prior to the time of birth, using standard techniques known to one skilled in the art, for communicable diseases including but not limited to, HIV, HBV, HCV, HTLV, syphilis, CMV, and other viral pathogens known to contaminate umbilical cord tissue.
  • One exemplary method for preparing the umbilical cord biomaterial of the invention comprises the following steps:
  • the umbilical cord is separated from the placental disc, and is typically massaged to remove umbilical cord blood.
  • the umbilical cord is sectioned into pieces of about 10 cm to about 15 cm in length.
  • the umbilical cord or umbilical cord sections can then be stored for up to about 72 hours in a sterile, preferably buffered, saline solution, such as 0.9% sterile NaCl solution.
  • the umbilical cord is stored under refrigeration, at a temperature of about 1° C. to about 5° C.
  • the umbilical cord can be slit or cut longitudinally using, e.g., a scalpel and forceps, grooved director, or the like.
  • the umbilical cord membrane can also be processed further without cutting and opening the membrane.
  • An umbilical cord vessel for example, can be removed from the cord by grasping the vessels with a forceps and gently pulling and massaging until the vessel is removed, leaving the umbilical cord membrane as an intact tube.
  • the umbilical vein of a fresh (less than 48 hours after delivery) umbilical cord is canalized using the blunt probe of a vein stripper.
  • the blunt probe is replaced with a small bullet probe, and the vein is tied to the probe with thread.
  • the stripper is then removed, and the process is repeated with the umbilical arteries.
  • the umbilical cord can be further processed “as is”, wherein the cord comprises the umbilical cord membrane, vessels, and Wharton's jelly.
  • the umbilical cord biomaterial can be substantially decellularized; that is, substantially all cellular material and cellular debris (e.g., all visible cellular material and cellular debris) can removed from the biomaterial.
  • Any decellularizing process known to one skilled in the art may be used, however, generally the process used for decellularizing the umbilical cord biomaterial of the invention does not disrupt the native conformation of the proteins making up the biomaterial.
  • “Substantially decellularized,” as used herein, means removal of at least 90% of the cells, more preferably at least 95% of the cells, and most preferably at least 99% of the cells associated with the umbilical cord membrane.
  • Decellularization can leave cellular material on the umbilical cord biomaterial; for example, decellularization can leave nuclear material detectable by 4′,6-diamidino-2-phenylindole (DAPI) and still be considered decellularized.
  • DAPI 4′,6-diamidino-2-phenylindole
  • Decellularization can comprise physical scraping, for example, with a sterile cell scraper, in combination with rinsing with a sterile solution.
  • the decellularization technique employed preferably does not result in gross disruption of the anatomy of the umbilical cord membrane or alter the biomechanical properties of the umbilical cord membrane.
  • the decellularization of the umbilical cord biomaterial can comprise contacting the membrane with a detergent-containing solution, such as one or more mild anionic or nonionic detergents, e.g., Triton X-100, sodium dodecyl sulfate, or the like, in an amount and for a time sufficient to decellularize the biomaterial.
  • a detergent-containing solution such as one or more mild anionic or nonionic detergents, e.g., Triton X-100, sodium dodecyl sulfate, or the like
  • Any mild detergent i.e., a non-caustic, low-foaming detergent, with a pH of about 6 to about 8, can be used to decellularize the umbilical cord biomaterial.
  • the biomaterial is contacted with about 0.01-1% deoxycholic acid (e.g., deoxycholic acid sodium salt monohydrate) for about 30 minutes to about 480 hours, preferably about 1 hour to about 240 hours, to decellularize the umbilical cord biomaterial.
  • deoxycholic acid e.g., deoxycholic acid sodium salt monohydrate
  • the umbilical cord biomaterial is decellularized in about 1% for about 20 days without scraping, followed by heat drying.
  • the biomaterial can be decellularized by any other method known to those in the art, including freezing to form intracellular ice (including, e.g., vapor phase freezing).
  • freezing is used to decellularize the biomaterial
  • a cryoprotectant e.g., polyvinylpyrollidone at, e.g., 10% w/v, or dialyzed hydroxyethyl starch at, e.g., 10% w/v, added to standard cryopreservation solutions such as, in a non-limiting example, DMEM comprising 10% DMSO and 10% fetal bovine serum.
  • any native or exogenous protease activity is inhibited or prevented in the preparation of the biomaterial.
  • Additives to the decellularization, rinse and/or storage solutions such as metal ion chelators, for example 1,10-phenanthroline and ethylenediaminetetraacetic acid (EDTA), create an environment unfavorable to many proteolytic enzymes.
  • metal ion chelators for example 1,10-phenanthroline and ethylenediaminetetraacetic acid (EDTA)
  • EDTA ethylenediaminetetraacetic acid
  • Providing sub-optimal conditions for proteases e.g., collagenase
  • Suboptimal conditions for proteases may be achieved by formulating the decellularization solution to eliminate or limit the amount of available calcium and zinc ions.
  • the decellularization solution is prepared, in part, by selecting conditions of pH, reduced availability of calcium and zinc ions, presence of metal ion chelators and the use of proteolytic inhibitors specific for collagenase, such that the solution will optimally lyse the native umbilical cord cells while protecting the umbilical cord biomaterial from proteolytic degradation.
  • a decellularization solution can include a buffered solution of water, pH 5.5 to 8, preferably pH 7 to 8, free from calcium and zinc ions and including a metal ion chelator such as EDTA. Decellularization can take place at, e.g., between 0° C. and 25° C., preferably below about 10° C., to reduce protease activity.
  • the decellularization treatment also limits the generation of new immunological sites. Since enzymatic degradation of, e.g., collagen is believed to lead to heightened immunogenicity, the invention encompasses treatment of the umbilical cord biomaterial with enzymes, e.g., nucleases, that are effective in inhibiting cellular metabolism, protein production and cell division, that minimize proteolysis of the components of the umbilical cord biomaterial thus preserving the underlying architecture of the amniotic biomaterial.
  • enzymes e.g., nucleases
  • nucleases that can be used in accordance with the methods of the invention are those effective in digestion of native cell DNA and RNA including both exonucleases and endonucleases.
  • exonucleases that inhibit cellular activity e.g., DNase I (SIGMA Chemical Company, St. Louis, Mo.) and RNase A (SIGMA Chemical Company, St. Louis, Mo.)
  • endonucleases that inhibit cellular activity e.g., EcoRI (SIGMA Chemical Company
  • the ionic concentration of the buffered solution, the treatment temperature and the length of treatment are selected by one skilled in the art by routine experimentation to assure the desired level of nuclease activity.
  • the buffer is preferably hypotonic to promote access of the nucleases to cell interiors.
  • the umbilical cord biomaterial is not decellularized prior to drying.
  • the umbilical cord after initial processing, is briefly rinsed in saline to remove blood from the umbilical cord surface.
  • the umbilical cord is then immersed in a cold deoxycholic acid solution at a concentration of about 0.1% to about 10%, and, in a specific embodiment, about 0.1% to about 2.0%.
  • the umbilical cord is then incubated in this solution at between about 1° C. to about 8° C. for about 5 days to about 6 months.
  • the umbilical cord is immersed, for example, for about 5 to about 15 days; about 5 to about 30 days, about 5 to about 60 days, or for up to about one year.
  • the deoxycholic acid solution is replaced during incubation every 2-5 days.
  • the umbilical cord is immersed in a deoxycholic acid solution at a concentration of about 1% at a temperature of 0° C. to about 8° C. for about 5 days to about 15 days.
  • This incubation serves two purposes. First, it allows time for serological tests to be performed on the umbilical cord and/or umbilical cord blood, so that umbilical cords failing to meet serological criteria are not processed further. Second, the longer incubation improves the removal of epithelial cells and fibroblasts, which allows for a significant reduction in the amount of time spent decellularizing the umbilical cord membrane by physically scraping.
  • the umbilical cord biomaterial can then be dried as described below.
  • the umbilical cord biomaterial is generally washed to assure removal of cellular debris (e.g., cellular proteins, cellular lipids, cellular nucleic acids, extracellular debris such as extracellular soluble proteins, lipids and proteoglycans, and the like).
  • the wash solution can be de-ionized water or an aqueous hypotonic buffer.
  • the umbilical cord biomaterial is gently agitated, e.g., for 15-120 minutes in the detergent, e.g., on a rocking platform, to assist in the decellularization.
  • the umbilical cord biomaterial after detergent decellularization, can again be physically decellularized as described above; the physical and detergent decellularization steps may be repeated as necessary, as long as the integrity of the umbilical cord biomaterial is maintained, until no visible'cellular material and cellular debris remain.
  • the umbilical cord biomaterial is dried immediately (i.e., within 30 minutes) after decellularization and/or washing.
  • the umbilical cord biomaterial may be refrigerated, e.g., stored at a temperature of about 1° C. to about 20° C., preferably from about 2° C. to about 8° C., for up to 28 days prior to drying.
  • the umbilical cord biomaterial e.g., decellularized umbilical cord biomaterial
  • the sterile solution covering the umbilical cord biomaterial is preferably changed periodically, e.g., every 1-3 days.
  • the biomaterial when the umbilical cord biomaterial is not refrigerated after washing, the biomaterial can be washed, e.g., washed at least 3 times, prior to proceeding to the next step of the preparation. In other embodiments, when the umbilical cord biomaterial has been refrigerated and the sterile solution has been changed once, the umbilical cord biomaterial can be washed at least twice prior to the next step of the preparation. In yet other embodiments, when the umbilical cord biomaterial has been refrigerated and the sterile solution has been changed twice or more, the umbilical cord biomaterial can be washed at least once prior to proceeding to the next step.
  • the final step in this embodiment comprises drying the decellularized umbilical cord membrane to produce the umbilical cord biomaterial of the invention.
  • Any method of drying the umbilical cord membrane can be used.
  • the membrane can be dried using heat, one or more hygroscopic compounds, freeze-drying, vacuum, microwaving, simple evaporation, and the like, or combinations of these methods.
  • the biomaterial is dried under vacuum.
  • the umbilical cord biomaterial can be dried in any useful conformation.
  • the umbilical cord biomaterial is dried so as to produce a flat, dry sheet.
  • the biomaterial can also be dried as a tube, strip, spiral, string or rope, or the like.
  • the biomaterial can be placed onto, or into, a form and dried, so that the dried biomaterial assumes the shape of the form or a part thereof.
  • the umbilical cord membrane can be supported by rubber hose or tubing inserted from one end, and freeze-dried to form a dried tube of the umbilical cord biomaterial.
  • the umbilical cord biomaterial can be, e.g., part of a complete umbilical cord that has been washed and rinsed; an umbilical cord biomaterial comprising Wharton's jelly but lacking vessels, an umbilical cord biomaterial that has had the interior components of the umbilical cord removed, and has been decellularized, etc.
  • the biomaterial can be dried.
  • an exemplary method for drying the umbilical cord biomaterial comprises the following steps:
  • the umbilical cord biomaterial is removed from the sterile solution, and the excess fluid is gently squeezed out.
  • the umbilical cord biomaterial is then gently stretched until it is flat with the epithelial side facing in a downward position, e.g., on a tray.
  • the umbilical cord biomaterial is then placed on a drying frame, preferably a plastic mesh drying frame (e.g., QUICK COUNT® Plastic Canvas, Uniek, Inc., Waunakee, Wis.).
  • the drying frame may be any autoclavable material, including but not limited to a stainless steel mesh.
  • a sterile gauze can be placed on the drying platform of a heat dryer (or gel-dryer) (e.g., Model 583, Bio-Rad Laboratories, Hercules, Calif.), so that an area slightly larger than the umbilical cord biomaterial resting on the plastic mesh drying frame is covered.
  • a heat dryer or gel-dryer
  • the total thickness of the gauze layer does not exceed the thickness of one folded 4 ⁇ 4 gauze.
  • Any heat drying apparatus may be used that is suitable for drying sheet-like material.
  • the drying frame is placed on top of the gauze on the drying platform so that the edges of the plastic frame extend above beyond the gauze edges, preferably between 0.1-1.0 cm, more preferably 0.5-1.0 cm.
  • another plastic framing mesh is placed on top of the umbilical cord biomaterial.
  • a sheet of thin plastic e.g., SW 182, clear PVC, AEP Industries Inc., South Hackensack, N.J.
  • a biocompatible silicone is placed on top of the biomaterial covered mesh so that the sheet extends well beyond all of the edges. In this embodiment, the second mesh frame is not needed.
  • the umbilical cord biomaterial is placed one or more sterile sheets of TYVEK® material (e.g., a sheet of TYVEK® for medical packaging, DuPont TYVEK®, Wilmington, Del.), optionally, with one sheet of TYVEK® on top of the biomaterial (prior to placing the plastic film).
  • TYVEK® material e.g., a sheet of TYVEK® for medical packaging, DuPont TYVEK®, Wilmington, Del.
  • This alternate process will produce a smoother version of the biomaterial (i.e., without the pattern of differential fiber compression regions along and perpendicular to the axis of the material), which may be advantageous for certain applications, such as for example for use as a matrix for expansion of cells.
  • the invention encompasses heat drying the umbilical cord biomaterial of the invention under vacuum. While the drying under vacuum may be accomplished at any temperature from about 0° C. to about 60° C., the umbilical cord biomaterial is preferably dried at between about 35° C. and about 50° C., and most preferably at about 50° C. It should be noted that some degradation of the collagen is to be expected at temperatures above 50° C.
  • the drying temperature is preferably set and verified using a calibrated digital thermometer using an extended probe. Any amount of vacuum that can be conveniently generated can be used, but preferably, the vacuum pressure is set to about ⁇ 22 inches of Hg.
  • the drying step is continued until the umbilical cord biomaterial is substantially dry, that is, contains less than 20% water by weight, and preferably, about 3-12% water by weight as determined for example by a moisture analyzer.
  • the umbilical cord biomaterial may be heat-vacuum dried, e.g., for approximately 60 minutes to achieve a dehydrated umbilical cord biomaterial.
  • the umbilical cord biomaterial is dried for about 30 minutes to 2 hours, preferably about 60 minutes.
  • low (e.g., ⁇ 50° C.) heat coupled with vacuum pressure allows the umbilical cord biomaterial to achieve the dehydrated state without denaturing collagen in the biomaterial.
  • the umbilical cord biomaterial can be cooled down, e.g., for approximately two minutes, with the vacuum pump running.
  • the umbilical cord biomaterial is dried, the biomaterial is gently lifted off the drying frame. Preferably, handling of the umbilical cord biomaterial at this stage is done with sterile gloves.
  • the umbilical cord biomaterial can be placed in a sterile container, e.g., peel pouch.
  • the umbilical cord biomaterial produced in accordance with the methods of the invention may be stored at room temperature for an extended period of time as described supra.
  • the umbilical cord biomaterial is prepared as above, but is not decellularized. That is, the umbilical cord membrane is obtained and dried, but the cells associated with the umbilical cord membrane are not removed.
  • the final, dried product thus comprises, e.g., the umbilical cord membrane and/or umbilical cord vessel(s), as well as cellular components.
  • the umbilical cord biomaterial can be dehydrated by other methods in place of, or in addition to, the vacuum-drying method outlined above.
  • the biomaterial can be freeze dried.
  • umbilical cord biomaterial can be frozen at a temperature between about ⁇ 170° C. and about 0° C. for time sufficient for the biomaterial to completely freeze.
  • the frozen biomaterial is freeze-dried process during which the ice crystals will be removed or avoided by sublimation under vacuum.
  • the biomaterial can be dehydrated using a solvent.
  • umbilical cord biomaterial can be dehydrated using, e.g., ethanol and acetone.
  • the biomaterial can be, e.g., soaked for a time in a series of ethanol-acetone mixtures (e.g., 20%, 40%, 60%, 80% and 100% ethanol, or a similar progression of equivalent solvents that act to extract water) such that the water inside the biomaterial is gradually replaced by the organic solvent.
  • the biomaterial can be placed in a well-ventilated place at room temperature (about 23° C.) for a time sufficient for the solvent to evaporate.
  • the biomaterial can alternately be vacuum-dried after the solvent soak.
  • the biomaterial can also be dehydrated by freeze drying.
  • the processed membrane can be first frozen and then transferred to a water miscible organic solvent. Ice crystals inside the membrane tissue may then be dissolved and replaced by the organic solvent using a series of progressive solvent soaks as described above. After the final soak, the biomaterial can be placed in a well-ventilated place at room temperature (e.g., about 20° C. to about 25° C.) for a time sufficient for the ethanol to evaporate. The biomaterial can alternately be vacuum-dried after the 100% ethanol soak.
  • Non-heat drying processes may be preferred if the porous structure of the biomaterial and/or bioactivity of the biological substances within the umbilical cord membrane need to be preserved, for example, if the biomaterial is to be used as a substrate or matrix for the transport of stem cells to a graft site, or if, e.g., the biomaterial is to be preloaded with a heat-sensitive drug as a drug release device, or if, e.g., the biomaterial is preloaded with a heat sensitive drug as a drug release device.
  • the membrane generally primarily comprises collagen (types I, III, IV, V, VI and VII), glycosaminoglycans (particularly hyaluronic acid); and growth factors, particularly fibroblast growth factor (FGF), basic fibroblast growth factor (b-FGF), epidermal growth factor (EGF), insulin-like growth factor I (IGF-I), platelet-derived growth factor (PDGF) and transforming growth factor beta (TGF- ⁇ ).
  • FGF fibroblast growth factor
  • b-FGF basic fibroblast growth factor
  • EGF epidermal growth factor
  • IGF-I insulin-like growth factor I
  • PDGF platelet-derived growth factor
  • TGF- ⁇ transforming growth factor beta
  • Dehydrated umbilical cord biomaterial may be stored, e.g., as dehydrated sheets, at room temperature (e.g., 25° C.) prior to use.
  • the umbilical cord biomaterial can be stored at a temperature of at least 10° C., at least 15° C., at least 20° C., at least 25° C., or at least 29° C.
  • umbilical cord biomaterial, in dehydrated form is not refrigerated.
  • the umbilical cord biomaterial may be refrigerated at a temperature of about 2° C. to about 8° C.
  • the umbilical cord biomaterial produced according to the methods of the invention can be stored at any of the specified temperatures for 12 months or more with no alteration in biochemical or structural integrity (e.g., no degradation), without any alteration of the biochemical or biophysical properties of the umbilical cord biomaterial.
  • the biomaterial can be stored for several years with no alteration in biochemical or structural integrity (e.g., no degradation), without any alteration of the biochemical or biophysical properties of the biomaterial.
  • the biomaterial can be stored in any container suitable for long-term storage.
  • the umbilical cord biomaterial of the invention is stored in a sterile double peel-pouch package.
  • the umbilical cord biomaterial in embodiments in which the material has been dried, may be hydrated prior to use, using, e.g., a sterile physiological buffer.
  • the sterile saline solution is a 0.9% NaCl solution.
  • the sterile saline solution is buffered.
  • the hydration of the umbilical cord biomaterial requires at least 2 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, or at least 20 minutes.
  • the hydration of the umbilical cord biomaterial is complete within 5 minutes.
  • the hydration of the umbilical cord biomaterial of the invention is complete within 10 minutes.
  • the hydration of the umbilical cord biomaterial takes no more than 10 minutes. Once hydrated, the umbilical cord biomaterial can be maintained in solution, e.g., in sterile 0.9% NaCl solution, for up to six months, with a change of solution, e.g., every three days.
  • Sterilization of the umbilical cord biomaterial may be accomplished by any medically-appropriate means, preferably means that do not significantly alter the tertiary and quaternary structure of the biomaterial proteins.
  • Sterilization can be accomplished, for example, using gas, e.g., ethylene dioxide.
  • Sterilization can be accomplished using radiation, for example, gamma radiation, and is preferably done by electron beam irradiation using methods known to one skilled in the art, e.g., Gorham, D. Byrom (ed.), 1991, Biomaterials, Stockton Press, New York, 55-122. Any dose of radiation sufficient to kill at least 99.9% of bacteria or other potentially contaminating organisms is within the scope of the invention. In a preferred embodiment, a dose of at least 18-25 kGy is used to achieve the terminal sterilization of the biomaterial.
  • the umbilical cord biomaterial may be laminated to provide greater load-bearing capacity and durability during the healing process.
  • Laminates of the umbilical cord biomaterial can comprise biomaterial from a single umbilical cord, wherein the composition is folded once, or a plurality of times, longitudinally or laterally, or both.
  • Laminates of the biomaterial can also comprise two or more sheets of biomaterial.
  • the umbilical cord biomaterial being anisotropic, has two orientations, longitudinal (that is, along the length of the umbilical cord membrane) and lateral (that is, around the width of the umbilical cord).
  • a laminate comprises two sheets of the umbilical cord biomaterial
  • the sheets can be laminated so that each sheet is oriented the same way (e.g., each sheet oriented longitudinally), or such that at least one sheet is oriented laterally and one longitudinally.
  • each of the layers of umbilical cord biomaterial can be laminated in any orientation with respect to any other layer of the biomaterial in the laminate.
  • the umbilical cord has a sidedness; that is, the umbilical cord biomaterial has an epithelial side (that is, the side towards the interior of the umbilical cord) and a mesothelial side (that is, the side towards the exterior of the umbilical cord).
  • Laminates can comprise two or more layers of the umbilical cord biomaterial in any sidedness configuration.
  • laminates can comprise layers of umbilical cord biomaterial in which only the endothelial sides of the layers are in contact; only the mesothelial sides of the layers are intact; or a combination of both.
  • a laminate comprises four layers, wherein two sets of two layers, contacted endothelial to mesothelial sides, are contacted by the exposed mesothelial side such that the two faces of the laminate show the endothelial sides.
  • Umbilical cord biomaterial can be laminated, e.g., by folding a single sheet of biomaterial, or by stacking 2 or more layers of the biomaterial one atop the other, and sealing or drying.
  • the biomaterial may be laminated either dry or after rehydration.
  • two or more layers of, e.g., umbilical cord biomaterial, or composition comprising an umbilical cord biomaterial can be laminated prior to initial drying after cell removal, e.g., after a cell scraping step (see Examples, below). If laminated prior to the initial drying, 2 or more biomaterial layers can be stacked one atop the other and subsequently dried, using, for example, a freeze-drying process, or drying under moderate heat with or without vacuum.
  • the heat applied preferably is not so intense as to cause breakdown or decomposition of the protein components, especially the collagen, of the umbilical cord biomaterial.
  • the heat applied is less than about 70° C., preferably less than about 60° C., and, more preferably, is approximately 50° C.
  • Lamination time varies with, e.g., the number of layers being laminated, but typically takes 1-2 hours at 50° C.
  • a method of preparing a laminate using a composition comprising umbilical cord membrane comprises layering a plurality of said membranes in contact with each other to form a laminate.
  • each of said membranes comprises less than 20% water by weight prior to said layering.
  • said laminate is dried to less than 20% water by weight after said layering.
  • the biomaterial may also be laminated using an adhesive applied between 2 or more layers of biomaterial or umbilical cord biomaterial or composition comprising an umbilical cord biomaterial.
  • an adhesive is preferably appropriate for medical applications, and can comprise a natural biological adhesive, for example fibrin glue, a synthetic adhesive, or combinations thereof.
  • the adhesive may further be chemically converted from precursors during the lamination process.
  • Laminates of the umbilical cord biomaterial can comprise, for example, umbilical cord biomaterial that has been decellularized, biomaterial that retains the cellular material (that is, where the cells have been killed, but not removed), or biomaterial comprising living umbilical cord cells or cells of another type (e.g., where cells have been cultured on a sheet of umbilical cord biomaterial).
  • a laminate can comprise a second type of material, e.g., one or more layers of umbilical cord biomaterial can be layered with one or more layers of a second biologically-compatible material, e.g., a sheetlike material such as, e.g., amniotic membrane.
  • a second biologically-compatible material e.g., a sheetlike material such as, e.g., amniotic membrane.
  • the umbilical cord biomaterial can be laminated such that the biomaterial lies with its longitudinal direction along, or alternatively across, the grain of the second material.
  • the umbilical cord membrane biomaterial is laminated with at least one other layer of a second material that has a relatively high load-bearing capacity.
  • the umbilical cord biomaterial can also be laminated with a non-biological material, e.g., plastic, e.g., TYVEK® or the like.
  • the umbilical cord biomaterial laminate comprises two layers of the biomaterial and one layer of plastic, e.g., TYVEK®, such that the plastic is sandwiched between the two layers of umbilical cord biomaterial, and the endothelial sides of the biomaterial contact the plastic.
  • one layer of umbilical cord membrane is placed on a plastic sheet with the epithelial side down, and a second piece of umbilical cord membrane is placed on the first epithelial side up. The resulting product is then heat-dried to produce an umbilical cord biomaterial laminate.
  • the second material has a load-bearing capacity or tensile strength, for, e.g., a 2-centimeter wide section, of at least 25 milliPascals (mPa), 50 mPa, 75 mPa, 100 mPa, 125 mPa, 150 mPa, 175 mPa, 200 mPa, 225 mPa, 250 mPa, 275 mPa, 300 mPa, 325 mPa, 350 mPa, 375 mPa, 400 mPa, 425 mPa, 450 mPa, 475 mPa, 500 mPa, 750 mPa, 1000 mPa, 1250 mPa, 1500 mPa, 1750 mPa or 2000 mPa.
  • mPa milliPascals
  • Such a second material can be sheetlike, or can be formed into a shape suitable for a particular application, and the umbilical cord biomaterial molded to the shape of the second material.
  • the second material can be, e.g., a material suitable for, and shaped for, tendon or ligament repair.
  • the umbilical cord biomaterial can be wrapped around such a second material so that the exterior of the laminate is biologically compatible, and the interior is load-bearing.
  • the load-bearing capacity of a particular umbilical cord biomaterial laminate, or piece of umbilical cord biomaterial can be tested by standard methods known in the art, such as ASTM D1708 (Standard Test Method for Tensile Properties of Plastics).
  • the laminate comprises at least two sheets of umbilical cord biomaterial approximately the same size and shape laid one atop the other so that the shape is substantially maintained. Such a laminate can be trimmed to finalize a particular shape.
  • the laminate comprises two or more sheets of umbilical cord biomaterial, wherein a portion of each of the sheets overlaps another. In this embodiment, several overlapping sheets of umbilical cord biomaterial can be laminated to form a larger sheet of the biomaterial than would be possible from a single umbilical cord.
  • such a laminate of overlapping sheets of the biomaterial can itself be laminated with another layer of a material, e.g., another overlapping biomaterial laminate; individual sheets of umbilical cord biomaterial; another type of biomaterial, e.g., an amniotic membrane-derived biomaterial; an artificial sheet or film; etc.
  • a material e.g., another overlapping biomaterial laminate; individual sheets of umbilical cord biomaterial; another type of biomaterial, e.g., an amniotic membrane-derived biomaterial; an artificial sheet or film; etc.
  • the umbilical cord biomaterial as described herein can also comprise stem or progenitor cells.
  • the umbilical cord biomaterial can comprise, e.g., mesenchymal or mesenchymal-like stem cells, for example, those described in U.S. Pat. Nos. 5,486,359, 6,261,549 and 6,387,367, or placental stem cells such as those described in U.S. Application Publication Nos. 2002/0123141, 2003/0032179 and 2003/0180269.
  • the umbilical cord biomaterial may comprise stem or progenitor cells, preferably mammalian stem or progenitor cells, from any tissue source.
  • the umbilical cord biomaterial can comprise embryonic stem cells or embryonic germ cells.
  • the umbilical cord biomaterial and stem or progenitor cells can be combined, e.g., in advance of a procedure in which the biomaterial is contacted with an individual having a disease, disorder or condition that would be amenable to treatment using an umbilical cord biomaterial.
  • stem cells can be contacted with, e.g., disposed onto, the biomaterial sufficiently in advance of such a procedure for a plurality, a majority, or substantially all of the stem cells to adhere to the biomaterial.
  • the stem cells can be contacted with the biomaterial immediately before the biomaterial is contacted with the individual.
  • the stem cells can also be contacted with the biomaterial in situ, after the biomaterial is contacted with the individual.
  • the number of stem or progenitor cells disposed onto the surface of the umbilical cord biomaterial may vary, but may be at least about 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , 1 ⁇ 10 10 , 5 ⁇ 10 10 , 1 ⁇ 10 11 , 5 ⁇ 10 11 , or 1 ⁇ 10 12 ; or may be no more than 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , 1 ⁇ 10 10 , 5 ⁇ 10 10 , 1 ⁇ 10 11 , 5 ⁇ 10 11 , or 1 ⁇ 10 12 stem or progenitor cells.
  • the stem cells can be contacted with one or more differentiation-modulating agents, for example, the differentiation-modulating agents described in U.S. Application Publication Nos. 2003/0235909 and/or 2004/0028660, the disclosures of which are incorporated by reverence in their entireties herein, or International Application Publication No. WO 03/087333.
  • Methods of differentiating stem cells to, for example, epidermal, mesodermal, and other cell types are known in the art, and are described, e.g., in U.S. Application Publication No. 2004/0028660.
  • the umbilical cord biomaterial can be used in a variety of medical applications.
  • the uses outlined in this section are non-limiting examples of such applications.
  • the umbilical cord biomaterial of the invention can be used, for example, to repair or replace ligaments, tendons and/or cartilage.
  • the biomaterial can be contacted with a ligament, tendon or cartilage in any medically-acceptable manner that tends to facilitate healing of a defect in the ligament, tendon or cartilage.
  • the umbilical cord biomaterial can be used, for example, to repair a defect in a tendon, ligament or cartilage where the defect is a tear, that is, a defect that is less than a complete failure.
  • the repair comprises contacting the tendon, ligament or cartilage with the umbilical cord biomaterial such that a part, or preferably all, of the defect is covered by the biomaterial.
  • the umbilical cord biomaterial can be, for example, wrapped around a tendon or ligament at the site of the defect, or a patch of umbilical cord biomaterial of sufficient size to cover the defect can be placed on the defect.
  • the biomaterial can be held in place using, e.g., a biologically-acceptable glue, e.g., a tissue glue, or can be sutured in place.
  • the umbilical cord biomaterial is held in place such that the biomaterial assumes at least part of the biomechanical load normally associated with the functional tendon, ligament or cartilage.
  • the umbilical cord biomaterial can be used, for example, to repair a defect in a tendon, ligament or cartilage where the defect is a complete rupture or failure of a tendon, ligament or cartilage.
  • the repair comprises contacting the tendon, ligament or cartilage with the umbilical cord biomaterial so as to partially, or preferably completely, cover the defect.
  • the contacting is such that the biomaterial assumes some, or all of the biomechanical load normally assumed by the tendon, ligament or cartilage.
  • a rupture in a tendon can be repaired by contacting the tendon at the site of the rupture with the biomaterial and affixing the biomaterial in place.
  • the two parts of the tendon at the site of the rupture are preferably brought into contact with each other, and the biomaterial is preferably wrapped around the site of the break.
  • the biomaterial can be sutured to the two parts of the ruptured tendon such that it acts as a splint, holding the to ends of the rupture together.
  • the biomaterial can be wrapped around such a rupture once, or preferably a plurality of times.
  • the umbilical cord biomaterial can be used, for example, to replace a tendon or ligament, or to support a joint where one or more of the native ligaments is weakened.
  • the biomaterial attaches two bones, or a muscle and bone, in place of a damaged, diseased or ruptured tendon or ligament, e.g., an anterior cruciate ligament.
  • tunnels can be drilled onto the femoral and tibial heads adjoining the knee.
  • the biomaterial e.g., folded lengthwise a plurality of times into a rope-like conformation, can be drawn through the tunnels, and the ends fastened to the respective bones.
  • Such fastening can be accomplished by any means known in the art, e.g., using screws, staples, or the like.
  • the ends of the umbilical cord biomaterial are folded over to provide a portion of the biomaterial of greater thickness for fastening.
  • Such replacement can comprise removing the native ligament during replacement, or can comprise adding the biomaterial and allowing the native ligament to remain.
  • the umbilical cord biomaterial is laminated with a load-bearing material, such as plastic sheeting, e.g., TYVEK® prior to folding into the rope-like conformation.
  • the umbilical cord biomaterial can be used to repair a tendon in the hand.
  • the umbilical cord biomaterial is inserted between one or more extensor tendons in the hand and bones to provide a gliding surface or a shield between tendon and bone, e.g., to prevent adhesion formation.
  • the umbilical cord biomaterial is stretched prior to repair or replacement of a ligament or tendon.
  • a weight, bearable by the particular piece of biomaterial can be suspended from the biomaterial for a time sufficient to allow up to, e.g., about 10%, 15%, or about 20% elongation. Such stretching tends to prevent loosening of the biomaterial after application.
  • the umbilical cord biomaterial in another embodiment, can also be used to repair or reinforce a rotator cuff tendon having a defect, e.g., a tear.
  • a piece of the umbilical cord biomaterial is used to completely cover the rotator cuff tendon defect.
  • the present invention provides methods and compositions for repair of a tympanic membrane using an umbilical cord biomaterial.
  • the present invention provides a method of repairing a perforated tympanic membrane, comprising contacting said tympanic membrane with a umbilical cord biomaterial. Said contacting can comprise shaping a flat piece of the biomaterial into the shape of an entire, or a portion of, a tympanic membrane, and contacting the biomaterial with the tympanic membrane.
  • said perforation has not healed spontaneously within two months of the appearance of the perforation.
  • the biomaterial can be contacted with the tympanic membrane while hydrated, or, preferably, while substantially dry (e.g., comprising less than 20% water by weight).
  • the biomaterial can be a single layer, or can be a laminate of two or more layers.
  • the umbilical cord biomaterial (whether a single layer or a laminate) contacted with the tympanic membrane is at least about 70 microns in thickness.
  • a tympanic membrane having a perforation is contacted with an umbilical cord biomaterial such that the biomaterial partially or totally occludes the perforation.
  • the perforation to be occluded may be a central perforation, that is, a perforation of any size that does not involve the margin of the tympanic membrane (i.e., the periphery seated in the auditory canal), or a marginal perforation (i.e., a perforation touching upon, or largely involving, the margin of the tympanic membrane).
  • the tympanic membrane is perforated, and no other ear structure is perforated or damaged.
  • occlusion of the perforation is an adjunct to at least one other surgical procedure involving the outer, middle, or inner ear.
  • the repair of the tympanic membrane is a tympanoplasty.
  • the repair of the tympanic membrane is a myringoplasty.
  • the benefits of closing a tympanic membrane perforation include prevention of water entering the ear while showering, bathing or swimming (which could cause ear infection), improved hearing, and diminished tinnitus. Closure also helps to prevent the development of cholesteatoma (skin cyst in the middle ear), which can cause chronic infection and destruction of ear structures.
  • cholesteatoma skin cyst in the middle ear
  • Tympanoplasty and myringoplasty are generally outpatient procedures.
  • the otolaryngologist may approach repair of a tympanic membrane perforation either through the auditory canal (trans-canal approach), or via a post-auricular incision followed by folding the ear forward to expose the tympanic membrane (post-auricular approach).
  • a hearing test is generally performed, and the patient is evaluated for Eustachian tube function, as partial or complete loss of Eustachian tube function can exacerbate a tympanic membrane puncture and interfere with the adherence of a graft to the tympanic membrane.
  • Repair of a perforated tympanic membrane generally comprises placing an occluding material on the membrane.
  • the patient is evaluated for complications, such as extension of squamous epithelium through the perforation and into the middle ear space.
  • tympanoplasty or myringoplasty is preferably accompanied, where possible, by remediation of the complication.
  • the present invention encompasses repair of a tympanic membrane with an umbilical cord biomaterial either as a first or subsequent therapy. That is, the biomaterial may be used to repair a tympanic membrane deformity, such as a perforation, before other remedial measures are tried. Alternatively, repair of a tympanic membrane with biomaterial may be performed after one or more other remedial measures have been tried and failed.
  • repair of a tympanic membrane with biomaterial may additionally comprise applying an anti-infective agent to the graft and/or surrounding ear canal.
  • the invention provides a method of repairing a tympanic membrane comprising contacting the tympanic membrane with an umbilical cord biomaterial and an anti-infective agent, e.g., one of the anti-infective agents listed in Section 5.1.2, above.
  • the anti-infective agent can be contacted either prior to, concurrently with, or subsequent to contacting the tympanic membrane with the umbilical cord biomaterial.
  • the anti-infective agent can be present separate from, or as an integral part of, the biomaterial.
  • the anti-infective agent can be present on the surface of the biomaterial, or can be impregnated in the biomaterial.
  • the anti-infective agent is an antibiotic, a bacteriostatic agent, antiviral compound, a virustatic agent, antifungal compound, a fungistatic agent, or an antimicrobial compound.
  • the anti-infective agent is ionic silver.
  • the ionic silver is contained within a hydrogel. Ionic silver hydrogel is a preferred anti-infective agent because it is broad spectrum, with no known bacterial resistance; its application and removal are pain-free, and the hydrogel supports autolytic debridement.
  • the umbilical cord biomaterial is impregnated with silver ions prior to application to the tympanic membrane. In another embodiment, the umbilical cord biomaterial is impregnated with silver ions after application of the biomaterial to the tympanic membrane, for example, by application of ear drops.
  • the invention further provides that the use of an umbilical cord biomaterial to repair a tympanic membrane deformity may be the sole treatment of the tympanic membrane, or may be in addition to another therapies or treatment used simultaneously in the course of treating or repairing a tympanic membrane.
  • the invention provides for the repair of a tympanic membrane comprising contacting the tympanic membrane with an umbilical cord biomaterial, and treating the tympanic membrane using an additional therapy not comprising contacting the tympanic membrane with an umbilical cord biomaterial, where the contacting and the additional therapy individually or together cause a measurable improvement in, maintenance of, or lessening of the worsening of, at least one aspect of a tympanic membrane deformity, as compared to a tympanic membrane not contacted with an umbilical cord biomaterial.
  • the invention further provides for the use of umbilical cord biomaterial to repair an ear condition in conjunction with repair of a tympanic membrane.
  • the umbilical cord biomaterial can be used to reconstruct or repair the outer or middle ear structures, including the auditory canal and middle ear chamber.
  • the umbilical cord biomaterial may be used to repair or line the mastoid cavity, particularly where mastoid reconstruction is indicated in addition to tympanoplasty.
  • the umbilical cord biomaterial may be used to line the mastoid cavity where the mastoid cavity comprises exposed bone, that is, bone with no covering epithelial cell layer.
  • the umbilical cord biomaterial may be used as a oval window graft in stapes surgery, either alone or in conjunction with tympanoplasty or myringoplasty.
  • the invention further provides for the use of umbilical cord biomaterial to repair a soft tissue injury or defect in an individual.
  • the soft tissue defect is an abdominal wall defect.
  • Such an abdominal wall defect e.g., hernia
  • the soft tissue defect is a hernia or abdominal wall defect in which the abdominal wall allows exit of at least part of an organ from the abdominal cavity.
  • the defect e.g., discontinuity or hernia
  • the umbilical cord biomaterial is sutured, stapled, or otherwise fastened to the defect such that repair is effected.
  • said soft tissue defect is a defect in the pelvic floor, an enteroceles, a rectoceles, or a cystoceles.
  • the abdominal wall defect is an opening of the abdominal wall to the exterior of the body, it is generally preferred that the defect be contacted with the mesothelial surface of the umbilical cord biomaterial.
  • the defect is incontinence
  • the umbilical cord biomaterial is used as an adjunct to a suburethral sling procedure to assist in the repair of gracilis muscle flaps sutured beneath the urethra.
  • the soft tissue defect is a leg ulcer such as, e.g., a venous leg ulcer, arterial leg ulcer, diabetic ulcer or decubitus ulcer.
  • Repair of a leg ulcer can comprise contacting a portion, or the entirety, of the leg ulcer with one or more pieces of umbilical cord biomaterial, either a single sheet or a laminate thereof, either dried or hydrated, such that the umbilical cord biomaterial becomes affixed to the leg ulcer.
  • the biomaterial becomes affixed to the leg ulcer without fastening; however, the biomaterial can be sutured, stapled or glued to the skin surrounding the ulcer, or can be held in place by, e.g., a bandage or compression boot, or by any other method known to those of skill in the art.
  • the soft tissue defect is a surgical adhesion.
  • the surgical adhesion is an adhesion resulting from, e.g., gynecological surgery. It is estimated that approximately 97% of surgical patients develop adhesions after surgery and of these, between 5% and 8% develop complications.
  • the umbilical cord biomaterial would be an effective barrier to adhesions in that the amnion epithelial cells on one side of the biomaterials would occupy cell binding sites, making it difficult for host cells, such as fibroblasts, to attach and penetrate.
  • the umbilical cord biomaterial can be used to prevent surgical adhesion by placing the biomaterial between two tissues that would ordinarily be expected to form a post-surgical adhesion.
  • the soft tissue defect is a nasal septal perforation.
  • the septal perforation may arise from any cause, e.g., inherited defect, trauma, drug use, etc.
  • a piece of umbilical cord biomaterial, suitably shaped, can be placed along the septum in order to partially or completely occlude the perforation, and can be held in place by one or several sutures or tissue glue.
  • the invention further provides for the use of umbilical cord biomaterial in the formation of an ocular plug.
  • An ocular plug at least partially, or, preferably, completely, occludes a hole in, e.g., the sclera, that is, e.g., caused by an injection, formed as a part of a surgical procedure to, e.g., allow insertion of a surgical tool into the lumen of the eye; caused by trauma; etc.
  • Ocular plugs may be configured in any shape to accomplish the particular purpose at hand, e.g., occluding injection or ocular surgery-related holes in the sclera, prevention of leakage, drug delivery, anchoring of the plug, etc.
  • the invention provides an ocular plug that comprises a shaft attached to and extending from a cap.
  • the cap is circular when viewed from the upper face.
  • the cap may be oval, square, rectangular, polygonal, irregular, or may appear as a plurality of flanges extending substantially perpendicularly from the shaft.
  • the upper face of the cap distal to the shaft may be hemispherical, curved to a degree other than completely hemispherical, or may be substantially flat.
  • the surface of the upper face of the cap is shaped to approximate the curvature of the eye to promote comfort and reduce the possibility of inflammation or irritation associated with the eyelid moving over the face of the cap.
  • the lower face of the cap proximal to the shaft may be substantially flat, but is preferably shaped to approximate the curvature of the eye.
  • the cap tapers towards the edges so that a smooth transition is made from sclera to cap when the eyelid passes over the cap.
  • the cap need not taper from center towards the edges, and may have a discernibly blunt edge.
  • the cap is preferably of a sufficient diameter to promote seating and maintenance of position of the plug within the hole in the sclera, and to reduce the possibility of the shaft from passing completely through the sclera during or after insertion of the plug into the sclera.
  • the outer diameter of the cap may be from 1-10 times the diameter of the shaft; preferably, the outer diameter of the cap is between 1-3 times the diameter of the shaft.
  • the shaft as the remainder of the plug, may be configured to accomplish occlusion of an injection- or ocular surgery-related scleral hole.
  • the shaft may be thin enough, for example, to occlude the hole made by a 33 gauge, or thinner, needle after intravitreous injection, or may be as thick as 1-2 mm in diameter, or more, to occlude holes created during, for example, macular hole surgery.
  • the shaft may be of any size appropriate to occlude a particular discontinuity in the sclera.
  • the shaft is preferably at least as long as a sclera is thick, but may be shorter than the thickness of a sclera, or may be longer.
  • a typical sclera is 0.35-0.55 mm thick, but may be thicker or thinner. The thickness depends upon the particular individual, as well as the position of the discontinuity in the sclera; for example, the sclera tends to thin away from the iris and towards the retina. Where the shaft is longer than the thickness of a sclera, the shaft, when the plug comprising it is fully inserted, projects through the sclera an into the vitreous humor.
  • the surface of the shaft may be smooth or textured.
  • the surface of the shaft may be rough, ribbed or knurled so as to enhance contact between the plug and sclera, thereby reducing the potential for the plug to work its way out of the scleral hole.
  • the shaft may be ribbed or knurled directionally; that is, ribbed or knurled to promote insertion of the plug into the scleral hole and to discourage passage of the plug in the opposite direction, i.e., back out of the scleral hole.
  • the shaft is substantially cylindrical. In another embodiment, the shaft is substantially cylindrical along its entire length. In other embodiments, the shaft is ovoid, square, rectangular, square or rectangular with rounded edges, polygonal, or irregular in cross-section. In another embodiment, the shaft comprises a narrow portion and a wide portion. Typically, the shaft is attached to the cap through the narrow portion; the wide portion, distal to the cap, facilitates anchoring of the plug into the sclera. In one embodiment, the cross-sectional area of the wide portion is greater than that of the narrow portion.
  • the wide portion of the shaft may be manufactured in a variety of configurations. For example, the shaft may flare.
  • Such a flare may be substantially continuous along the length of the shaft, or may begin at any point along the length of the shaft.
  • the wide portion is a flange or protrusion from a portion of the main body of the shaft, e.g., from one side of the shaft.
  • Such a flange or protrusion may have any shape that facilitates maintenance of the plug within the scleral hole while not substantially increasing the difficulty of insertion or the potential for scleral damage during insertion.
  • the wide portion comprises a flange or other protrusion that substantially encircles the shaft.
  • the wide portion may be an inverted cone or frustum, wherein the larger radius of the frustum is wider than the diameter of the shaft.
  • the wider portion of the shaft is a cylinder having a radius larger than the radius of the shaft.
  • the wider portion of the shaft has substantially the same cross-sectional shape as the shaft, but a cross-sectional area larger than the cross-sectional area of the shaft.
  • the shaft comprises a thread spirally disposed along a portion or all of the length of the shaft, so that the shaft of the plug functions as a screw. In this embodiment, the thread may proceed clockwise or counterclockwise along the shaft.
  • the plug may be constructed so that the portion of the shaft distal to the cap comprises one or more flaps that may be folded against the shaft during insertion of the plug into the sclera, and which open, or fold away, from the shaft once the flap has been pushed completely through the sclera.
  • the one or more flaps would act as an anchor.
  • the wider portion of the shaft extends into the sclera itself, and serves as an anchor. In another embodiment, part or all of the wide portion of the shaft extends into the vitreous humor.
  • the end of the shaft distal to the cap may be flat, rounded, or tapered, or may be irregular.
  • the surface of the end may be substantially perpendicular to the longitudinal axis of the shaft, or may be tilted, giving the end of the shaft a barbed appearance.
  • the ocular plug comprises an opening extending at least the portion of the cap distal to the shaft, and, optionally, into the shaft.
  • the opening can be used, for example, to receive a wire of fixed gauge.
  • the wire is used to pick up the ocular plug and guide the ocular plug into a scleral discontinuity.
  • the ocular plug does not comprise a cap.
  • the ocular plug may comprise a shaft only.
  • the shaft may be formed in any of the configurations as for the shafts of a plug with a cap as discussed above.
  • the plug may simply be a cylinder, with a smooth, ribbed, knurled or textured surface, or may comprise one or more wide portions that can act as anchors.
  • the shaft (that is, the plug) comprises two wide portions.
  • the shaft is dumbbell-shaped.
  • the dumbbell shape may be accomplished, for example, by thickening the ends of the shaft so that the change in thickness from center of the shaft to either end is continuous; alternatively, the change in thickness from center of the shaft to either end is discontinuous.
  • the length of the shaft between the wide portions e.g., the ends of the dumbbell
  • the thickness of the sclera is at least the thickness of the sclera.
  • Ocular plugs may be pre-made to standard sizes, or may be custom-made to fill particular scleral holes or discontinuities, whether anticipated (as in the case of surgery) or unanticipated.
  • the invention provides an ocular plug, wherein said ocular plug has a shaft of a reproducible, standard size.
  • the ocular plug may also be custom-made for a particular discontinuity.
  • the standard or custom-made diameter size of a shaft for said ocular plug is a diameter sufficient to substantially occlude a scleral hole caused by passage of a 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12 or larger gauge needle.
  • the standard or custom-made diameter of a shaft for said ocular plug is about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5 millimeters, or wider.
  • the standard length of the shaft of said plug is 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1.00, 1.05, 1.10, 1.15, 1.20, 1.25, 1.30, 1.35, 1.40, 1.45 or 1.50 millimeters.
  • Ocular plugs may be made from umbilical cord biomaterial by any method used to create or produce molded devices.
  • the umbilical cord biomaterial used to make an ocular plug is a dried, decellularized biomaterial.
  • Ocular plugs may be made, for example, by stamping the plugs from a sheet of the biomaterial using a shaped stamp. Alternatively, the plugs may be cut from a sheet of the biomaterial, or may be formed by removal of unwanted material from a block of the biomaterial. In a preferred embodiment, an ocular plug is formed using a mold. Where the ocular plug is formed using a mold, the biomaterial is preferably first made into a liquid, slurry, paste, or similar material amenable to forming in a mold.
  • umbilical cord biomaterial is first reduced to a collection of particles; that is, the biomaterial is micronized.
  • the biomaterial may be micronized to a particle size of anywhere from about 1 micron to about 1 millimeter. Generally, the larger the particle size, the more porous the plug. Any method may be used to micronize the biomaterial, for example, ultrasound, physical shearing, homogenization, etc. Such micronization may be done dry (that is, by micronizing the biomaterial without any additional liquid), or may be done using a micronization liquid or carrier.
  • the liquid may be any physiologically acceptable liquid or solution that does not significantly degrade the biodegradable material, e.g., the tertiary structure of the proteins comprising the biomaterial.
  • the ratio of biomaterial to liquid is 25 mg/ml to 300 mg/ml, but more or less of the biomaterial may be used. Determination that a desired particle size has been achieved may be accomplished by any means known in the art, e.g., microscopic examination, comparison to bead size standards, etc.
  • the micronized biomaterial is injected or otherwise forced into the mold and allowed to set.
  • the wet micronized biomaterial is forced into the mold and is then frozen, e.g., at a temperature of from ⁇ 5° C. to ⁇ 160° C. (though higher or lower temperatures would also work) for a time sufficient to allow ice crystals to form and grow, e.g., 2 hours to several days.
  • the frozen plug is then freeze dried to substantial dryness, that is, to a water content of about 20% by weight or less.
  • any plugs formed using the biomaterial are freeze-dried to substantial dryness.
  • Freeze-drying is particularly preferred as the process allows for the development of pores in the biomaterial constituting the plug.
  • Plugs may also be heat-dried, but heat applied to dry the plugs is preferably not heat that would cause the breakdown of any component of the biomaterial.
  • an ocular plug formed from biomaterial may be dried at about 70° C., about 65° C., about 60° C., about 55° C., about 50° C. or about 45° C., or less than about 70° C., less than about 65° C., less than about 60° C., less than about 55° C., less than about 50° C. or less than about 45° C.
  • the biomaterial is preferably cross-linked to provide mechanical stability and integrity.
  • Crosslinking may be accomplished by any method known in the art; particularly preferred are radiation, chemical, or heat crosslinking. Radiation crosslinking is preferred. Radiation used may be any known in the art to be useful for such a purpose, for example, electron-beam or e-beam radiation, gamma radiation or ultraviolet radiation. E-beam radiation is preferred. See, e.g., Odland, U.S. Pat. No. 5,989,498 “E-beam Sterilization of Biological Materials,” The intensity of radiation used may be that ordinarily used for the sterilization of medical instruments.
  • the biomaterial may also be chemically crosslinked using any chemical crosslinking methodology known in the art, for example, thiol-thiol crosslinking, amide-amide crosslinking, amine-thiol crosslinking, amine-carboxylic acid and thiol-carboxylic acid crosslinking, etc., as appropriate for the material from which the plug is made.
  • the plug may also be heat crosslinked, typically using a thermal dehydration process. Most preferably the heat used for such crosslinking does not significantly degrade or structurally weaken the material in any way.
  • Plugs may be heat crosslinked, for example, by placing the plugs in a vacuum oven at 105° C. for 1-5 hours, or until the desired structural integrity or degree of crosslinking is achieved.
  • the umbilical cord biomaterial can be used in any other medical application in which like materials are used, e.g., as a patch for wound repair.
  • the umbilical cord biomaterial is used as a wrapping or covering for a replacement eye orb, e.g., a hydroxyapatite sphere.
  • the umbilical cord biomaterial can be used to repair or ameliorate a cardiac defect.
  • Such defects include, but are not limited to, cardiac wall defects, areas of necrosis due, e.g., to ischemia; repair of a cardiac valve; repair of patent foramen ovale, and the like.
  • the umbilical cord biomaterial is seeded or inoculated with cardiomyocytes. Immediately, or following culture to allow proliferation of the cardiomyocytes, the umbilical cord biomaterial is implanted into a cardiac defect as a living patch. In preferred embodiments, the umbilical cord biomaterial is placed so as to cover the defect completely.
  • kits comprising one or more pieces of umbilical cord membrane biomaterial.
  • each of the one or more pieces the biomaterial is individually sterilely wrapped, e.g., in a peel-pouch.
  • the kit comprises a piece of umbilical cord biomaterial that is at least about 2 ⁇ 8 cm.
  • said kit comprises a piece of umbilical cord biomaterial approximately the size of an eardrum.
  • the kit may comprise one or more pieces of umbilical cord biomaterial and any other medical device, disposable or drug that would facilitate treatment of a disease, disorder or condition treatable using the biomaterial.
  • each piece of the umbilical cord biomaterial in the kit is provided as a single sheet or patch in a sterile container or wrapping separate from the remainder of kit contents.
  • the kit comprises two or more pieces of umbilical cord biomaterial, separately wrapped or contained.
  • said kit comprises a support for the umbilical cord biomaterial.
  • the support may be a natural or a synthetic material.
  • said support is a plastic film, plastic sheet, or a stretchable plastic wrap.
  • said kit comprises one or more disposables, e.g., paper tissues or towels, mats, cotton swabs, plastic or rubber gloves, disposable forceps, or the like.
  • said disposables are bandages, means for sterilizing skin, swabs, gloves, or sterile sheets.
  • said kit comprises an anti-infective agent, for example, an antibiotic ointment, cream, or spray.
  • said kit comprises a piece of umbilical cord biomaterial and one or more wound healing agents.
  • said wound healing agent is PDGF, TGF, hyaluronic acid, fibrin, or fibronectin.
  • said kit comprises umbilical cord biomaterial and a means for applying compression to a part of the body.
  • the kit comprises an instruction sheet suitable for use by a non-medical end user; an instruction sheet suitable for use by an end user in a medical profession; or a materials safety data sheet; or a combination thereof.
  • the following example demonstrates one method of preparing umbilical cord biomaterial.
  • a sterile field was set.
  • the placenta was removed from the transport container and placed into a sterile stainless steel tray. Using surgical damps and scissors, the umbilical cord was cut off approximately 2 inches from the placental disc.
  • the umbilical cord was rinsed with sterile 0.9% NaCl solution as many times as necessary to remove as much blood as possible; optionally, fingers were used to squeeze remaining blood from vessels.
  • the umbilical cord was optionally placed in a separate sterile container cup prefilled with sterile 0.9% NaCl solution, if the cord did not have to be processed immediately.
  • the harvested umbilical cord was placed in a refrigerator at 4° C. until use.
  • the placental disk was placed back into the transport container to be utilized for other projects, or discarded.
  • the umbilical cord was processed as follows. The umbilical cord was removed from the specimen container, and squeezed to remove any remaining blood from vessels prior to introducing the umbilical cord to a processing tray. The umbilical cord was placed into a sterile stainless steel processing tray, and cut into segments 12 to 15 cm in length. The umbilical cord vein was then located for each segment, and canalized using an Adson bayonet forceps or grooved director. The vein and umbilical cord were then cut longitudinally, using scissors, until both the vein and umbilical cord were fully open. The umbilical cord, was placed on the processing tray with the opened vein side facing upward.
  • the umbilical cord and vein were then bluntly dissected longitudinally between the vein wall and the umbilical cord wall with sterile tweezers or mosquito clamps. When both sides were separated, the vein was carefully removed. After vein removal, the two arteries were located and removed in the same manner.
  • the resulting umbilical cord membrane biomaterial was placed in either in saline solution or in 1% deoxycholic acid solution (a decellularizing solution) and stored at 4° C. until serological testing results become available.
  • the umbilical cord membrane obtained as outlined above, was kept in sterile 0.9% saline solution or 1% deoxycholic acid solution for 10-20 days at 4° C. until serological test results, if ordered, were available. Saline solution, where used, was changed every 3 days. 1% deoxycholic acid solution, when used, was changed every 5 days.
  • a sterile field was set with a new set of sterilized trays as above.
  • the Umbilical cord membrane was removed from the refrigerator and placed into a stainless steel processing tray.
  • Sterile 0.9% saline solution is added to cover the bottom of the tray.
  • All, or substantially all, residual deoxycholic solution, where used, was removed, and remaining cells and debris were removed from both sides of the tissue using a cell scraper and sterile tweezers.
  • Sterile 0.9% saline solution was used as needed to aid in removal of the cells and debris.
  • the umbilical cord membrane was rinsed three times in a separate stainless steel rinsing tray filled with sterile 0.9% saline solution. The saline solution was changed between each cleaning step.
  • the umbilical cord membrane was then placed into a new sterile specimen container containing about 150 mL saline solution, and placed on a rocking platform for agitation for 5 minutes at setting #6. The scraping and rinsing steps were repeated once as necessary.
  • the umbilical cord membrane was then placed into a sterile specimen container containing 150 mL sterile water, and placed on a rocking platform for agitation for 20 minutes at setting #6. This rinsing step was repeated three times.
  • a TYVEK® sheet was placed onto a stainless steel processing tray.
  • the cleaned umbilical cord membrane segments were removed from the specimen container one piece at a time, and excess fluid was gently squeezed out.
  • the membrane segments were then placed on the surface of the TYVEK® sheet, epithelium side up, and gently stretched until flat.
  • the membrane was then dried at about 50° C. ⁇ 1.0° C. in a vacuum dryer.
  • Sterile gauze was placed on the drying platform of the vacuum dryer, covering an area slightly larger than the area of the TYVEK® sheet. The total thickness of the gauze layer did not exceed the thickness of one folded 4 ⁇ 4 gauze.
  • a sheet of silicone framing mesh was placed on top the gauze, smooth side up.
  • the TYVEK® sheet with the tissue was then placed on the heat dryer platform on top of the silicone mesh. Another TYVEK® sheet was then placed on top of the tissue. A piece of PVC wrap film was then cut large enough to cover the entire drying platform, and pulled so that the film pulled tightly against the TYVEK® sheet (that is, was “sucked in” by vacuum) and so that there were no air leaks and no wrinkles over the tissue area).
  • the vacuum pump was then set to approximately ⁇ 22 inches Hg, and heat/vacuum drying was allowed to proceed for a total of about 120 minutes. Approximately 30-45 minutes into the drying process, the sterile gauze layer was replaced.
  • a new sterile field was set with a sterilized drying kit and cutting board. With the pump still running, the plastic film was removed from the TYVEK®, and the sheet and tissue were placed on a cutting board with the epithelium side of the tissue facing upward. The dried membrane (now umbilical cord biomaterial) was then gently removed from the TYVEK® sheet. The biomaterial segments were then cut with a scalpel into segments of a specified size, typically 2 ⁇ 2 cm or 1 ⁇ 1 cm. The dried, sized umbilical cord biomaterial was then placed and sealed into a peel-pouch package.
  • Umbilical cord membrane from a 23.5 hour-old placenta was collected and processed as in Example 1 up to the point of drying. The final size of the membrane was approximately 35 cm by 4 cm. The membrane was cut into three pieces approximately 10 cm long. The pieces were arranged so as to overlap by about 2 cm on the long edge, and were dried at 50° C. between two sheets of TYVEK®.
  • Umbilical cord membrane can also be laminated by placing two or more pieces of the biomaterial, interior (of the umbilical cord) side down, on a substrate in a mounting frame. The laminated membrane is then placed in a gel dryer and dried to substantial dryness ( ⁇ about 20% water content by weight) to produce a laminated umbilical cord biomaterial.
  • Another method of constructing a thicker biomaterial is to laminate intact umbilical cord (including Wharton's jelly but lacking arteries and vein).
  • the intact cord is then processed, e.g., by rinsing, soaking in a solution such as a buffered saline solution, e.g., phosphate buffered saline, or a mild ionic or nonionic detergent solution.
  • a solution such as a buffered saline solution, e.g., phosphate buffered saline, or a mild ionic or nonionic detergent solution.
  • the cord is dried in a vacuum dryer to create an intact, double-layer biomaterial. Two or more layers of this double-layer material can be laminated by layering the biomaterials and drying further in a heated vacuum dryer.
  • the drying/dehydration process can be heat drying or any other processes as described below.
  • HUC human umbilical cord membrane
  • FIG. 1 summarizes the results of the rehydration for HUC incubated for 10 and 20 days.
  • the initial water uptake of the control (nonsterilized samples) was much higher for HUC incubated for 20 days than for HUC incubated for 10 days, possibly due to the loosening of the membrane proteins by the detergent effect of the deoxycholic acid in the D-cell solution.
  • Water uptake and equilibrium water content closely matched for the 10 and 20 days samples that were sterilized at all radiation doses. There was a linear decrease in the water uptake and the equilibrium water content of both sets of samples with increasing radiation dose. Even at the highest radiation dose, the membranes took up at least their own weight in water.
  • the average dry thickness of the HUC was ⁇ 70 ⁇ m, with the 10 and 20 day samples, having average thickness of 57 and 86 ⁇ m respectively (Table 1 and Table 2).
  • Table 1 and Table 2 There appeared to be no correlation between radiation dose and dry thickness of the HUC.
  • After rehydration there was a marked difference in thickness between the sterilized and non-sterilized samples.
  • the difference observed can be due to cross-linking of collagen molecules caused by irradiation.
  • FIG. 3 graphically summarizes the denaturation temperature results for the HUC samples. The results are identical for samples incubated for 10 and 20 days. Onset and peak temperatures were only a few degrees different for each of the samples, and both onset and peak temperatures decreased linearly with increasing radiation dose. There was very low variability in the results.
  • the umbilical cord biomaterial was determined to have a superior suture pull-out strength compared to dried human amniotic membrane.
  • one short side of a 1 ⁇ 2 section of umbilical cord biomaterial was glued to vellum paper, and the other short side sutured to a second piece of vellum paper, as depicted in FIG. 5A .
  • the pieces of vellum paper were held by grips, and a load was applied to the suture at a rate of about 12.7 mm/min.
  • the umbilical cord biomaterial demonstrated an average pull-out resistance of about 1.4 Newtons (N), with a range of about 0.75 N to about 2.4 N, while the dried amniotic membrane demonstrated a pull-out resistance averaging about 0.3 N. See FIG. 5B .
  • This study evaluated the host response to an implant made of umbilical cord biomaterial during absorption following subcutaneous implantation in a rat model.
  • Test materials for implantation consisted of umbilical cord biomaterial or high density polyethylene (HDPE; control).
  • Umbilical cord biomaterial was provided as dried umbilical cord membrane measuring approximately 1 cm ⁇ 1 cm sections prepared in either 0.9% NaCl (Test article A; non-decellularized) or 1% deoxycholic acid (Test articles B and C; decellularized).
  • the 16 rats used in the experiments were ten week old male Rattus norvegicus strain H1A®:(SD)CVF® (Hilltop Lab Animals, Inc.). Animal weight at the time of implantation ranged from 344 grams to 392 grams. Maintenance of animals during the experiment conformed to Standard Operating Procedures based on the “Guide for the Care and Use of Laboratory Animals”.
  • each rat was identified and weighed. Groups of four animals were arbitrarily assigned to be terminated 1 week, 3 weeks, 6 weeks or eight weeks after implantation (Table 5).
  • the animals were anesthetized by intraperitoneal injection of ketamine hydrochloride and xylazine (66 mg/kg and 9 mg/kg, respectively) dosed at 2.25 ml/kg.
  • the implant region was scrubbed with a germicidal soap and wiped with 70% alcohol. Separate incisions were made on each side of the back through the skin and parallel to the lumbar region of the vertebral column. A pocket was formed by blunt dissection in the subcutaneous tissue on each side of the back.
  • One section of the test material was implanted into each pocket such that it lay as flat in the pocket as reasonably possible.
  • a nonsorbable suture was cut into approximately 1 cm length sections and placed at each test article implantation site as a location marker.
  • One section of the negative control article (HDPE) was similarly implanted caudally to the sections of the test article. The skin was closed with wound clips.
  • HDPE negative control article
  • the designated animals were weighed and euthanized by carbon dioxide inhalation. Macroscopic observation of the viscera was conducted. The general appearance of the skin at the implantation sites was recorded. The implant sites were exposed by incision along the midline from the proximal to the distal end of the rat, and the skin was gently pulled away from the implantation site. The implanted materials were measured to the nearest millimeter (length and width) and the color and consistency of the surrounding tissue was documented. The sites were also photographed. The implant sites and any abnormal tissues were excised and preserved in 10% neutral buffered formalin (NBF) until further processing. After fixation, the implant sites and any abnormal tissues were histologically processed (embedded, sectioned and stained with hematosylin and eosin) for microscopic evaluation by a pathologist.
  • NBF neutral buffered formalin
  • Implantation sites were evaluated to assess any change in the integrity of the form of the test material.
  • the local tissue response was evaluated and compared to the reactions at the negative control article sited.
  • the evaluation included characterization of the test material in regard to acute inflammation, chronic inflammation, granulation tissue formation, foreign body reaction, and foreign body giant cell formation.
  • the formation and the thickness change of the fibrous capsule around the implants, the change in implants' characteristics at degradation were also evaluated. Microscopic cellular changes were graded according to severity on a scale of 0 to 4.
  • Implant absorption Implant size at the various termination points is presented in Tables 6 and 7. In several cases, the implant was found to have been completely absorbed by the host. Average implant sizes for the test articles at each of the terminations points is shown in Table 8.
  • both decellularized and non-decellularized umbilical cord biomaterial have excellent biocompatibility.
  • Animal responses to both biomaterials was similar to control USP grade HDPE.
  • No fibrous tissue encapsulations developed during the course of the implantation study, whereas HDPE implants developed fibrosis at later stages of the study.
  • both decellularized and non-decellularized biomaterials were biodegradable, it was noted that implanted decellularized membranes lasted longer (e.g., about 6 to 8 weeks) than non-decellularized biomaterials (about 3 to 6 weeks).

Abstract

The present invention provides a biomaterial comprising a mammalian umbilical cord membrane. The biomaterial can additionally comprise Wharton's jelly and/or one or more umbilical cord vessels. The biomaterial is preferably dry, and can be flat, tubular, or shaped to fit a particular body structure. The invention further provides laminates comprising at least one layer of an umbilical cord membrane biomaterial. The invention further provides methods of making the biomaterial, and laminates comprising the biomaterial, and methods of using the biomaterial.

Description

    1. FIELD OF THE INVENTION
  • The present invention generally relates to biomaterials derived from the umbilical cord membrane, compositions comprising the umbilical cord membrane, and methods of treatment using the compositions.
  • 2. BACKGROUND OF THE INVENTION
  • The repair or treatment of various body tissues, such as skin, organs, and the like, has been accomplished using collagen compositions, including membranes comprising collagen. A need exists, however, for additional such compositions, including ones that are able to handle loads well.
  • 3. SUMMARY OF THE INVENTION
  • The present invention provides biomaterials derived from umbilical cord. In certain embodiments, the biomaterial comprises an umbilical cord membrane. The present invention provides methods of making the umbilical cord biomaterial, and of using the biomaterial, e.g., to repair organs and tissues.
  • In one aspect, the present invention provides a biomaterial comprising an isolated mammalian umbilical cord membrane. In a specific embodiment, the biomaterial comprises at least one umbilical vessel (e.g., an umbilical artery or umbilical vein). In another specific embodiment, the biomaterial comprises Wharton's jelly. In another specific embodiment, the biomaterial comprises Wharton's jelly but lacks umbilical vessels. In another specific embodiment, the biomaterial comprises the umbilical cord membrane, Wharton's jelly, and all three umbilical vessels. The umbilical cord biomaterial is not, however, an umbilical cord that has not been processed in any manner.
  • In more specific embodiments of any of the above embodiments, the biomaterial comprises less water by weight than native umbilical cord membrane in vivo, e.g., the biomaterial comprises 40%, 30%, 20%, 10% or less water by weight. In other more specific embodiments, the biomaterial comprises at least 60%, at least 70%, or at least 80% water by weight. In another specific embodiment, the biomaterial is decellularized prior to use. In another specific embodiment, the biomaterial is not decellularized prior to use. In another specific embodiment, the umbilical cord membrane of the biomaterial is cut or slit longitudinally. In another specific embodiment, the biomaterial is substantially flat. In another specific embodiment, the biomaterial is substantially tubular. In another specific embodiment, the biomaterial comprises artificially crosslinked proteins.
  • In another specific embodiment, the umbilical cord biomaterial comprises an exogenous bioactive molecule, that is, a bioactive molecule not obtained from the umbilical cord used to make the biomaterial. In a more specific embodiment, said bioactive molecule is a cytokine or growth factor. In another more specific embodiment, said bioactive molecule is an extracellular matrix protein. In another more specific embodiment, said extracellular matrix protein is collagen, fibronectin, elastin, vitronectin, or hyaluronic acid. In a more specific embodiment, said bioactive molecule is hyaluronic acid. In an even more specific embodiment, said hyaluronic acid is crosslinked to said umbilical cord membrane. In another specific embodiment, said biomaterial comprises an exogenous polymer. In a more specific embodiment, said exogenous polymer is a synthetic biodegradable polymer or an anionic polymer. In another more specific embodiment, said synthetic biodegradable polymer is a polyhydroxyalkanoate. In a more specific embodiment, said anionic polymer is dextran sulfate or pentosan polysulfate. In another specific embodiment, the bioactive molecule is an antibiotic, a hormone, a growth factor, an anti-tumor agent, an anti-fungal agent, an anti-viral agent, a pain medication, an anti-histamine, an anti-inflammatory agent, an anti-infective agent, a wound healing agent, a wound sealant, a cellular attractant or a scaffolding reagent. In another specific embodiment, the bioactive molecule is a small molecule, e.g., a small organic molecule, e.g., a drug.
  • In another specific embodiment, the umbilical cord biomaterial comprises a hydrogel composition. In a more specific embodiment, said hydrogel composition comprises a polyvinyl alcohol, a polyethylene glycol, a hyaluronic acid, a dextran, or a derivative or analog thereof.
  • The umbilical cord biomaterial can also comprise, e.g., be seeded with, one or more types of stem and/or progenitor cells. In a specific embodiment, the biomaterial comprises an exogenous stem cell, that is, a stem cell not native to the umbilical cord from which the biomaterial is derived, or from an individual different from the umbilical cord donor. In specific embodiments, the stem cell is a placental stem cell, a mesenchymal stem cell, an embryonic stem cell, an adult stem cell, or a somatic stem cell. In a more specific embodiment, the somatic stem cell is a neural stem cell, a hepatic stem cell, a pancreatic stem cell, an endothelial stem cell, a cardiac stem cell, a stromal cell, or a muscle stem cell. In another specific embodiment, the umbilical cord biomaterial comprises an exogenous adult (e.g., fully-differentiated or committed progenitor) cell.
  • In another embodiment, the invention provides laminates comprising the umbilical cord biomaterial. Such laminates can comprise layers of umbilical cord biomaterial, or one or more layers of umbilical cord biomaterial layered with one or more layers of another material. The layers can be substantially aligned, or can be offset, e.g., overlapping, to form, e.g., a laminate of biomaterial longer and/or wider than an umbilical cord. Thus, in one embodiment, the invention provides a laminate comprising a plurality (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) of layers, wherein at least one of the layers comprises an umbilical cord biomaterial. In a specific embodiment, said laminate comprises one or more layers of an amniotic membrane material. In a more specific embodiment, said amniotic membrane material is a dried amniotic membrane laminated to said umbilical cord membrane. In another more specific embodiment, said amniotic membrane material has been sonicated or otherwise disrupted. In another specific embodiment, at least some of the proteins in at least one of the layers of the laminate are artificially crosslinked, either to other proteins in the same layer, or to proteins in one or more adjoining layers.
  • The invention further provides laminates comprising the umbilical cord biomaterial and other useful compounds. For example, in one embodiment, the invention provides a laminate comprising umbilical cord biomaterial and an exogenous stem cell or an exogenous adult cell. In another embodiment, the laminate comprises a hydrogel composition. In a specific embodiment, said hydrogel composition comprises a polyvinyl alcohol, a polyethylene glycol, a hyaluronic acid, a dextran, or a derivative or analog thereof.
  • The invention also provides a method of producing a biomaterial comprising isolating and decellularizing a biomaterial comprising an umbilical cord membrane. In a specific embodiment, the biomaterial comprises Wharton's jelly, or one or more umbilical cord vessels. In another embodiment, the method comprises isolating said umbilical cord membrane from one or more umbilical cord vessels. The method may further comprise drying said biomaterial to less than 20% water by weight. In various embodiments, said biomaterial is dried at about 26° C. to about 65° C., or at about 35° C. to about 50° C. The composition can, for example, be dried with a hygroscopic compound. In another embodiment, the biomaterial is freeze dried, or dried using vacuum. Said decellularizing may, for example, comprise contacting the umbilical cord membrane with a detergent solution, for example, a solution comprising 0.01-2.0% deoxycholic acid. The detergent may be a nonionic detergent, an anionic detergent, or a combination thereof. In specific embodiments, said detergent is Triton X-100 or sodium dodecyl sulfate, or a combination thereof.
  • The invention further provides methods of making a laminate comprising an umbilical cord membrane biomaterial. Such a method can comprise crosslinking at least some proteins in at least one layer of said laminate. The method of making the laminate, in one embodiment, comprises layering a plurality of umbilical cord membranes in contact with each other to form a laminate. In a specific embodiment, one or more of said membranes comprise less than 20% water by weight prior to said layering. In another specific embodiment, said laminate is dried to less than 20% water by weight after said layering. In other specific embodiments, one or more of said membranes comprise at least 60%, at least 70% or at least 80% water by weight. In another specific embodiment, said laminate comprises at least 60%, at least 70%, or at least 80% water by weight.
  • The invention also provides for the use of the umbilical cord membrane biomaterial to deliver one or more therapeutic agents to a subject. In one embodiment, the invention provides a method of delivering a therapeutic agent to a subject comprising contacting the subject with an umbilical cord membrane biomaterial, or laminate thereof, wherein said composition or laminate comprises a therapeutic agent. In a specific embodiment, said subject is a human. In other specific embodiments, said therapeutic agent is an antibiotic, an anti-cancer agent, an anti-bacterial agent, an anti-viral agent, a vaccine, an anesthetic, an analgesic, an anti-asthmatic agent, an anti-inflammatory agent, an anti-depressant, an anti-diabetic agent, an anti-psychotic, a central nervous system stimulant, a hormone, an immunosuppressant, a muscle relaxant, or a prostaglandin.
  • The invention further provides a method of using an umbilical cord biomaterial, e.g., one that comprises an isolated umbilical cord membrane, in the repair of a tympanic membrane deformity, comprising contacting said tympanic membrane with such a biomaterial. In a specific embodiment, the deformity is a perforation, which may be, e.g., a central perforation or a marginal perforation. The perforation may be caused by, e.g., trauma, or as part of a surgical procedure. In a more specific embodiment, said contacting is sufficient to occlude the perforation. In another more specific embodiment, said perforation has not healed spontaneously within two months of developing the perforation. In another specific embodiment, said deformity is an atelectatic tympanic membrane, a deformity relating to a choleastoma, a retraction pocket or a deformity resulting from a tympanosclerosis.
  • The invention also provides for the repair of other deformities using the umbilical cord membrane biomaterial. For example, the invention provides a method of repairing a nasal septum having a perforation, comprising contacting said septum with a biomaterial comprising an umbilical cord biomaterial, e.g., one comprising an isolated mammalian umbilical cord membrane. In a more specific embodiment, said method comprises contacting cartilage within said septum with said biomaterial.
  • As used herein, “exogenous bioactive compound” means a molecule introduced into the biomaterial that has a detectable effect on one or more biological systems. Examples of bioactive compounds are listed, without limitation, in Section 5.1.2, below.
  • As used herein, “substantially flat,” in reference to umbilical cord biomaterial, means that the majority of the umbilical cord biomaterial is planar, but can comprise differences in thickness, whether naturally-occurring or artificially-induced, e.g., natural variations in membrane thickness, ridges, patterns, raised areas, warps, and the like, induced, e.g., during drying on a mesh.
  • As used herein, “substantially tubular,” in reference to umbilical cord biomaterial, means that the majority of the umbilical cord biomaterial is tubular (i.e., circular, ovoid, or irregular in cross-section, and comprising the material defining an interior or lumen). The substantially tubular biomaterial can be a closed tube or partially open tube.
  • As used herein, “umbilical cord biomaterial” means any biomaterial comprising an isolated umbilical cord outer membrane, particularly a biomaterial manufactured or derived from umbilical cord, and includes, e.g., dried whole umbilical cord, dried umbilical cord membrane (with or without vessels), umbilical cord membrane laminated with a second material, etc. The term does not, however, encompass an umbilical cord that has not been treated or manipulated in any manner, that is, an umbilical cord that has not been modified from an in vivo state.
  • 4. BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A and 1B depict the effect of radiation dose on water uptake [⋄, (Ww−Wd)/Wd*100] and equilibrium water content [□, (Ww−Wd)/Ww*100] for human umbilical cord biomaterial incubated for 10 (A) and 20 (B) days in 1% D-cell solution.
  • FIGS. 2A and 2B depict a comparison of the change in thickness (□) and water uptake (▪) during rehydration of gamma sterilized human umbilical cord biomaterial incubated for 10 (A) and 20 (B) days in 1% D-cell solution. Error bars indicate standard deviation.
  • FIG. 3 depicts a comparison of the denaturation temperature of rehydrated human umbilical cord membrane that had previously been incubated in 1% D-cell solution for 10 or 20 days. There is essentially no difference between the different incubation times and there is a linear decrease in denaturation temperature with increasing radiation dose.
  • FIG. 4 depicts a schematic of tensile testing of membrane samples based on American Society for Testing and Materials (ASTM) standard D1708. A dog bone shaped sample 1 is mounted in a rectangular support of vellum paper. The membrane and the support are incubated in PBS for 1 hour at 37° C. The support keeps the membrane flat and eases loading into the mechanical tester. The tester comprises and upper grip 2 and a lower grip 3. Once the support and membrane have been secured in the grips of the mechanical tester with glue 4, the supporting struts 5 are cut and the sample can be tested with no interference from the vellum support.
  • FIG. 5A depicts a suture pull-out assay apparatus with an lower grip 10 holding vellum paper 11 to which a 1×2 cm section of umbilical cord biomaterial 12 is glued, and an upper grip 13 attached to a suture 14 that passes through the umbilical cord biomaterial. Force was applied upwards through the suture at approximately 12.7 mm/min. FIG. 5B depicts results of a comparison of the human umbilical cord biomaterial (HUC) and dried human amniotic membrane pull-out resistance in Newtons (N).
  • 5. DETAILED DESCRIPTION OF THE INVENTION 5.1 Umbilical Cord Biomaterial
  • 5.1.1 Description
  • The umbilical cord biomaterial of the invention is derived from a mammalian umbilical cord or part thereof that comprises an umbilical cord membrane (that is, the outer membrane of the umbilical cord). The umbilical cord is a substantially tubular organ, typically 10-15 cm in length, that connects the fetus to the placenta and houses the umbilical vessels. The umbilical cord comprises an outer membrane that wraps around two umbilical arteries and one umbilical vein, which are contained within a ground substance known as Wharton's jelly. The main components of Wharton's jelly are proteoglycans. Wharton's jelly also contains large, stellate fibroblasts and macrophages.
  • The umbilical cord membrane biomaterial of the invention typically comprises only the umbilical cord membrane, but can also comprise Wharton's jelly and/or one or more of the umbilical vessels. In one embodiment, the umbilical cord biomaterial is an umbilical cord membrane substantially isolated from the remaining umbilical cord components (e.g., Wharton's jelly and umbilical vessels). In another embodiment, the umbilical cord biomaterial comprises an umbilical cord membrane and Wharton's jelly (that is, the ground material in which the umbilical cord vessels are contained in the intact umbilical cord) that are isolated from the remaining umbilical cord components (e.g., umbilical cord vessels). In another specific embodiment, the umbilical cord membrane biomaterial comprises the membrane, Wharton's jelly and one or more umbilical cord vessels. In another embodiment, the umbilical cord biomaterial comprises an isolated umbilical cord (e.g., comprising Wharton's jelly and vessels, Wharton's jelly only, or vessels only) that has been flattened into a sheet or strip. The umbilical cord membrane biomaterial can be a substantially tubular structure from which the contents (Wharton's jelly and vessels) have been removed. The biomaterial can also comprise an umbilical cord membrane that has been slit or cut for part or all of the length of the umbilical cord to expose the contents of the umbilical cord.
  • In a specific embodiment, the biomaterial, comprising umbilical cord membrane and/or Wharton's jelly and/or vessels) can be decellularized. In another specific embodiment, the biomaterial comprises umbilical cord membrane-associated cells or Wharton's jelly-associated cells that have been killed. In another specific embodiment, the biomaterial comprises umbilical cord membrane-associated cells or Wharton's jelly-associated cells that have been maintained in a living state.
  • The umbilical cord biomaterial of the invention can be derived from the umbilical cord of any mammal, for example, from equine, bovine, porcine or catarrhine sources, but is most preferably derived from human umbilical cord.
  • The umbilical cord biomaterial is preferably dry or substantially dry. In a preferred embodiment, the umbilical cord biomaterial is substantially dry, i.e., is 20% or less water by weight. When dry, the umbilical cord biomaterial can be substantially flat. The dry biomaterial may, in another embodiment, substantially retain the shape of the native umbilical cord, that is, the dry membrane may be substantially tubular. The umbilical cord biomaterial can also be shaped to assume different conformations, e.g., can be curved, cut, molded, or the like, to fit to a part of the body.
  • In another preferred embodiment, the umbilical cord biomaterial has not been protease-treated, heat-denatured or artificially (e.g., chemically or radiologically) crosslinked. In another embodiment, the umbilical cord biomaterial comprises artificially crosslinked proteins, e.g., chemically or radiologically crosslinked collagen. In other embodiments, the umbilical cord biomaterial contains substantially no structural proteins that are artificially crosslinked. For example, in one embodiment, the umbilical cord biomaterial is not fixed. A preferred umbilical cord biomaterial is produced by the methods disclosed herein (see Section 5.1.4, below, and Examples 1 and 2).
  • When the umbilical cord biomaterial is substantially dry, it is typically about 0.001 g/cm2 to about 0.006 g/cm2. In a specific embodiment, a single layer of the acellular, dried umbilical cord biomaterial is approximately 50 microns to 250 microns in thickness, typically approximately 90 microns to 220 microns in thickness. In other specific embodiments, a single layer of the umbilical cord biomaterial is approximately 75-200 microns, 100-200 microns, 100-220 microns, 120-220 microns, or 150-250 microns in thickness in the dry state. In another embodiment, the average thickness of the umbilical cord biomaterial is about 157 microns (e.g., ±20%). In another specific embodiment, the pull out strength of the dried umbilical cord biomaterial is approximately 1.5 Newtons (N), compared to a pull out strength of a dried amniotic membrane material at approximately 0.4N.
  • Generally, the umbilical cord biomaterial is sided, that is, the umbilical cord biomaterial comprises an epithelial side (from the interior of the umbilical cord), and an outer, mesothelial side (from the exterior of the umbilical cord).
  • Generally, the umbilical cord biomaterial is non-immunogenic.
  • In various embodiments, the umbilical cord biomaterial comprises particular cytokines, i.e., interleukin (IL)-1b, IL-2, IL-3, IL-6, IL-7, IL-12, IL-15, IFN-α, MIP-1b, and/or MCP-1.
  • In one embodiment, the umbilical cord biomaterial is translucent. In other embodiments, the umbilical cord biomaterial is opaque, or is colored or dyed, e.g., permanently colored or dyed, using a medically-acceptable dyeing or coloring agent. Such an agent may be adsorbed onto the biomaterial, or the biomaterial may be impregnated or coated with such an agent. In this embodiment, any known non-toxic, non-irritating coloring agent or dye may be used.
  • The umbilical cord biomaterial comprises of collagen (types I, III and IV; typically about 75%-80%% of the matrix of the biomaterial), fibronectin, elastin, and may further comprise glycosaminoglycans, (GAGs, e.g., hyaluronic acid) and/or proteoglycans. Typically, laminin is not present, or is present in trace amounts (i.e., less than 0.1% of the dry weight of the biomaterial). Typically, the umbilical cord biomaterial comprises collagen types I, III, IV, V, VI and VII. In certain embodiments, the umbilical cord biomaterial can comprise non-structural components, such as, for example, one or more growth factors, e.g., platelet-derived growth factors (PDGFs), vascular-endothelial growth factor (VEGF), fibroblast growth factor (FGF), transforming growth factor-β1, and the like. In certain embodiments, the umbilical cord biomaterial comprises growth factors such as FGF, b-FGF, EGF, IGF-1, PDGF and TGF-β. The composition of the umbilical cord biomaterial may thus be ideally suited to encourage the migration of fibroblasts and macrophages, and thus, e.g., the promotion of wound healing.
  • In one embodiment, the invention provides an umbilical cord biomaterial wherein at least 50% of the dry weight of the biomaterial is collagen I. In various more specific embodiments, at least 55%, 60%, 65% or 70% of the dry weight of the biomaterial is collagen I. In another specific embodiment, the invention provides an umbilical cord biomaterial wherein at most 5% of the dry weight of the biomaterial is collagen III. In various more specific embodiments, at most 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4.0%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3.0% or 2.9% of the dry weight of the biomaterial is collagen III. In another specific embodiment, the invention provides an umbilical cord biomaterial wherein at least 4% of the dry weight of the biomaterial is collagen IV. In various more specific embodiments, at least 5%, 6%, 7%, 8%, 9%, 10% or 11% of the dry weight of said biomaterial is collagen IV. In another specific embodiment, the invention provides an umbilical cord biomaterial wherein at most 4% of the dry weight of the biomaterial is elastin. In various more specific embodiments, at most 3.8%, 3.6%, 3.4%, 3.2%, 3.0%, 2.8%, 2.6%, 2.4%, 2.2%, 2.0%, or 1.8% of the dry weight of the biomaterial is elastin. In another specific embodiment, at least 4% of the dry weight of the biomaterial is glycosaminoglycan. In various more specific embodiments, at least 4.1%, 4.2%, 4.3%, 4.4%, 4.5% or 4.6% of the dry weight of said biomaterial is glycosaminoglycan.
  • The umbilical cord biomaterial may be used in a single-layered format, for example, as a single-layer sheet or an un-laminated membrane. Alternatively, the umbilical cord biomaterial may be used in a double-layer or multiple-layer format, e.g., the umbilical cord biomaterial may be laminated. Lamination can provide greater stiffness and durability, for example, during the healing process. The umbilical cord biomaterial may be, for example, laminated as described below (see Section 5.1.7).
  • The umbilical cord biomaterial may further comprise collagen from a non-umbilical cord source. For example, one or more layers of umbilical cord biomaterial may comprise, e.g., be coated or impregnated with, or layered with, purified extracted collagen. Such collagen may be obtained, for example, from commercial sources, or may be produced according to known methods, such as those disclosed in U.S. Pat. Nos. 4,420,339, 5,814,328, and 5,436,135, the disclosures of which are hereby incorporated by reference. Such collagen can also be obtained from a placental source, including a placenta obtained from the same donor as the umbilical cord biomaterial.
  • The umbilical cord biomaterial can comprise one or more compounds or substances that are not present in the umbilical cord material from which the biomaterial is derived. Moreover, the umbilical cord biomaterial can comprise non-naturally-occurring amounts of one or more compounds or substances that are normally present in the umbilical cord from which the biomaterial is derived. For example, the umbilical cord biomaterial can comprise, e.g., can be impregnated with, a bioactive compound, such as those listed in Section 5.1.2, below. Such bioactive compounds include, but are not limited to, small organic molecules (e.g., drugs), antibiotics (such as, for example, Clindamycin, Minocycline, Doxycycline, Gentamycin), hormones, growth factors, anti-tumor agents, anti-fungal agents, anti-viral agents, pain medications, anti-histamines, anti-inflammatory agents, anti-infectives including but not limited to silver (such as silver salts, including but not limited to silver nitrate and silver sulfadiazine), elemental silver, antibiotics, bactericidal enzymes (such as lysozyme), wound healing agents (such as cytokines including but not limited to PDGF, TGF; thymosin), hyaluronic acid as a wound healing agent, wound sealants (such as fibrin with or without thrombin), cellular attractant and scaffolding reagents (such as added fibronectin) and the like. In a specific example, the umbilical cord biomaterial may be impregnated with at least one growth factor, for example, fibroblast growth factor, epithelial growth factor, etc. The biomaterial may also be impregnated with small organic molecules such as specific inhibitors of particular biochemical processes e.g., membrane receptor inhibitors, kinase inhibitors, growth inhibitors, anticancer drugs, antibiotics, etc. Impregnating the umbilical cord biomaterial with a bioactive compound may be accomplished, e.g., by immersing the biomaterial in a solution of the bioactive compound of the desired concentration for a time sufficient to allow the biomaterial to absorb and to equilibrate with the solution. In a specific embodiment, the biomaterial so impregnated is a dried biomaterial, and the solution partially or fully re-hydrates the biomaterial, compared to an umbilical cord or umbilical cord membrane in vivo. In another embodiment, the biomaterial is impregnated prior to drying the biomaterial, e.g., to substantial dryness.
  • In other embodiments, the umbilical cord biomaterial may be combined with a hydrogel to form a composite. The use of any hydrogel composition known to one skilled in the art is encompassed within the invention, e.g., any of the hydrogel compositions disclosed in the following reviews: Graham, 1998, Med. Device Technol. 9 (1): 18-22; Peppas et al., 2000, Eur. J. Pharm. Biopharm. 50 (1): 27-46; Nguyen et al., 2002, Biomaterials, 23 (22): 4307-14; Henincl et al., 2002, Adv. Drug Deliv. Rev 54 (1): 13-36; Skelhome et al., 2002, Med. Device. Technol. 13 (9): 19-23; Schmedlen et al., 2002, Biomaterials 23: 4325-32. In a specific embodiment, the hydrogel composition is applied on the umbilical cord biomaterial, that is, is disposed on the surface of the biomaterial. The hydrogel composition for example, may be sprayed onto the umbilical cord biomaterial or coated onto the surface of the biomaterial, or the biomaterial may, for example, be soaked, bathed or saturated with the hydrogel composition. In another specific embodiment, the hydrogel is sandwiched between two or more layers of umbilical cord biomaterial. In an even more specific embodiment, the hydrogel is sandwiched between two layers of umbilical cord biomaterial, wherein the edges of the two layers of biomaterial are sealed so as to substantially or completely contain the hydrogel.
  • The hydrogels useful in the methods and compositions of the invention can be made from any water-interactive, or water soluble polymer known in the art, including but not limited to, polyvinylalcohol (PVA), polyhydroxyehthyl methacrylate, polyethylene glycol, polyvinyl pyrrolidone, hyaluronic acid, alginate, collagen, gelatin, dextran or derivatives and analogs thereof.
  • In some embodiments, a composition comprises an umbilical cord biomaterial of the invention, one or more bioactive compounds and a hydrogel. In other embodiments, a composition comprises an umbilical cord biomaterial and a hydrogel composition that comprises one or more bioactive compounds. In yet another embodiment, a composition comprises an umbilical cord biomaterial comprising one or more bioactive compounds and a hydrogel composition comprising one or more bioactive compounds. The bioactive compounds can be, for example, one or more compounds as described in Section 5.1.2, below.
  • 5.1.2 Bioactive Compounds
  • The umbilical cord biomaterial of the invention can comprise (e.g., be impregnated with or coated with) one or more bioactive or medicinal compounds, such as small organic molecules (e.g., drugs), antibiotics, antiviral agents, antimicrobial agents, anti-inflammatory agents, antiproliferative agents, cytokines, enzyme or protein inhibitors, antihistamines, and the like. In various embodiments, the umbilical cord biomaterial may be coated or impregnated with antibiotics (such as Clindamycin, Minocycline, Doxycycline, Gentamycin), hormones, growth factors, anti-tumor agents, anti-fungal agents, anti-viral agents, pain medications (including XYLOCAINE®, Lidocaine, Procaine, Novocaine, etc.), antihistamines (e.g., diphenhydramine, BENADRYL®, etc.), anti-inflammatory agents, anti-infectives including but not limited to silver (such as silver salts, including but not limited to silver nitrate and silver sulfadiazine), elemental silver, antibiotics, bactericidal enzymes (such as lysozome), wound healing agents (such as cytokines including but not limited to PDGF (e.g., REGRANEX®), TGF; thymosin), hyaluronic acid as a wound healing agent, wound sealants (such as fibrin with or without thrombin), cellular attractant and scaffolding reagents (such as fibronectin), and the like, or combinations of any of the foregoing, or of the foregoing and other compounds not listed. Such impregnation or coating may be accomplished by any means known in the art, and a portion or the whole of the umbilical cord biomaterial may be so coated or impregnated.
  • The umbilical cord biomaterial, or composites comprising umbilical cord biomaterial, may comprise any of the compounds listed herein, without limitation, individually or in any combination. Any of the biologically active compounds listed herein may be formulated by known methods for immediate release or extended release. Additionally, the umbilical cord biomaterial may comprise two or more biologically active compounds in different manners; e.g., the biomaterial may be impregnated with one biologically active compound and coated with another. In another embodiment, the umbilical cord biomaterial comprises one biologically active compound formulated for extended release, and a second biologically active compound formulated for immediate release.
  • Wound healing requires adequate nutrition, particularly the presence of iron, zinc, vitamin C, arginine, and the like. Thus, the umbilical cord biomaterial may comprise, e.g., be impregnated or coated with, a physiologically-available form of one or more nutrients required for wound healing. Preferably, the nutrient is formulated for extended release.
  • The umbilical cord biomaterial, or composite comprising umbilical cord biomaterial, may comprise an antibiotic. In certain embodiments, the antibiotic is a macrolide (e.g., tobramycin (TOBI®)), a cephalosporin (e.g., cephalexin (KEFLEX®)), cephradine (VELOSEF®)), cefuroxime (CEFTIN®, cefprozil (CEFZIL®), cefaclor (CECLOR®), cefixime (SUPRAX® or cefadroxil (DURICEF®), a clarithromycin (e.g., clarithromycin (Biaxin)), an erythromycin (e.g., erythromycin (EMYCIN®)), a penicillin (e.g., penicillin V (V-CILLINK® or PEN VEEK®)) or a quinolone (e.g., ofloxacin (FLOXIN®), ciprofloxacin (CIPRO®) ornorfloxacin (NOROXIN®)), aminoglycoside antibiotics (e.g., apramycin, arbekacin, bambermycins, butirosin, dibekacin, neomycin, neomycin, undecylenate, netilmicin, paromomycin, ribostamycin, sisomicin, and spectinomycin), amphenicol antibiotics (e.g., azidamfenicol, chloramphenicol, florfenicol, and thiamphenicol), ansamycin antibiotics (e.g., rifamide and rifampin), carbacephems (e.g., loracarbef), carbapenems (e.g., biapenem and imipenem), cephalosporins (e.g., cefaclor, cefadroxil, cefamandole, cefatrizine, cefazedone, cefozopran, cefpimizole, cefpiramide, and cefpirome), cephamycins (e.g., cefbuperazone, cefinetazole, and cefminox), monobactams (e.g., aztreonam, carumonam, and tigemonam), oxacephems (e.g., flomoxef, and moxalactam), penicillins (e.g., amdinocillin, amdinocillin pivoxil, amoxicillin, bacampicillin, benzylpenicillinic acid, benzylpenicillin sodium, epicillin, fenbenicillin, floxacillin, penamccillin, penethamate hydriodide, penicillin o-benethamine, penicillin 0, penicillin V, penicillin V benzathine, penicillin V hydrabamine, penimepicycline, and phencihicillin potassium), lincosamides (e.g., clindamycin, and lincomycin), macrolides (e.g., azithromycin, carbomycin, clarithomycin, dirithromycin, erythromycin, and erythromycin acistrate), amphomycin, bacitracin, capreomycin, colistin, enduracidin, enviomycin, tetracyclines (e.g., apicycline, chlortetracycline, clomocycline, and demeclocycline), 2,4-diaminopyrimidines (e.g., brodimoprim), nitrofurans (e.g., furaltadone, and furazolium chloride), quinolones and analogs thereof (e.g., cinoxacin, ciprofloxacin, clinafloxacin, flumequine, and grepagloxacin), sulfonamides (e.g., acetyl sulfamethoxypyrazine, benzylsulfamide, noprylsulfamide, phthalylsulfacetamide, sulfachrysoidine, and sulfacytine), sulfones (e.g., diathymosulfone, glucosulfone sodium, and solasulfone), cycloserine, mupirocin and tuberin.
  • The umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial, may comprise, e.g., be coated or impregnated with, an antifungal agent. Suitable antifungal agents include but are not limited to amphotericin B, itraconazole, ketoconazole, fluconazole, intrathecal, flucytosine, miconazole, butoconazole, clotrimazole, nystatin, terconazole, tioconazole, ciclopirox, econazole, haloprogrin, naftifine, terbinafine, undecylenate, and griseofuldin.
  • The umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial, may comprise, e.g., be coated or impregnated with, an anti-inflammatory agent. Useful anti-inflammatory agents include, but are not limited to, non-steroidal anti-inflammatory drugs such as salicylic acid, acetylsalicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, acetaminophen, indomethacin, sulindac, etodolac, mefenamic acid, meclofenamate sodium, tolmetin, ketorolac, dichlofenac, ibuprofen, naproxen, naproxen sodium, fenoprofen, ketoprofen, flurbinprofen, oxaprozin, piroxicam, meloxicam, ampiroxicam, droxicam, pivoxicam, tenoxicam, nabumetome, phenylbutazone, oxyphenbutazone, antipyrine, aminopyrine, apazone and nimesulide; leukotriene antagonists including, but not limited to, zileuton, aurothioglucose, gold sodium thiomalate and auranofin; and other anti-inflammatory agents including, but not limited to, methotrexate, colchicine, allopurinol, probenecid, sulfinpyrazone and benzbromarone.
  • The umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial, may comprise, e.g., be coated or impregnated with, an antiviral agent. Useful antiviral agents include, but are not limited to, nucleoside analogs, such as zidovudine, acyclovir, gangcyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin, as well as foscarnet, amantadine, rimantadine, saquinavir, indinavir, ritonavir, and the alpha-interferons.
  • The umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial, comprises, e.g., may be coated or impregnated with, a cytokine receptor modulator. Examples of cytokine receptor modulators include, but are not limited to, soluble cytokine receptors (e.g., the extracellular domain of a TNF-α receptor or a fragment thereof, the extracellular domain of an IL-10 receptor or a fragment thereof, and the extracellular domain of an IL-6 receptor or a fragment thereof), cytokines or fragments thereof (e.g., interleukin (IL)-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-15, TNF-α, TNF-β, interferon (IFN)-α, IFN-β, IFN-γ, and GM-CSF), anti-cytokine receptor antibodies (e.g., anti-IFN receptor antibodies, anti-IL-2 receptor antibodies (e.g., Zenapax (Protein Design Labs)), anti-IL-4 receptor antibodies, anti-IL-6 receptor antibodies, anti-IL-10 receptor antibodies, and anti-IL-12 receptor antibodies), anti-cytokine antibodies (e.g., anti-IFN antibodies, anti-TNF-α antibodies, anti-IL-10 antibodies, anti-IL-6 antibodies, anti-IL-8 antibodies (e.g., ABX-IL-8 (Abgenix)), and anti-IL-12 antibodies). In a specific embodiment, a cytokine receptor modulator is IL-4, IL-10, or a fragment thereof. In another embodiment, a cytokine receptor modulator is an anti-IL-1 antibody, anti-IL-6 antibody, anti-IL-12 receptor antibody, or anti-TNF-α antibody. In another embodiment, a cytokine receptor modulator is the extracellular domain of a TNF-α receptor or a fragment thereof. In certain embodiments, a cytokine receptor modulator is not a TNF-α antagonist.
  • In a preferred embodiment, proteins, polypeptides or peptides (including antibodies) that are utilized as immunomodulatory agents are derived from the same species as the recipient of the proteins, polypeptides or peptides so as to reduce the likelihood of an immune response to those proteins, polypeptides or peptides. In another preferred embodiment, when the subject is a human, the proteins, polypeptides, or peptides that are utilized as immunomodulatory agents are human or humanized.
  • The umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial, may also comprise, e.g., be coated or impregnated with. a cytokine. Examples of cytokines include, but are not limited to, colony stimulating factor 1 (CSF-1), interleukin-2 (IL-2), interleukin-3 (IL-3), interleukin-4 (IL-4), interleukin-5 (IL-5), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-9 (IL-9), interleukin-10 (IL-10), interleukin-12 (IL-12), interleukin 15 (IL-15), interleukin 18 (IL-18), insulin-like growth factor 1 (IGF-1), platelet derived growth factor (PDGF), erythropoietin (Epo), epidermal growth factor (EGF), fibroblast growth factor (FGF) (basic or acidic), granulocyte macrophage stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), heparin binding epidermal growth factor (HEGF), macrophage colony stimulating factor (M-CSF), prolactin, and interferon (IFN), e.g., IFN-alpha, and IFN-gamma), transforming growth factor alpha (TGF-α), TGFβ1, TGFβ2, tumor necrosis factor alpha (TNF-α), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), etc.
  • The umbilical cord biomaterial may also comprise, e.g., be coated or impregnated with, a hormone. Examples of hormones include, but are not limited to, luteinizing hormone releasing hormone (LHRH), growth hormone (GH), growth hormone releasing hormone, ACTH, somatostatin, somatotropin, somatomedin, parathyroid hormone, hypothalamic releasing factors, insulin, glucagon, enkephalins, vasopressin, calcitonin, heparin, low molecular weight heparins, heparinoids, synthetic and natural opioids, insulin thyroid stimulating hormones, and endorphins. Examples of β-interferons include, but are not limited to, interferon β 1-a and interferon β 1-b.
  • The umbilical cord biomaterial, or composite comprising umbilical cord biomaterial, may also comprise, e.g., be coated or impregnated with, an alkylating agent. Examples of alkylating agents include, but are not limited to nitrogen mustards, ethylenimines, methylmelamines, alkyl sulfonates, nitrosoureas, triazenes, mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil, hexamethylmelaine, thiotepa, busulfan, carmustine, streptozocin, dacarbazine and temozolomide.
  • The umbilical cord biomaterial, or a composite comprising umbilical cord biomaterial, may also comprise, e.g., be coated or impregnated with, an immunomodulatory agent, including but not limited to methothrexate, leflunomide, cyclophosphamide, cyclosporine A, macrolide antibiotics (e.g., FK506 (tacrolimus)), methylprednisolone (MP), corticosteroids, steroids, mycophenolate mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar, malononitriloamindes (e.g., leflunamide), T cell receptor modulators, and cytokine receptor modulators, peptide mimetics, and antibodies (e.g., human, humanized, chimeric, monoclonal, polyclonal, Fvs, ScFvs, Fab or F(ab)2 fragments or epitope binding fragments), nucleic acid molecules (e.g., antisense nucleic acid molecules and triple helices), small molecules, organic compounds, and inorganic compounds. In particular, immunomodulatory agents include, but are not limited to, methothrexate, leflunomide, cyclophosphamide, cytoxan, Immuran, cyclosporine A, minocycline, azathioprine, antibiotics (e.g., FK506 (tacrolimus)), methylprednisolone (MP), corticosteroids, steroids, mycophenolate mofetil, rapamycin (sirolimus), mizoribine, deoxyspergualin, brequinar, malononitriloamindes (e.g., leflunamide), T cell receptor modulators, and cytokine receptor modulators. Examples of T cell receptor modulators include, but are not limited to, anti-T cell receptor antibodies (e.g., anti-CD4 antibodies (e.g., cM-T412 (Boehringer), IDEC-CE9.Is (IDEC and SKB), mAb 4162W94, Orthoclone and OKTcdr4a (Janssen-Cilag)), anti-CD3 antibodies (e.g., Nuvion (Product Design Labs), OKT3 (Johnson & Johnson), or Rituxan (IDEC)), anti-CD5 antibodies (e.g., an anti-CD5 ricin-linked immunoconjugate), anti-CD7 antibodies (e.g., CHH-380 (Novartis)), anti-CD8 antibodies, anti-CD40 ligand monoclonal antibodies (e.g., IDEC-131 (IDEC)), anti-CD52 antibodies (e.g., CAMPATH 1H (Ilex)), anti-CD2 antibodies, anti-CD1 1a antibodies (e.g., Xanelim (Genentech)), and anti-B7 antibodies (e.g., IDEC-114) (IDEC))) and CTLA4-immunoglobulin. In a specific embodiment, a T cell receptor modulator is a CD2 antagonist. In other embodiments, a T cell receptor modulator is not a CD2 antagonist. In another specific embodiment, a T cell receptor modulator is a CD2 binding molecule, preferably MEDI-507. In other embodiments, a T cell receptor modulator is not a CD2 binding molecule.
  • The umbilical cord biomaterial, or composite comprising umbilical cord biomaterial, may also comprise, e.g., be coated or impregnated with a class of immunomodulatory compounds known as IMIDS®. As used herein and unless otherwise indicated, the term “IMID®” and “IMIDS®” (Celgene Corporation) encompasses small organic molecules that markedly inhibit TNF-α, LPS induced monocyte IL-1β and IL-12, and partially inhibit IL-6 production. Specific immunomodulatory compounds are discussed below.
  • Specific examples of such immunomodulatory compounds, include, but are not limited to, cyano and carboxy derivatives of substituted styrenes such as those disclosed in U.S. Pat. No. 5,929,117; 1-oxo-2-(2,6-dioxo-3-fluoropiperidin-3-yl)isoindolines and 1,3-dioxo-2-(2,6-dioxo-3-fluoropiperidine-3-yl)isoindolines such as those described in U.S. Pat. Nos. 5,874,448 and 5,955,476; the tetra substituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolines described in U.S. Pat. No. 5,798,368; 1-oxo and 1,3-dioxo-2-(2,6-dioxopiperidin-3-yl)isoindolines (e.g., 4-methyl derivatives of thalidomide), including, but not limited to, those disclosed in U.S. Pat. Nos. 5,635,517, 6,476,052, 6,555,554, and 6,403,613; 1-oxo and 1,3-dioxoisoindolines substituted in the 4- or 5-position of the indoline ring (e.g., 4-(4-amino-1,3-dioxoisoindoline-2-yl)-4-carbamoylbutanoic acid) described in U.S. Pat. No. 6,380,239; isoindoline-1-one and isoindoline-1,3-dione substituted in the 2-position with 2,6-dioxo-3-hydroxypiperidin-5-yl (e.g., 2-(2,6-dioxo-3-hydroxy-5-fluoropiperidin-5-yl)-4-aminoisoindolin-1-one) described in U.S. Pat. No. 6,458,810; a class of non-polypeptide cyclic amides disclosed in U.S. Pat. Nos. 5,698,579 and 5,877,200; aminothalidomide, as well as analogs, hydrolysis products, metabolites, derivatives and precursors of aminothalidomide, and substituted 2-(2,6-dioxopiperidin-3-yl)phthalimides and substituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoles such as those described in U.S. Pat. Nos. 6,281,230 and 6,316,471; and isoindole-imide compounds such as those described in U.S. patent application Ser. No. 09/972,487 filed on Oct. 5, 2001, U.S. patent application Ser. No. 10/032,286 filed on Dec. 21, 2001, and International Application No. PCT/US01/50401 (International Publication No. WO 02/059106). The entireties of each of the patents and patent applications identified herein are incorporated herein by reference. Immunomodulatory compounds do not include thalidomide.
  • Other specific immunomodulatory compounds include, but are not limited to, 1-oxo- and 1,3dioxo-2-(2,6-dioxopiperidin-3-yl)isoindolines substituted with amino in the benzo ring as described in U.S. Pat. No. 5,635,517 which is incorporated herein by reference. These compounds have the structure I:
  • Figure US20120114712A1-20120510-C00001
  • in which one of X and Y is C═O, the other of X and Y is C═O or CH2, and R2 is hydrogen or lower alkyl, in particular methyl. Specific immunomodulatory compounds include, but are not limited to:
    • 1-oxo-2-(2,6-dioxopiperidin-3-yl)-4-aminoisoindoline;
    • 1-oxo-2-(2,6-dioxopiperidin-3-yl)-5-aminoisoindoline;
    • 1-oxo-2-(2,6-dioxopiperidin-3-yl)-6-aminoisoindoline;
    • 1-oxo-2-(2,6-dioxopiperidin-3-yl)-7-aminoisoindoline;
    • 1,3-dioxo-2-(2,6-dioxopiperidin-3-yl)-4-aminoisoindoline; and
    • 1,3-dioxo-2-(2,6-dioxopiperidin-3-yl)-5-aminoisoindoline.
  • Other specific immunomodulatory compounds belong to a class of substituted 2-(2,6-dioxopiperidin-3-yl)phthalimides and substituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindoles, such as those described in U.S. Pat. Nos. 6,281,230; 6,316,471; 6,335,349; and 6,476,052, and International Patent Application No. PCT/US97/13375 (International Publication No. WO 98/03502), each of which is incorporated herein by reference. Representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00002
  • in which:
  • one of X and Y is C═O and the other of X and Y is C═O or CH2;
  • (i) each of R1, R2, R3, and R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3, and R4 is —NHR5 and the remaining of R1, R2, R3, and R4 are hydrogen;
  • R5 is hydrogen or alkyl of 1 to 8 carbon atoms;
  • R6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzyl, or halo;
  • provided that R6 is other than hydrogen if X and Y are C═O and (i) each of R1, R2, R3, and R4 is fluoro or (ii) one of R1, R2, R3, or R4 is amino.
  • Compounds representative of this class are of the formulas:
  • Figure US20120114712A1-20120510-C00003
  • wherein R1 is hydrogen or methyl. In a separate embodiment, the invention encompasses the use of enantiomerically pure forms (e.g., optically pure (R) or (S) enantiomers) of these compounds.
  • Still other specific immunomodulatory compounds belong to a class of isoindole-imides disclosed in U.S. Patent Application Publication Nos. US 2003/0096841 and US 2003/0045552, and International Application No. PCT/US01/50401 (International Publication No. WO 02/059106), each of which are incorporated herein by reference. Representative compounds are of formula II:
  • Figure US20120114712A1-20120510-C00004
  • and pharmaceutically acceptable salts, hydrates, solvates, clathrates, enantiomers, diastereomers, racemates, and mixtures of stereoisomers thereof, wherein:
  • one of X and Y is C═O and the other is CH2 or C═O;
  • R1 is H, (C1-C8)alkyl, (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2-C5)heteroaryl, C(O)R3, C(S)R3, C(O)OR4, (C1-C8)alkyl-N(R6)2, (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, C(O)NHR3, C(S)NHR3, C(O)NR3R3′, C(S)NR3R3′ or (C1-C8)alkyl-O(CO)R5;
  • R2 is H, F, benzyl, (C1-C8)alkyl, (C2-C8)alkenyl, or (C2-C8)alkynyl;
  • R3 and R3′ are independently (C1-C8)alkyl, (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2-C5)heteroaryl, (C0-C8)alkyl-N(R6)2, (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, (C1-C8)alkyl-O(CO)R5, or C(O)OR5;
  • R4 is (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C1-C4)alkyl-OR5, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, or (C0-C4)alkyl-(C2-C5)heteroaryl;
  • R5 is (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, or (C2-C5)heteroaryl;
  • each occurrence of R6 is independently H, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, (C2-C5)heteroaryl, or (C0-C8)alkyl-C(O)O—R5 or the R6 groups can join to form a heterocycloalkyl group;
  • n is 0 or 1; and
  • * represents a chiral-carbon center.
  • In specific compounds of formula II, when n is 0 then R1 is (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2-C5)heteroaryl, C(O)R3, C(O)OR4, (C1-C8)alkyl-N(R6)2, (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, C(S)NHR3, or (C1-C8)alkyl-O(CO)R5;
  • R2 is H or (C1-C8)alkyl; and
  • R3 is (C1-C8)alkyl, (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2-C5)heteroaryl, (C5-C8)alkyl-N(R6)2; (C0-C8)alkyl-NH—C(O)O—R5; (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, (C1-C8)alkyl-O(CO)R5, or C(O)OR5; and the other variables have the same definitions.
  • In other specific compounds of formula II, R2 is H or (C1-C4)alkyl.
  • In other specific compounds of formula II, R1 is (C1-C8)alkyl or benzyl.
  • In other specific compounds of formula II, R1 is H, (C1-C8)alkyl, benzyl, CH2OCH3, CH2CH2OCH3, or
  • Figure US20120114712A1-20120510-C00005
  • In another embodiment of the compounds of formula II, R1 is
  • Figure US20120114712A1-20120510-C00006
  • wherein Q is O or S, and each occurrence of R7 is independently H, (C1-C8)alkyl, (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, halogen, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2-C5)heteroaryl, (C0-C8)alkyl-N(R6)2, (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, (C1-C8)alkyl-O(CO)R5, or C(O)OR5, or adjacent occurrences of R7 can be taken together to form a bicyclic alkyl or aryl ring.
  • In other specific compounds of formula II, R1 is C(O)R3.
  • In other specific compounds of formula II, R3 is (C0-C4)alkyl-(C2-C5)heteroaryl, (C1-C8)alkyl, aryl, or (C0-C4)alkyl-OR5.
  • In other specific compounds of formula II, heteroaryl is pyridyl, furyl, or thienyl.
  • In other specific compounds of formula II, R1 is C(O)OR4.
  • In other specific compounds of formula II, the H of C(O)NHC(O) can be replaced with (C1-C4)alkyl, aryl, or benzyl.
  • Further examples of the compounds in this class include, but are not limited to: [2-(2,6-dioxo-piperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-4-ylmethyl]-amide; (2-(2,6-dioxo-piperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-4-ylmethyl)-carbamic acid tert-butyl ester; 4-(aminomethyl)-2-(2,6-dioxo(3-piperidyl))-isoindoline-1,3-dione; N-(2-(2,6-dioxo-piperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-isoindol-4-ylmethyl)-acetamide; N-{(2-(2,6-dioxo(3-piperidyl)-1,3-dioxoisoindolin-4-yl)methyl}cyclopropyl-carboxamide; 2-chloro-N-{(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)methyl}acetamide; N-(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)-3-pyridylcarboxamide; 3-{1-oxo-4-(benzylamino)isoindolin-2-yl}piperidine-2,6-dione; 2-(2,6-dioxo(3-piperidyl))-4-(benzylamino)isoindoline-1,3-dione; N-{(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)methyl}propanamide; N-{(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)methyl}-3-pyridylcarboxamide; N-{(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)methyl}heptanamide; N-{(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)methyl}-2-furylcarboxamide; {N-(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)carbamoyl}methyl acetate; N-(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)pentanamide; N-(2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl)-2-thienylcarboxamide; N-{[2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl]methyl}(butylamino)carboxamide; N-{[2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl]methyl}(octylamino)carboxamide; and N-{[2-(2,6-dioxo(3-piperidyl))-1,3-dioxoisoindolin-4-yl]methyl}(benzylamino)carboxamide.
  • Still other specific immunomodulatory compounds belong to a class of isoindole-imides disclosed in U.S. Patent Application Publication Nos. US 2002/0045643, International Publication No. WO 98/54170, and U.S. Pat. No. 6,395,754, each of which is incorporated herein by reference. Representative compounds are of formula III:
  • Figure US20120114712A1-20120510-C00007
  • and pharmaceutically acceptable salts, hydrates, solvates, clathrates, enantiomers, diastereomers, racemates, and mixtures of stereoisomers thereof, wherein:
  • one of X and Y is C═O and the other is CH2 or C═O;
  • R is H or CH2OCOR′;
  • (i) each of R1, R2, R3, or R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3, or R4 is nitro or —NHR5 and the remaining of R1, R2, R3, or R4 are hydrogen;
  • R5 is hydrogen or alkyl of 1 to 8 carbons
  • R6 hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro;
  • R′ is R7—CHR10—N(R8R9);
  • R7 is m-phenylene or p-phenylene or —(CnH2n)— in which n has a value of 0 to 4;
  • each of R8 and R9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R8 and R9 taken together are tetramethylene, pentamethylene, hexamethylene, or —CH2CH2X1CH2CH2— in which X1 is —O—, —S—, or —NH—;
  • R10 is hydrogen, alkyl of to 8 carbon atoms, or phenyl; and
  • * represents a chiral-carbon center.
  • Other representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00008
  • wherein:
  • one of X and Y is C═O and the other of X and Y is C═O or CH2;
  • (i) each of R1, R2, R3, or R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3, and R4 is —NHR5 and the remaining of R1, R2, R3, and R4 are hydrogen;
  • R5 is hydrogen or alkyl of 1 to 8 carbon atoms;
  • R6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro;
  • R7 is m-phenylene or p-phenylene or —(CnH2n)— in which n has a value of 0 to 4;
  • each of R8 and R9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R8 and R9 taken together are tetramethylene, pentamethylene, hexamethylene, or —CH2CH2X1CH2CH2— in which X1 is —O—, —S—, or —NH—;
  • R10 is hydrogen, alkyl of to 8 carbon atoms, or phenyl.
  • Other representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00009
  • in which:
  • one of X and Y is C═O and the other of X and Y is C═O or CH2;
  • each of R1, R2, R3, and R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3, and R4 is nitro or protected amino and the remaining of R1, R2, R3, and R4 are hydrogen; and
  • R6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro.
  • Other representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00010
  • in which:
  • one of X and Y is C═O and the other of X and Y is C═O or CH2;
  • (i) each of R1, R2, R3, and R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3, and R4 is —NHR5 and the remaining of R1, R2, R3, and R4 are hydrogen;
  • R5 is hydrogen, alkyl of 1 to 8 carbon atoms, or CO—R7—CH(R10)NR8R9 in which each of R7, R8, R9, and R10 is as herein defined; and
  • R6 is alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro.
  • Specific examples of the compounds are of formula:
  • Figure US20120114712A1-20120510-C00011
  • in which:
  • one of X and Y is C═O and the other of X and Y is C═O or CH2;
  • R6 is hydrogen, alkyl of 1 to 8 carbon atoms, benzyl, chloro, or fluoro;
  • R7 is m-phenylene, p-phenylene or —(CnH2n)— in which n has a value of 0 to 4;
  • each of R8 and R9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R8 and R9 taken together are tetramethylene, pentamethylene, hexamethylene, or —CH2CH2X1CH2CH2— in which X1 is —O—, —S— or —NH—; and
  • R10 is hydrogen, alkyl of 1 to 8 carbon atoms, or phenyl.
  • Preferred immunomodulatory compounds are 4-(amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-1,3-dione and 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione. The compounds can be obtained via standard, synthetic methods (see e.g., U.S. Pat. No. 5,635,517, incorporated herein by reference). The compounds are available from Celgene Corporation, Warren, N.J. 4-(Amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-1,3-dione has the following chemical structure:
  • Figure US20120114712A1-20120510-C00012
  • The compound 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione has the following chemical structure:
  • Figure US20120114712A1-20120510-C00013
  • In another embodiment, specific immunomodulatory compounds encompass polymorphic forms of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione such as Form A, B, C, D, E, F, G and H, disclosed in U.S. provisional application no. 60/499,723 filed on Sep. 4, 2003, and U.S. non-provisional application Ser. No. 10/934,863, filed Sep. 3, 2004, both of which are incorporated herein by reference. For example, Form A of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is an unsolvated, crystalline material that can be obtained from non-aqueous solvent systems. Form A has an X-ray powder diffraction pattern comprising significant peaks at approximately 8, 14.5, 16, 17.5, 20.5, 24 and 26 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 270° C. Form A is weakly or not hygroscopic and appears to be the most thermodynamically stable anhydrous polymorph of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidine-2,6-dione discovered thus far.
  • Form B of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a hemihydrated, crystalline material that can be obtained from various solvent systems, including, but not limited to, hexane, toluene, and water. Form B has an X-ray powder diffraction pattern comprising significant peaks at approximately 16, 18, 22 and 27 degrees 2θ, and has endotherms from DSC curve of about 146 and 268° C., which are identified dehydration and melting by hot stage microscopy experiments. Interconversion studies show that Form B converts to Form E in aqueous solvent systems, and converts to other forms in acetone and other anhydrous systems.
  • Form C of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a hemisolvated crystalline material that can be obtained from solvents such as, but not limited to, acetone. Form C has an X-ray powder diffraction pattern comprising significant peaks at approximately 15.5 and 25 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 269° C. Form C is not hygroscopic below about 85% RH, but can convert to Form B at higher relative humidities.
  • Form D of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a crystalline, solvated polymorph prepared from a mixture of acetonitrile and water. Form D has an X-ray powder diffraction pattern comprising significant peaks at approximately 27 and 28 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 270° C. Form D is either weakly or not hygroscopic, but will typically convert to Form B when stressed at higher relative humidities.
  • Form E of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a dihydrated, crystalline material that can be obtained by slurrying 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione in water and by a slow evaporation of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione in a solvent system with a ratio of about 9:1 acetone:water. Form E has an X-ray powder diffraction pattern comprising significant peaks at approximately 20, 24.5 and 29 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 269° C. Form E can convert to Form C in an acetone solvent system and to Form G in a THF solvent system. In aqueous solvent systems, Form E appears to be the most stable form. Desolvation experiments performed on Form E show that upon heating at about 125° C. for about five minutes, Form E can convert to Form B. Upon heating at 175° C. for about five minutes, Form B can convert to Form F.
  • Form F of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is an unsolvated, crystalline material that can be obtained from the dehydration of Form E. Form F has an X-ray powder diffraction pattern comprising significant peaks at approximately 19, 19.5 and 25 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 269° C.
  • Form G of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is an unsolvated, crystalline material that can be obtained from slurrying forms B and E in a solvent such as, but not limited to, tetrahydrofuran (THF). Form G has an X-ray powder diffraction pattern comprising significant peaks at approximately 21, 23 and 24.5 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 267° C.
  • Form H of 3-(4-amino-1-oxo-1,3dihydro-isoindol-2-yl)-piperidene-2,6-dione is a partially hydrated (about 0.25 moles) crystalline material that can be obtained by exposing Form E to 0% relative humidity. Form H has an X-ray powder diffraction pattern comprising significant peaks at approximately 15, 26 and 31 degrees 2θ, and has a differential scanning calorimetry melting temperature maximum of about 269° C.
  • Other specific immunomodulatory compounds include, but are not limited to, 1-oxo-2-(2,6-dioxo-3-fluoropiperidin-3yl)isoindolines and 1,3-dioxo-2-(2,6-dioxo-3-fluoropiperidine-3-yl)isoindolines such as those described in U.S. Pat. Nos. 5,874,448 and 5,955,476, each of which is incorporated herein by reference. Representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00014
  • wherein Y is oxygen or H2 and
  • each of R1, R2, R3, and R4, independently of the others, is hydrogen, halo, alkyl of 1 to 4 carbon atoms; alkoxy of 1 to 4 carbon atoms, or amino.
  • Other specific immunomodulatory compounds include, but are not limited to, the tetra substituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolines described in U.S. Pat. No. 5,798,368, which is incorporated herein by reference. Representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00015
  • wherein each of R1, R2, R3, and R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms.
  • Other specific immunomodulatory compounds include, but are not limited to, 1-oxo and 1,3-dioxo-2-(2,6-dioxopiperidin-3-yl)isoindolines disclosed in U.S. Pat. No. 6,403,613, which is incorporated herein by reference. Representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00016
  • in which
  • Y is oxygen or H2,
  • a first of R1 and R2 is halo, alkyl, alkoxy, alkylamino, dialkylamino, cyano, or carbamoyl, the second of R1 and R2, independently of the first, is hydrogen, halo, alkyl, alkoxy, alkylamino, dialkylamino, cyano, or carbamoyl, and
  • R3 is hydrogen, alkyl, or benzyl.
  • Specific examples of the compounds are of formula:
  • Figure US20120114712A1-20120510-C00017
  • wherein a first of R1 and R2 is halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl,
  • the second of R1 and R2, independently of the first, is hydrogen, halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, alkylamino in which alkyl is of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl, and
  • R3 is hydrogen, alkyl of from 1 to 4 carbon atoms, or benzyl. Specific examples include, but are not limited to, 1-oxo-2-(2,6-dioxopiperidin-3-yl)-4-methylisoindoline.
  • Other representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00018
  • wherein a first of R1 and R2 is halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl,
  • the second of R1 and R2, independently of the first, is hydrogen, halo, alkyl of from 1 to 4 carbon atoms, alkoxy of from 1 to 4 carbon atoms, alkylamino in which alkyl is of from 1 to 4 carbon atoms, dialkylamino in which each alkyl is of from 1 to 4 carbon atoms, cyano, or carbamoyl, and
  • R3 is hydrogen, alkyl of from 1 to 4 carbon atoms, or benzyl.
  • Specific examples include, but are not limited to, 1-oxo-2-(2,6-dioxopiperidin-3-yl)-4-methylisoindoline.
  • Other specific immunomodulatory compounds include, but are not limited to, 1-oxo and 1,3-dioxoisoindolines substituted in the 4- or 5-position of the indoline ring described in U.S. Pat. No. 6,380,239 and co-pending U.S. application Ser. No. 10/900,270, filed Jul. 28, 2004, which are incorporated herein by reference. Representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00019
  • in which the carbon atom designated C* constitutes a center of chirality (when n is not zero and R1 is not the same as R2); one of X1 and X2 is amino, nitro, alkyl of one to six carbons, or NH—Z, and the other of X1 or X2 is hydrogen; each of R1 and R2 independent of the other, is hydroxy or NH—Z; R3 is hydrogen, alkyl of one to six carbons, halo, or haloalkyl; Z is hydrogen, aryl, alkyl of one to six carbons, formyl, or acyl of one to six carbons; and n has a value of 0, 1, or 2; provided that if X1 is amino, and n is 1 or 2, then R1 and R2 are not both hydroxy; and the salts thereof.
  • Further representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00020
  • in which the carbon atom designated C* constitutes a center of chirality when n is not zero and R1 is not R2; one of X1 and X2 is amino, nitro, alkyl of one to six carbons, or NH—Z, and the other of X1 or X2 is hydrogen; each of R1 and R2 independent of the other, is hydroxy or NH—Z; R3 is alkyl of one to six carbons, halo, or hydrogen; Z is hydrogen, aryl or an alkyl or acyl of one to six carbons; and n has a value of 0, 1, or 2.
  • Specific examples include, but are not limited to, 2-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-4-carbamoyl-butyric acid and 4-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-4-cabamoyl-butyric acid, which have the following structures, respectively, and pharmaceutically acceptable salts, solvates, prodrugs, and stereoisomers thereof:
  • Figure US20120114712A1-20120510-C00021
  • Other representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00022
  • in which the carbon atom designated C* constitutes a center of chirality when n is not zero and R1 is not R2; one of X1 and X2 is amino, nitro, alkyl of one to six carbons, or NH—Z, and the other of X1 or X2 is hydrogen; each of R1 and R2 independent of the other, is hydroxy or NH—Z; R3 is alkyl of one to six carbons, halo, or hydrogen; Z is hydrogen, aryl, or an alkyl or acyl of one to six carbons; and n has a value of 0, 1, or 2; and the salts thereof.
  • Specific examples include, but are not limited to, 4-carbamoyl-4-{4-[(furan-2-yl-methyl)-amino]-1,3-dioxo-1,3-dihydro-isoindol-2-yl}-butyric acid, 4-carbamoyl-2-{4-[(furan-2-yl-methyl)-amino]-1,3-dioxo-1,3-dihydro-isoindol-2-yl}-butyric acid, 2-{4-[(furan-2-yl-methyl)-amino]-1,3-dioxo-1,3-dihydro-isoindol-2-yl}-4-phenylcarbamoyl-butyric acid, and 2-{4-[(furan-2-yl-methyl)-amino]-1,3-dioxo-1,3-dihydro-isoindol-2-yl}-pentanedioic acid, which have the following structures, respectively, and pharmaceutically acceptable salts, solvate, prodrugs, and stereoisomers thereof:
  • Figure US20120114712A1-20120510-C00023
  • Other specific examples of the compounds are of formula:
  • Figure US20120114712A1-20120510-C00024
  • wherein one of X1 and X2 is nitro, or NH—Z, and the other of X1 or X2 is hydrogen;
  • each of R1 and R2, independent of the other, is hydroxy or NH—Z;
  • R3 is alkyl of one to six carbons, halo, or hydrogen;
  • Z is hydrogen, phenyl, an acyl of one to six carbons, or an alkyl of one to six carbons; and
  • n has a value of 0, 1, or 2;
  • provided that if one of X1 and X2 is nitro, and n is 1 or 2, then R1 and R2 are other than hydroxy; and if —COR2 and —(CH2)nCOR1 are different, the carbon atom designated C* constitutes a center of chirality. Other representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00025
  • wherein one of X1 and X2 is alkyl of one to six carbons;
  • each of R1 and R2, independent of the other, is hydroxy or NH—Z;
  • R3 is alkyl of one to six carbons, halo, or hydrogen;
  • Z is hydrogen, phenyl, an acyl of one to six carbons, or an alkyl of one to six carbons;
  • n has a value of 0, 1, or 2; and
  • if —COR2 and —(CH2)nCOR1 are different, the carbon atom designated C* constitutes a center of chirality.
  • Still other specific immunomodulatory compounds include, but are not limited to, isoindoline-1-one and isoindoline-1,3-dione substituted in the 2-position with 2,6-dioxo-3-hydroxypiperidin-5-yl described in U.S. Pat. No. 6,458,810, which is incorporated herein by reference. Representative compounds are of formula:
  • Figure US20120114712A1-20120510-C00026
  • wherein:
  • the carbon atoms designated * constitute centers of chirality;
  • X is —C(O)— or —CH2—;
  • R1 is alkyl of 1 to 8 carbon atoms or —NHR3;
  • R2 is hydrogen, alkyl of 1 to 8 carbon atoms, or halogen; and
  • R3 is hydrogen,
  • alkyl of 1 to 8 carbon atoms, unsubstituted or substituted with alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms,
  • cycloalkyl of 3 to 18 carbon atoms,
  • phenyl, unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms,
  • benzyl, unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms, or —COR4 in which
  • R4 is hydrogen,
  • alkyl of 1 to 8 carbon atoms, unsubstituted or substituted with alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms,
  • cycloalkyl of 3 to 18 carbon atoms,
  • phenyl, unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms, or
  • benzyl, unsubstituted or substituted with alkyl of 1 to 8 carbon atoms, alkoxy of 1 to 8 carbon atoms, halo, amino, or alkylamino of 1 to 4 carbon atoms.
  • The immunomodulatory compounds disclosed herein can either be commercially purchased or prepared according to the methods described in the patents or patent publications disclosed herein. Further, optically pure compounds can be asymmetrically synthesized or resolved using known resolving agents or chiral columns as well as other standard synthetic organic chemistry techniques.
  • As used herein and unless otherwise indicated, the term “pharmaceutically acceptable salt” encompasses non-toxic acid and base addition salts of the compound to which the term refers. Acceptable non-toxic acid addition salts include those derived from organic and inorganic acids or bases know in the art, which include, for example, hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulphonic acid, acetic acid, tartaric acid, lactic acid, succinic acid, citric acid, malic acid, maleic acid, sorbic acid, aconitic acid, salicylic acid, phthalic acid, embolic acid, enanthic acid, and the like.
  • Compounds that are acidic in nature are capable of forming salts with various pharmaceutically acceptable bases. The bases that can be used to prepare pharmaceutically acceptable base addition salts of such acidic compounds are those that form non-toxic base addition salts, i.e., salts containing pharmacologically acceptable cations such as, but not limited to, alkali metal or alkaline earth metal salts and the calcium, magnesium, sodium or potassium salts in particular. Suitable organic bases include, but are not limited to, N,N-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumaine (N-methylglucamine), lysine, and procaine.
  • As used herein, and unless otherwise specified, the term “solvate” means a compound of the present invention or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • As used herein and unless otherwise indicated, the term “prodrug” means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide the compound. Examples of prodrugs include, but are not limited to, derivatives of immunomodulatory compounds of the invention that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. Other examples of prodrugs include derivatives of immunomodulatory compounds of the invention that comprise —NO, —NO2, —ONO, or —ONO2 moieties. Prodrugs can typically be prepared using well-known methods, such as those described in 1 Burger's Medicinal Chemistry and Drug Discovery, 172-178, 949-982 (Manfred E. Wolff ed., 5th ed. 1995), and Design of Prodrugs (H. Bundgaard ed., Elselvier, New York 1985).
  • As used herein and unless otherwise indicated, the terms “biohydrolyzable amide,” “biohydrolyzable ester,” “biohydrolyzable carbamate,” “biohydrolyzable carbonate,” “biohydrolyzable ureide,” “biohydrolyzable phosphate” mean an amide, ester, carbamate, carbonate, ureide, or phosphate, respectively, of a compound that either: 1) does not interfere with the biological activity of the compound but can confer upon that compound advantageous properties in vivo, such as uptake, duration of action, or onset of action; or 2) is biologically inactive but is converted in vivo to the biologically active compound. Examples of biohydrolyzable esters include, but are not limited to, lower alkyl esters, lower acyloxyalkyl esters (such as acetoxylmethyl, acetoxyethyl, aminocarbonyloxymethyl, pivaloyloxymethyl, and pivaloyloxyethyl esters), lactonyl esters (such as phthalidyl and thiophthalidyl esters), lower alkoxyacyloxyalkyl esters (such as methoxycarbonyl-oxymethyl, ethoxycarbonyloxyethyl and isopropoxycarbonyloxyethyl esters), alkoxyalkyl esters, choline esters, and acylamino alkyl esters (such as acetamidomethyl esters). Examples of biohydrolyzable amides include, but are not limited to, lower alkyl amides, α-amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides. Examples of biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, amino acids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.
  • As used herein, and unless otherwise specified, the term “stereoisomer” encompasses all enantiomerically/stereomerically pure and enantiomerically/stereomerically enriched compounds of this invention.
  • As used herein, and unless otherwise indicated, the term “stereomerically pure” or “enantiomerically pure” means that a compound comprises one stereoisomer and is substantially free of its counter stereoisomer or enantiomer. For example, a compound is stereomerically or enantiomerically pure when the compound contains 80%, 90%, or 95% or more of one stereoisomer and 20%, 10%, or 5% or less of the counter stereoisomer. In certain cases, a compound of the invention is considered optically active or stereomerically/enantiomerically pure (i.e., substantially the R-form or substantially the S-form) with respect to a chiral center when the compound is about 80% ee (enantiomeric excess) or greater, preferably, equal to or greater than 90% ee with respect to a particular chiral center, and more preferably 95% ee with respect to a particular chiral center.
  • As used herein, and unless otherwise indicated, the term “stereomerically enriched” or “enantiomerically enriched” encompasses racemic mixtures as well as other mixtures of stereoisomers of compounds of this invention (e.g., R/S=30/70, 35/65, 40/60, 45/55, 55/45, 60/40, 65/35 and 70/30). Various immunomodulatory compounds of the invention contain one or more chiral centers, and can exist as racemic mixtures of enantiomers or mixtures of diastereomers. This invention encompasses the use of stereomerically pure forms of such compounds, as well as the use of mixtures of those forms. For example, mixtures comprising equal or unequal amounts of the enantiomers of a particular immunomodulatory compounds of the invention may be used in methods and compositions of the invention. These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral columns or chiral resolving agents. See, e.g., Jacques, J., et al., Enantiomers, Racemates and Resolutions (Wiley-Interscience, New York, 1981); Wilen, S. H., et al., Tetrahedron 33:2725 (1977); Eliel, E. L., Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, S. H., Tables of Resolving Agents and Optical Resolutions p. 268 (E. L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, Ind., 1972).
  • It should be noted that if there is a discrepancy between a depicted structure and a name given that structure, the depicted structure is to be accorded more weight. In addition, if the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold or dashed lines, the structure or portion of the structure is to be interpreted as encompassing all stereoisomers of it.
  • The amount of the bioactive compound coating or impregnating the umbilical cord biomaterial may vary, and will preferably depend upon the particular bioactive compound to be delivered, and the effect desired. For example, where the bioactive compound is an anti-inflammatory agent, the amount of the anti-inflammatory agent on or contained by the umbilical cord biomaterial is an amount sufficient to measurably reduce one or more symptoms or indicia of inflammation in a tissue contacted by, or proximal to, e.g., an umbilical cord biomaterial implant.
  • In various embodiments, the umbilical cord biomaterial of the invention may comprise, e.g., be coated or impregnated with, at least 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 100, 1250, 1500, 2000, 2500, 300, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000 or at least 1000000 nanograms of a bioactive compound. In another embodiment, the umbilical cord biomaterial of the invention may be coated with, or impregnated with, no more than 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 100, 1250, 1500, 2000, 2500, 300, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000 or at least 1000000 nanograms of a bioactive compound.
  • 5.1.3 Conformation of the Umbilical Cord Biomaterial
  • The umbilical cord biomaterial may be formed into any shape or conformation that will facilitate its use in the methods of the invention. For example, the umbilical cord biomaterial can be formed into any shape or conformation that will facilitate, e.g., the occlusion of a tympanic membrane perforation, particularly in the context of a tympanoplasty or myringoplasty; repair of a joint, ligament or tendon; etc. The umbilical cord biomaterial may, for example, be provided as an extended membrane, e.g., the entire membrane from a single umbilical cord. The umbilical cord biomaterial can also be provided as square, rectangular, circular or oval shaped pieces, or may be cut to conform generally to the shape of, e.g., a tympanic membrane, tendon, or other bodily structure. The umbilical cord biomaterial can be tubular. In various embodiments, umbilical cord biomaterial may be provided as pieces measuring approximately 1×1 cm, 1.5×1.5 cm, 2×2 cm, 2.5×2.5 cm, 1×1.5 cm, 1×2 cm, 1×2.5 cm, 1×3 cm, 1×3.5 cm, 1×4 cm, 1×4.5 cm, 1×5 cm, 1×5.5 cm, 1×6 cm, 1×6.5 cm, 1×7 cm, 1.5×2 cm, 1.5×2.5 cm, 1.5×3 cm, 1.5×3.5 cm, 1.5×4 cm, 1.5×4.5 cm, 1.5×5 cm, 1.5×5.5 cm, 1.5×6 cm, 1.5×6.5 cm, 1.5×7 cm, 2×2.5 cm, 2×3 cm, 2×3.5 cm, 2×4 cm, 2×4.5 cm, 2×5 cm, 2×5.5 cm, 2×6 cm, 2×6.5 cm, 2×7 cm, 2.5×2.5 cm, 2.5×3 cm, 2.5×3.5 cm, 2.5×4 cm, 2.5×4.5 cm, 2.5×5 cm, 2.5×5.5 cm, 2.5×6 cm, 2.5×6.5 cm, 2.5×7 cm, 3×3 cm, 3×3.5 cm, 3×4 cm, 3×4.5 cm, 3×5 cm, 3×5.5 cm, 3×6 cm, 3×6.5 cm, 3×7 cm, 3.5×3.5 cm, 3.5×4 cm, 3.5×4.5 cm, 3.5×5 cm, 3.5×5.5 cm, 3.5×6 cm, 3.5×6.5 cm, 3.5×7 cm, 4×2.5 cm, 4×4 cm, 4×4.5 cm, 4×5 cm, 4×5.5 cm, 4×6 cm, 4×6.5 cm, or 4×7 cm in size, or may be no smaller, or no larger, than 1×1 cm, 1.5×1.5 cm, 2×2 cm, 2.5×2.5 cm, 1×1.5 cm, 1×2 cm, 1×2.5 cm, 1×3 cm, 1×3.5 cm, 1×4 cm, 1×4.5 cm, 1×5 cm, 1×5.5 cm, 1×6 cm, 1×6.5 cm, 1×7 cm, 1.5×2 cm, 1.5×2.5 cm, 1.5×3 cm, 1.5×3.5 cm, 1.5×4 cm, 1.5×4.5 cm, 1.5×5 cm, 1.5×5.5 cm, 1.5×6 cm, 1.5×6.5 cm, 1.5×7 cm, 2×2.5 cm, 2×3 cm, 2×3.5 cm, 2×4 cm, 2×4.5 cm, 2×5 cm, 2×5.5 cm; 2×6 cm, 2×6.5 cm, 2×7 cm, 2.5×2.5 cm, 2.5×3 cm, 2.5×3.5 cm, 2.5×4 cm, 2.5×4.5 cm, 2.5×5 cm, 2.5×5.5 cm, 2.5×6 cm, 2.5×6.5 cm, 2.5×7 cm, 3×3 cm, 3×3.5 cm, 3×4 cm, 3×4.5 cm, 3×5 cm, 3×5.5 cm, 3×6 cm, 3×6.5 cm, 3×7 cm, 3.5×3.5 cm, 3.5×4 cm, 3.5×4.5 cm, 3.5×5 cm, 3.5×5.5 cm, 3.5×6 cm, 3.5×6.5 cm, 3.5×7 cm, 4×2.5 cm, 4×4 cm, 4×4.5 cm, 4×5 cm, 4×5.5 cm, 4×6 cm, 4×6.5 cm, or 4×7 cm, though the biomaterial may be cut to different dimensions. In preferred embodiments, wherein the umbilical cord biomaterial is used as a wound covering, the biomaterial is about 2.5 cm×2.5 cm to 3.5 cm×3.5 cm, or, more preferably, about 3×3 cm2. Longer and/or wider pieces can be formed by laminating two or more smaller pieces, as described elsewhere herein. Further, the biomaterial may be provided as a sheet from which an end user may cut two or more pieces, or may be provided as a roll or strip.
  • The biomaterial may be provided to the end user either dry, or pre-wetted in a suitable physiologically-compatible, medically-useful liquid, such as a saline solution. In one embodiment, the solution comprises one or more bioactive compounds, as described in Section 5.1.2, above. Preferably, said bioactive compound is disposed onto or within the umbilical cord biomaterial such that the majority of the bioactive compound contacts the tympanic membrane at some point during the time the umbilical cord biomaterial contacts the tympanic membrane.
  • 5.1.4 Methods of Making Umbilical Cord Biomaterial
  • The umbilical cord biomaterial of the invention can be made in a number of ways. For example, the biomaterial is preferably produced by any means that preserves the biochemical and structural characteristics of the membrane's components—chiefly collagen, elastin, laminin, and fibronectin. That is, the biomaterial can be made so as to preserve, or substantially preserve, the native structure of the protein components of the biomaterial. The biomaterial may also be altered, e.g., the proteins of the biomaterial can be crosslinked, so as to improve the strength (e.g., tensile strength) of the biomaterial. The biomaterial can be completely, or substantially completely, decellularized prior to use, that is, can comprise only, or substantially only, an umbilical cord outer membrane, or can be made to retain other components of the umbilical cord (e.g., Wharton's jelly, umbilical vessel(s), umbilical cord cells, and the like). Generally, an umbilical cord is separated from a placenta obtained by normal birth. The umbilical cord is then cleaned and disinfected, and optionally stored for further processing, e.g., decellularization and/or drying.
  • In one embodiment, the umbilical cord is separated from the placenta as soon as possible after delivery of the newborn. The umbilical cord may be used immediately, or may be stored for 2-5 days from the time of delivery prior to any further treatment. Preferably, the expectant mother is screened prior to the time of birth, using standard techniques known to one skilled in the art, for communicable diseases including but not limited to, HIV, HBV, HCV, HTLV, syphilis, CMV, and other viral pathogens known to contaminate umbilical cord tissue. One exemplary method for preparing the umbilical cord biomaterial of the invention comprises the following steps:
  • The umbilical cord is separated from the placental disc, and is typically massaged to remove umbilical cord blood. Optionally, the umbilical cord is sectioned into pieces of about 10 cm to about 15 cm in length. The umbilical cord or umbilical cord sections can then be stored for up to about 72 hours in a sterile, preferably buffered, saline solution, such as 0.9% sterile NaCl solution. Preferably, the umbilical cord is stored under refrigeration, at a temperature of about 1° C. to about 5° C.
  • At this time, the umbilical cord can be slit or cut longitudinally using, e.g., a scalpel and forceps, grooved director, or the like. This allows the umbilical cord membrane to be laid flat, allowing, e.g., removal of the Wharton's jelly, and/or one or more of the umbilical cord arteries, e.g., with a forceps. The umbilical cord membrane can also be processed further without cutting and opening the membrane. An umbilical cord vessel, for example, can be removed from the cord by grasping the vessels with a forceps and gently pulling and massaging until the vessel is removed, leaving the umbilical cord membrane as an intact tube. In a preferred embodiment of deveining, the umbilical vein of a fresh (less than 48 hours after delivery) umbilical cord is canalized using the blunt probe of a vein stripper. The blunt probe is replaced with a small bullet probe, and the vein is tied to the probe with thread. The stripper is then removed, and the process is repeated with the umbilical arteries.
  • The umbilical cord can be further processed “as is”, wherein the cord comprises the umbilical cord membrane, vessels, and Wharton's jelly.
  • Continuing the embodiment, the umbilical cord biomaterial can be substantially decellularized; that is, substantially all cellular material and cellular debris (e.g., all visible cellular material and cellular debris) can removed from the biomaterial. Any decellularizing process known to one skilled in the art may be used, however, generally the process used for decellularizing the umbilical cord biomaterial of the invention does not disrupt the native conformation of the proteins making up the biomaterial. “Substantially decellularized,” as used herein, means removal of at least 90% of the cells, more preferably at least 95% of the cells, and most preferably at least 99% of the cells associated with the umbilical cord membrane. Decellularization can leave cellular material on the umbilical cord biomaterial; for example, decellularization can leave nuclear material detectable by 4′,6-diamidino-2-phenylindole (DAPI) and still be considered decellularized.
  • Decellularization can comprise physical scraping, for example, with a sterile cell scraper, in combination with rinsing with a sterile solution. The decellularization technique employed preferably does not result in gross disruption of the anatomy of the umbilical cord membrane or alter the biomechanical properties of the umbilical cord membrane.
  • The decellularization of the umbilical cord biomaterial can comprise contacting the membrane with a detergent-containing solution, such as one or more mild anionic or nonionic detergents, e.g., Triton X-100, sodium dodecyl sulfate, or the like, in an amount and for a time sufficient to decellularize the biomaterial. Any mild detergent, i.e., a non-caustic, low-foaming detergent, with a pH of about 6 to about 8, can be used to decellularize the umbilical cord biomaterial. In a specific embodiment, the biomaterial is contacted with about 0.01-1% deoxycholic acid (e.g., deoxycholic acid sodium salt monohydrate) for about 30 minutes to about 480 hours, preferably about 1 hour to about 240 hours, to decellularize the umbilical cord biomaterial. In a preferred embodiment, the umbilical cord biomaterial is decellularized in about 1% for about 20 days without scraping, followed by heat drying.
  • The biomaterial can be decellularized by any other method known to those in the art, including freezing to form intracellular ice (including, e.g., vapor phase freezing). Where freezing is used to decellularize the biomaterial, preferably a cryoprotectant is used, e.g., polyvinylpyrollidone at, e.g., 10% w/v, or dialyzed hydroxyethyl starch at, e.g., 10% w/v, added to standard cryopreservation solutions such as, in a non-limiting example, DMEM comprising 10% DMSO and 10% fetal bovine serum.
  • Preferably, any native or exogenous protease activity is inhibited or prevented in the preparation of the biomaterial. Additives to the decellularization, rinse and/or storage solutions such as metal ion chelators, for example 1,10-phenanthroline and ethylenediaminetetraacetic acid (EDTA), create an environment unfavorable to many proteolytic enzymes. Providing sub-optimal conditions for proteases (e.g., collagenase) assists in protecting umbilical cord biomaterial components such as collagen from degradation during the cell lysis step. Suboptimal conditions for proteases may be achieved by formulating the decellularization solution to eliminate or limit the amount of available calcium and zinc ions. Many proteases are active in the presence of calcium and zinc ions and lose much of their activity in calcium and zinc ion free environments. Preferably, the decellularization solution is prepared, in part, by selecting conditions of pH, reduced availability of calcium and zinc ions, presence of metal ion chelators and the use of proteolytic inhibitors specific for collagenase, such that the solution will optimally lyse the native umbilical cord cells while protecting the umbilical cord biomaterial from proteolytic degradation. For example, a decellularization solution can include a buffered solution of water, pH 5.5 to 8, preferably pH 7 to 8, free from calcium and zinc ions and including a metal ion chelator such as EDTA. Decellularization can take place at, e.g., between 0° C. and 25° C., preferably below about 10° C., to reduce protease activity.
  • It is preferred that the decellularization treatment also limits the generation of new immunological sites. Since enzymatic degradation of, e.g., collagen is believed to lead to heightened immunogenicity, the invention encompasses treatment of the umbilical cord biomaterial with enzymes, e.g., nucleases, that are effective in inhibiting cellular metabolism, protein production and cell division, that minimize proteolysis of the components of the umbilical cord biomaterial thus preserving the underlying architecture of the amniotic biomaterial. Examples of nucleases that can be used in accordance with the methods of the invention are those effective in digestion of native cell DNA and RNA including both exonucleases and endonucleases. A non-limiting example of nucleases that can be used in accordance with the methods of the invention include exonucleases that inhibit cellular activity, e.g., DNase I (SIGMA Chemical Company, St. Louis, Mo.) and RNase A (SIGMA Chemical Company, St. Louis, Mo.) and endonucleases that inhibit cellular activity, e.g., EcoRI (SIGMA Chemical Company, St. Louis, Mo.) and HindIII (SIGMA Chemical Company, St. Louis, Mo.). It is preferable that the selected nucleases are applied in a physiological buffer solution which contains ions, e.g., magnesium, calcium, which are optimal for the activity of the nuclease. Preferably, the ionic concentration of the buffered solution, the treatment temperature and the length of treatment are selected by one skilled in the art by routine experimentation to assure the desired level of nuclease activity. The buffer is preferably hypotonic to promote access of the nucleases to cell interiors.
  • In another embodiment of the invention, the umbilical cord biomaterial is not decellularized prior to drying.
  • In another embodiment of the above steps, the umbilical cord, after initial processing, is briefly rinsed in saline to remove blood from the umbilical cord surface. The umbilical cord is then immersed in a cold deoxycholic acid solution at a concentration of about 0.1% to about 10%, and, in a specific embodiment, about 0.1% to about 2.0%. The umbilical cord is then incubated in this solution at between about 1° C. to about 8° C. for about 5 days to about 6 months. In specific embodiments, the umbilical cord is immersed, for example, for about 5 to about 15 days; about 5 to about 30 days, about 5 to about 60 days, or for up to about one year. Typically, the deoxycholic acid solution is replaced during incubation every 2-5 days. In another specific embodiment, the umbilical cord is immersed in a deoxycholic acid solution at a concentration of about 1% at a temperature of 0° C. to about 8° C. for about 5 days to about 15 days. This incubation serves two purposes. First, it allows time for serological tests to be performed on the umbilical cord and/or umbilical cord blood, so that umbilical cords failing to meet serological criteria are not processed further. Second, the longer incubation improves the removal of epithelial cells and fibroblasts, which allows for a significant reduction in the amount of time spent decellularizing the umbilical cord membrane by physically scraping. The umbilical cord biomaterial can then be dried as described below.
  • Following decellularization, the umbilical cord biomaterial is generally washed to assure removal of cellular debris (e.g., cellular proteins, cellular lipids, cellular nucleic acids, extracellular debris such as extracellular soluble proteins, lipids and proteoglycans, and the like). The wash solution can be de-ionized water or an aqueous hypotonic buffer. Preferably, the umbilical cord biomaterial is gently agitated, e.g., for 15-120 minutes in the detergent, e.g., on a rocking platform, to assist in the decellularization. The umbilical cord biomaterial, after detergent decellularization, can again be physically decellularized as described above; the physical and detergent decellularization steps may be repeated as necessary, as long as the integrity of the umbilical cord biomaterial is maintained, until no visible'cellular material and cellular debris remain.
  • In certain embodiments, the umbilical cord biomaterial is dried immediately (i.e., within 30 minutes) after decellularization and/or washing. Alternatively, when further processing is not done immediately, the umbilical cord biomaterial may be refrigerated, e.g., stored at a temperature of about 1° C. to about 20° C., preferably from about 2° C. to about 8° C., for up to 28 days prior to drying. When the umbilical cord biomaterial, e.g., decellularized umbilical cord biomaterial, is stored for more than three days, the sterile solution covering the umbilical cord biomaterial is preferably changed periodically, e.g., every 1-3 days.
  • In certain embodiments, when the umbilical cord biomaterial is not refrigerated after washing, the biomaterial can be washed, e.g., washed at least 3 times, prior to proceeding to the next step of the preparation. In other embodiments, when the umbilical cord biomaterial has been refrigerated and the sterile solution has been changed once, the umbilical cord biomaterial can be washed at least twice prior to the next step of the preparation. In yet other embodiments, when the umbilical cord biomaterial has been refrigerated and the sterile solution has been changed twice or more, the umbilical cord biomaterial can be washed at least once prior to proceeding to the next step.
  • The final step in this embodiment comprises drying the decellularized umbilical cord membrane to produce the umbilical cord biomaterial of the invention. Any method of drying the umbilical cord membrane can be used. For example, the membrane can be dried using heat, one or more hygroscopic compounds, freeze-drying, vacuum, microwaving, simple evaporation, and the like, or combinations of these methods. Preferably, the biomaterial is dried under vacuum.
  • The umbilical cord biomaterial can be dried in any useful conformation. Preferably, the umbilical cord biomaterial is dried so as to produce a flat, dry sheet. The biomaterial can also be dried as a tube, strip, spiral, string or rope, or the like. For three-dimensional shapes, the biomaterial can be placed onto, or into, a form and dried, so that the dried biomaterial assumes the shape of the form or a part thereof. In a specific embodiment, for example, the umbilical cord membrane can be supported by rubber hose or tubing inserted from one end, and freeze-dried to form a dried tube of the umbilical cord biomaterial. At this point, the umbilical cord biomaterial can be, e.g., part of a complete umbilical cord that has been washed and rinsed; an umbilical cord biomaterial comprising Wharton's jelly but lacking vessels, an umbilical cord biomaterial that has had the interior components of the umbilical cord removed, and has been decellularized, etc. In each case, the biomaterial can be dried.
  • In a specific embodiment, an exemplary method for drying the umbilical cord biomaterial comprises the following steps:
  • Assembly of the umbilical cord biomaterial for drying. The umbilical cord biomaterial is removed from the sterile solution, and the excess fluid is gently squeezed out. The umbilical cord biomaterial is then gently stretched until it is flat with the epithelial side facing in a downward position, e.g., on a tray. The umbilical cord biomaterial is then placed on a drying frame, preferably a plastic mesh drying frame (e.g., QUICK COUNT® Plastic Canvas, Uniek, Inc., Waunakee, Wis.). In other embodiments, the drying frame may be any autoclavable material, including but not limited to a stainless steel mesh. Once the umbilical cord biomaterial is positioned on the drying frame, a sterile gauze can be placed on the drying platform of a heat dryer (or gel-dryer) (e.g., Model 583, Bio-Rad Laboratories, Hercules, Calif.), so that an area slightly larger than the umbilical cord biomaterial resting on the plastic mesh drying frame is covered. Preferably, the total thickness of the gauze layer does not exceed the thickness of one folded 4×4 gauze. Any heat drying apparatus may be used that is suitable for drying sheet-like material. The drying frame is placed on top of the gauze on the drying platform so that the edges of the plastic frame extend above beyond the gauze edges, preferably between 0.1-1.0 cm, more preferably 0.5-1.0 cm. In some embodiments, another plastic framing mesh is placed on top of the umbilical cord biomaterial. In another embodiments, a sheet of thin plastic (e.g., SW 182, clear PVC, AEP Industries Inc., South Hackensack, N.J.) or a biocompatible silicone is placed on top of the biomaterial covered mesh so that the sheet extends well beyond all of the edges. In this embodiment, the second mesh frame is not needed.
  • In an alternative embodiment, the umbilical cord biomaterial is placed one or more sterile sheets of TYVEK® material (e.g., a sheet of TYVEK® for medical packaging, DuPont TYVEK®, Wilmington, Del.), optionally, with one sheet of TYVEK® on top of the biomaterial (prior to placing the plastic film). This alternate process will produce a smoother version of the biomaterial (i.e., without the pattern of differential fiber compression regions along and perpendicular to the axis of the material), which may be advantageous for certain applications, such as for example for use as a matrix for expansion of cells.
  • Drying the umbilical cord biomaterial. In a preferred embodiment, the invention encompasses heat drying the umbilical cord biomaterial of the invention under vacuum. While the drying under vacuum may be accomplished at any temperature from about 0° C. to about 60° C., the umbilical cord biomaterial is preferably dried at between about 35° C. and about 50° C., and most preferably at about 50° C. It should be noted that some degradation of the collagen is to be expected at temperatures above 50° C. The drying temperature is preferably set and verified using a calibrated digital thermometer using an extended probe. Any amount of vacuum that can be conveniently generated can be used, but preferably, the vacuum pressure is set to about −22 inches of Hg. The drying step is continued until the umbilical cord biomaterial is substantially dry, that is, contains less than 20% water by weight, and preferably, about 3-12% water by weight as determined for example by a moisture analyzer. To accomplish this, the umbilical cord biomaterial may be heat-vacuum dried, e.g., for approximately 60 minutes to achieve a dehydrated umbilical cord biomaterial. In some embodiments, the umbilical cord biomaterial is dried for about 30 minutes to 2 hours, preferably about 60 minutes. Although not intending to be bound by any mechanism of action, it is believed that low (e.g., <50° C.) heat coupled with vacuum pressure allows the umbilical cord biomaterial to achieve the dehydrated state without denaturing collagen in the biomaterial. After completion of the drying process in accordance with the invention, the umbilical cord biomaterial can be cooled down, e.g., for approximately two minutes, with the vacuum pump running.
  • Packaging and Storing of the Umbilical Cord Biomaterial. Once the umbilical cord biomaterial is dried, the biomaterial is gently lifted off the drying frame. Preferably, handling of the umbilical cord biomaterial at this stage is done with sterile gloves. The umbilical cord biomaterial can be placed in a sterile container, e.g., peel pouch. When dried, the umbilical cord biomaterial produced in accordance with the methods of the invention may be stored at room temperature for an extended period of time as described supra.
  • In another embodiment, the umbilical cord biomaterial is prepared as above, but is not decellularized. That is, the umbilical cord membrane is obtained and dried, but the cells associated with the umbilical cord membrane are not removed. The final, dried product thus comprises, e.g., the umbilical cord membrane and/or umbilical cord vessel(s), as well as cellular components.
  • The umbilical cord biomaterial can be dehydrated by other methods in place of, or in addition to, the vacuum-drying method outlined above. For example, in one embodiment, the biomaterial can be freeze dried. Typically, umbilical cord biomaterial can be frozen at a temperature between about −170° C. and about 0° C. for time sufficient for the biomaterial to completely freeze. The frozen biomaterial is freeze-dried process during which the ice crystals will be removed or avoided by sublimation under vacuum.
  • In another embodiment, the biomaterial can be dehydrated using a solvent. For example, umbilical cord biomaterial can be dehydrated using, e.g., ethanol and acetone. In this specific embodiment, the biomaterial can be, e.g., soaked for a time in a series of ethanol-acetone mixtures (e.g., 20%, 40%, 60%, 80% and 100% ethanol, or a similar progression of equivalent solvents that act to extract water) such that the water inside the biomaterial is gradually replaced by the organic solvent. After the final soak, the biomaterial can be placed in a well-ventilated place at room temperature (about 23° C.) for a time sufficient for the solvent to evaporate. The biomaterial can alternately be vacuum-dried after the solvent soak.
  • In another embodiment, the biomaterial can also be dehydrated by freeze drying. For example, in a specific embodiment in a combination of the above two processes, the processed membrane can be first frozen and then transferred to a water miscible organic solvent. Ice crystals inside the membrane tissue may then be dissolved and replaced by the organic solvent using a series of progressive solvent soaks as described above. After the final soak, the biomaterial can be placed in a well-ventilated place at room temperature (e.g., about 20° C. to about 25° C.) for a time sufficient for the ethanol to evaporate. The biomaterial can alternately be vacuum-dried after the 100% ethanol soak.
  • Non-heat drying processes may be preferred if the porous structure of the biomaterial and/or bioactivity of the biological substances within the umbilical cord membrane need to be preserved, for example, if the biomaterial is to be used as a substrate or matrix for the transport of stem cells to a graft site, or if, e.g., the biomaterial is to be preloaded with a heat-sensitive drug as a drug release device, or if, e.g., the biomaterial is preloaded with a heat sensitive drug as a drug release device.
  • When the above steps are complete, the membrane generally primarily comprises collagen (types I, III, IV, V, VI and VII), glycosaminoglycans (particularly hyaluronic acid); and growth factors, particularly fibroblast growth factor (FGF), basic fibroblast growth factor (b-FGF), epidermal growth factor (EGF), insulin-like growth factor I (IGF-I), platelet-derived growth factor (PDGF) and transforming growth factor beta (TGF-β).
  • 5.1.5 Storage and Handling of Umbilical Cord Biomaterial
  • Dehydrated umbilical cord biomaterial may be stored, e.g., as dehydrated sheets, at room temperature (e.g., 25° C.) prior to use. In certain embodiments, the umbilical cord biomaterial can be stored at a temperature of at least 10° C., at least 15° C., at least 20° C., at least 25° C., or at least 29° C. Preferably, umbilical cord biomaterial, in dehydrated form, is not refrigerated. In some embodiments, the umbilical cord biomaterial may be refrigerated at a temperature of about 2° C. to about 8° C. The umbilical cord biomaterial produced according to the methods of the invention can be stored at any of the specified temperatures for 12 months or more with no alteration in biochemical or structural integrity (e.g., no degradation), without any alteration of the biochemical or biophysical properties of the umbilical cord biomaterial. The biomaterial can be stored for several years with no alteration in biochemical or structural integrity (e.g., no degradation), without any alteration of the biochemical or biophysical properties of the biomaterial. The biomaterial can be stored in any container suitable for long-term storage. Preferably, the umbilical cord biomaterial of the invention is stored in a sterile double peel-pouch package.
  • The umbilical cord biomaterial, in embodiments in which the material has been dried, may be hydrated prior to use, using, e.g., a sterile physiological buffer. In a specific embodiment, the sterile saline solution is a 0.9% NaCl solution. In some embodiments the sterile saline solution is buffered. In certain embodiments, the hydration of the umbilical cord biomaterial requires at least 2 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, or at least 20 minutes. In a preferred embodiment, the hydration of the umbilical cord biomaterial is complete within 5 minutes. In yet another preferred embodiment, the hydration of the umbilical cord biomaterial of the invention is complete within 10 minutes. In yet another embodiment, the hydration of the umbilical cord biomaterial takes no more than 10 minutes. Once hydrated, the umbilical cord biomaterial can be maintained in solution, e.g., in sterile 0.9% NaCl solution, for up to six months, with a change of solution, e.g., every three days.
  • 5.1.6 Sterilization
  • Sterilization of the umbilical cord biomaterial may be accomplished by any medically-appropriate means, preferably means that do not significantly alter the tertiary and quaternary structure of the biomaterial proteins. Sterilization can be accomplished, for example, using gas, e.g., ethylene dioxide. Sterilization can be accomplished using radiation, for example, gamma radiation, and is preferably done by electron beam irradiation using methods known to one skilled in the art, e.g., Gorham, D. Byrom (ed.), 1991, Biomaterials, Stockton Press, New York, 55-122. Any dose of radiation sufficient to kill at least 99.9% of bacteria or other potentially contaminating organisms is within the scope of the invention. In a preferred embodiment, a dose of at least 18-25 kGy is used to achieve the terminal sterilization of the biomaterial.
  • 5.1.7 Laminates
  • The umbilical cord biomaterial may be laminated to provide greater load-bearing capacity and durability during the healing process. Laminates of the umbilical cord biomaterial can comprise biomaterial from a single umbilical cord, wherein the composition is folded once, or a plurality of times, longitudinally or laterally, or both. Laminates of the biomaterial can also comprise two or more sheets of biomaterial.
  • The umbilical cord biomaterial, being anisotropic, has two orientations, longitudinal (that is, along the length of the umbilical cord membrane) and lateral (that is, around the width of the umbilical cord). Where a laminate comprises two sheets of the umbilical cord biomaterial, the sheets can be laminated so that each sheet is oriented the same way (e.g., each sheet oriented longitudinally), or such that at least one sheet is oriented laterally and one longitudinally. In other embodiments, each of the layers of umbilical cord biomaterial can be laminated in any orientation with respect to any other layer of the biomaterial in the laminate.
  • The umbilical cord has a sidedness; that is, the umbilical cord biomaterial has an epithelial side (that is, the side towards the interior of the umbilical cord) and a mesothelial side (that is, the side towards the exterior of the umbilical cord). Laminates can comprise two or more layers of the umbilical cord biomaterial in any sidedness configuration. For example, laminates can comprise layers of umbilical cord biomaterial in which only the endothelial sides of the layers are in contact; only the mesothelial sides of the layers are intact; or a combination of both. In one embodiment, a laminate comprises four layers, wherein two sets of two layers, contacted endothelial to mesothelial sides, are contacted by the exposed mesothelial side such that the two faces of the laminate show the endothelial sides.
  • Umbilical cord biomaterial can be laminated, e.g., by folding a single sheet of biomaterial, or by stacking 2 or more layers of the biomaterial one atop the other, and sealing or drying. The biomaterial may be laminated either dry or after rehydration. Alternatively, two or more layers of, e.g., umbilical cord biomaterial, or composition comprising an umbilical cord biomaterial, can be laminated prior to initial drying after cell removal, e.g., after a cell scraping step (see Examples, below). If laminated prior to the initial drying, 2 or more biomaterial layers can be stacked one atop the other and subsequently dried, using, for example, a freeze-drying process, or drying under moderate heat with or without vacuum. The heat applied preferably is not so intense as to cause breakdown or decomposition of the protein components, especially the collagen, of the umbilical cord biomaterial. Typically, the heat applied is less than about 70° C., preferably less than about 60° C., and, more preferably, is approximately 50° C. Lamination time varies with, e.g., the number of layers being laminated, but typically takes 1-2 hours at 50° C. Thus, a method of preparing a laminate using a composition comprising umbilical cord membrane comprises layering a plurality of said membranes in contact with each other to form a laminate. In some embodiments, each of said membranes comprises less than 20% water by weight prior to said layering. In certain embodiments, said laminate is dried to less than 20% water by weight after said layering.
  • The biomaterial may also be laminated using an adhesive applied between 2 or more layers of biomaterial or umbilical cord biomaterial or composition comprising an umbilical cord biomaterial. Such an adhesive is preferably appropriate for medical applications, and can comprise a natural biological adhesive, for example fibrin glue, a synthetic adhesive, or combinations thereof. The adhesive may further be chemically converted from precursors during the lamination process.
  • Laminates of the umbilical cord biomaterial can comprise, for example, umbilical cord biomaterial that has been decellularized, biomaterial that retains the cellular material (that is, where the cells have been killed, but not removed), or biomaterial comprising living umbilical cord cells or cells of another type (e.g., where cells have been cultured on a sheet of umbilical cord biomaterial).
  • A laminate can comprise a second type of material, e.g., one or more layers of umbilical cord biomaterial can be layered with one or more layers of a second biologically-compatible material, e.g., a sheetlike material such as, e.g., amniotic membrane. Where the second material has a “grain” or orientation, the umbilical cord biomaterial can be laminated such that the biomaterial lies with its longitudinal direction along, or alternatively across, the grain of the second material. In a specific, preferred embodiment, the umbilical cord membrane biomaterial is laminated with at least one other layer of a second material that has a relatively high load-bearing capacity. The umbilical cord biomaterial can also be laminated with a non-biological material, e.g., plastic, e.g., TYVEK® or the like. In a preferred embodiment, the umbilical cord biomaterial laminate comprises two layers of the biomaterial and one layer of plastic, e.g., TYVEK®, such that the plastic is sandwiched between the two layers of umbilical cord biomaterial, and the endothelial sides of the biomaterial contact the plastic. In another preferred embodiment, one layer of umbilical cord membrane is placed on a plastic sheet with the epithelial side down, and a second piece of umbilical cord membrane is placed on the first epithelial side up. The resulting product is then heat-dried to produce an umbilical cord biomaterial laminate.
  • In various embodiments, the second material has a load-bearing capacity or tensile strength, for, e.g., a 2-centimeter wide section, of at least 25 milliPascals (mPa), 50 mPa, 75 mPa, 100 mPa, 125 mPa, 150 mPa, 175 mPa, 200 mPa, 225 mPa, 250 mPa, 275 mPa, 300 mPa, 325 mPa, 350 mPa, 375 mPa, 400 mPa, 425 mPa, 450 mPa, 475 mPa, 500 mPa, 750 mPa, 1000 mPa, 1250 mPa, 1500 mPa, 1750 mPa or 2000 mPa. Load-bearing capacities of sections that are wider or narrower would be accordingly more or less. Such a second material can be sheetlike, or can be formed into a shape suitable for a particular application, and the umbilical cord biomaterial molded to the shape of the second material. For example, the second material can be, e.g., a material suitable for, and shaped for, tendon or ligament repair. The umbilical cord biomaterial can be wrapped around such a second material so that the exterior of the laminate is biologically compatible, and the interior is load-bearing.
  • The load-bearing capacity of a particular umbilical cord biomaterial laminate, or piece of umbilical cord biomaterial, can be tested by standard methods known in the art, such as ASTM D1708 (Standard Test Method for Tensile Properties of Plastics).
  • In one embodiment, the laminate comprises at least two sheets of umbilical cord biomaterial approximately the same size and shape laid one atop the other so that the shape is substantially maintained. Such a laminate can be trimmed to finalize a particular shape. In another embodiment, the laminate comprises two or more sheets of umbilical cord biomaterial, wherein a portion of each of the sheets overlaps another. In this embodiment, several overlapping sheets of umbilical cord biomaterial can be laminated to form a larger sheet of the biomaterial than would be possible from a single umbilical cord. In a specific embodiment, such a laminate of overlapping sheets of the biomaterial can itself be laminated with another layer of a material, e.g., another overlapping biomaterial laminate; individual sheets of umbilical cord biomaterial; another type of biomaterial, e.g., an amniotic membrane-derived biomaterial; an artificial sheet or film; etc.
  • 5.1.8 Stem Cells
  • The umbilical cord biomaterial as described herein can also comprise stem or progenitor cells. The umbilical cord biomaterial can comprise, e.g., mesenchymal or mesenchymal-like stem cells, for example, those described in U.S. Pat. Nos. 5,486,359, 6,261,549 and 6,387,367, or placental stem cells such as those described in U.S. Application Publication Nos. 2002/0123141, 2003/0032179 and 2003/0180269. However, the umbilical cord biomaterial may comprise stem or progenitor cells, preferably mammalian stem or progenitor cells, from any tissue source. The umbilical cord biomaterial can comprise embryonic stem cells or embryonic germ cells.
  • The umbilical cord biomaterial and stem or progenitor cells can be combined, e.g., in advance of a procedure in which the biomaterial is contacted with an individual having a disease, disorder or condition that would be amenable to treatment using an umbilical cord biomaterial. For example, stem cells can be contacted with, e.g., disposed onto, the biomaterial sufficiently in advance of such a procedure for a plurality, a majority, or substantially all of the stem cells to adhere to the biomaterial. The stem cells can be contacted with the biomaterial immediately before the biomaterial is contacted with the individual. The stem cells can also be contacted with the biomaterial in situ, after the biomaterial is contacted with the individual. The number of stem or progenitor cells disposed onto the surface of the umbilical cord biomaterial may vary, but may be at least about 1×106, 5×106, 1×107, 5×107, 1×108, 5×108, 1×109, 5×109, 1×1010, 5×1010, 1×1011, 5×1011, or 1×1012; or may be no more than 1×106, 5×106, 1×107, 5×107, 1×108, 5×108, 1×109, 5×109, 1×1010, 5×1010, 1×1011, 5×1011, or 1×1012 stem or progenitor cells.
  • The stem cells, at any of the times noted above, can be contacted with one or more differentiation-modulating agents, for example, the differentiation-modulating agents described in U.S. Application Publication Nos. 2003/0235909 and/or 2004/0028660, the disclosures of which are incorporated by reverence in their entireties herein, or International Application Publication No. WO 03/087333. Methods of differentiating stem cells to, for example, epidermal, mesodermal, and other cell types are known in the art, and are described, e.g., in U.S. Application Publication No. 2004/0028660.
  • 5.2 Uses of Umbilical Cord Biomaterial
  • The umbilical cord biomaterial can be used in a variety of medical applications. The uses outlined in this section are non-limiting examples of such applications.
  • 5.2.1 Repair of Joints, Ligaments, and Tendons
  • The umbilical cord biomaterial of the invention can be used, for example, to repair or replace ligaments, tendons and/or cartilage. The biomaterial can be contacted with a ligament, tendon or cartilage in any medically-acceptable manner that tends to facilitate healing of a defect in the ligament, tendon or cartilage.
  • The umbilical cord biomaterial can be used, for example, to repair a defect in a tendon, ligament or cartilage where the defect is a tear, that is, a defect that is less than a complete failure. The repair comprises contacting the tendon, ligament or cartilage with the umbilical cord biomaterial such that a part, or preferably all, of the defect is covered by the biomaterial. The umbilical cord biomaterial can be, for example, wrapped around a tendon or ligament at the site of the defect, or a patch of umbilical cord biomaterial of sufficient size to cover the defect can be placed on the defect. The biomaterial can be held in place using, e.g., a biologically-acceptable glue, e.g., a tissue glue, or can be sutured in place. Preferably, the umbilical cord biomaterial is held in place such that the biomaterial assumes at least part of the biomechanical load normally associated with the functional tendon, ligament or cartilage.
  • The umbilical cord biomaterial can be used, for example, to repair a defect in a tendon, ligament or cartilage where the defect is a complete rupture or failure of a tendon, ligament or cartilage. The repair comprises contacting the tendon, ligament or cartilage with the umbilical cord biomaterial so as to partially, or preferably completely, cover the defect. Preferably, the contacting is such that the biomaterial assumes some, or all of the biomechanical load normally assumed by the tendon, ligament or cartilage. For example, a rupture in a tendon can be repaired by contacting the tendon at the site of the rupture with the biomaterial and affixing the biomaterial in place. The two parts of the tendon at the site of the rupture are preferably brought into contact with each other, and the biomaterial is preferably wrapped around the site of the break. The biomaterial can be sutured to the two parts of the ruptured tendon such that it acts as a splint, holding the to ends of the rupture together. The biomaterial can be wrapped around such a rupture once, or preferably a plurality of times.
  • The umbilical cord biomaterial can be used, for example, to replace a tendon or ligament, or to support a joint where one or more of the native ligaments is weakened. In such an embodiment, the biomaterial attaches two bones, or a muscle and bone, in place of a damaged, diseased or ruptured tendon or ligament, e.g., an anterior cruciate ligament.
  • In a specific embodiment, for example, to replace a cruciate ligament, e.g., an anterior cruciate ligament, tunnels can be drilled onto the femoral and tibial heads adjoining the knee. The biomaterial, e.g., folded lengthwise a plurality of times into a rope-like conformation, can be drawn through the tunnels, and the ends fastened to the respective bones. Such fastening can be accomplished by any means known in the art, e.g., using screws, staples, or the like. In a specific embodiment, the ends of the umbilical cord biomaterial are folded over to provide a portion of the biomaterial of greater thickness for fastening. Such replacement can comprise removing the native ligament during replacement, or can comprise adding the biomaterial and allowing the native ligament to remain. In preferred embodiments, the umbilical cord biomaterial is laminated with a load-bearing material, such as plastic sheeting, e.g., TYVEK® prior to folding into the rope-like conformation.
  • In another embodiment, the umbilical cord biomaterial can be used to repair a tendon in the hand. In a specific embodiment, for example, the umbilical cord biomaterial is inserted between one or more extensor tendons in the hand and bones to provide a gliding surface or a shield between tendon and bone, e.g., to prevent adhesion formation.
  • Preferably, the umbilical cord biomaterial is stretched prior to repair or replacement of a ligament or tendon. For example, a weight, bearable by the particular piece of biomaterial, can be suspended from the biomaterial for a time sufficient to allow up to, e.g., about 10%, 15%, or about 20% elongation. Such stretching tends to prevent loosening of the biomaterial after application.
  • The umbilical cord biomaterial, in another embodiment, can also be used to repair or reinforce a rotator cuff tendon having a defect, e.g., a tear. In a preferred embodiment, a piece of the umbilical cord biomaterial is used to completely cover the rotator cuff tendon defect.
  • 5.2.2 Tympanic Membrane Repair and Other Aural Applications
  • In another embodiment, the present invention provides methods and compositions for repair of a tympanic membrane using an umbilical cord biomaterial. In one embodiment, the present invention provides a method of repairing a perforated tympanic membrane, comprising contacting said tympanic membrane with a umbilical cord biomaterial. Said contacting can comprise shaping a flat piece of the biomaterial into the shape of an entire, or a portion of, a tympanic membrane, and contacting the biomaterial with the tympanic membrane. In another specific embodiment, said perforation has not healed spontaneously within two months of the appearance of the perforation. As with other applications, the biomaterial can be contacted with the tympanic membrane while hydrated, or, preferably, while substantially dry (e.g., comprising less than 20% water by weight). The biomaterial can be a single layer, or can be a laminate of two or more layers. In another embodiment, the umbilical cord biomaterial (whether a single layer or a laminate) contacted with the tympanic membrane is at least about 70 microns in thickness.
  • In one embodiment of repairing a tympanic membrane, a tympanic membrane having a perforation is contacted with an umbilical cord biomaterial such that the biomaterial partially or totally occludes the perforation. The perforation to be occluded may be a central perforation, that is, a perforation of any size that does not involve the margin of the tympanic membrane (i.e., the periphery seated in the auditory canal), or a marginal perforation (i.e., a perforation touching upon, or largely involving, the margin of the tympanic membrane). In another embodiment, only the tympanic membrane is perforated, and no other ear structure is perforated or damaged. In another embodiment, occlusion of the perforation is an adjunct to at least one other surgical procedure involving the outer, middle, or inner ear. In another embodiment, the repair of the tympanic membrane is a tympanoplasty. In another embodiment, the repair of the tympanic membrane is a myringoplasty.
  • The benefits of closing a tympanic membrane perforation include prevention of water entering the ear while showering, bathing or swimming (which could cause ear infection), improved hearing, and diminished tinnitus. Closure also helps to prevent the development of cholesteatoma (skin cyst in the middle ear), which can cause chronic infection and destruction of ear structures.
  • Tympanoplasty and myringoplasty are generally outpatient procedures. The otolaryngologist may approach repair of a tympanic membrane perforation either through the auditory canal (trans-canal approach), or via a post-auricular incision followed by folding the ear forward to expose the tympanic membrane (post-auricular approach).
  • Before attempting any correction of the perforation, a hearing test is generally performed, and the patient is evaluated for Eustachian tube function, as partial or complete loss of Eustachian tube function can exacerbate a tympanic membrane puncture and interfere with the adherence of a graft to the tympanic membrane. Repair of a perforated tympanic membrane generally comprises placing an occluding material on the membrane. The patient is evaluated for complications, such as extension of squamous epithelium through the perforation and into the middle ear space. In such instances, tympanoplasty or myringoplasty is preferably accompanied, where possible, by remediation of the complication.
  • The present invention encompasses repair of a tympanic membrane with an umbilical cord biomaterial either as a first or subsequent therapy. That is, the biomaterial may be used to repair a tympanic membrane deformity, such as a perforation, before other remedial measures are tried. Alternatively, repair of a tympanic membrane with biomaterial may be performed after one or more other remedial measures have been tried and failed.
  • In one embodiment, repair of a tympanic membrane with biomaterial may additionally comprise applying an anti-infective agent to the graft and/or surrounding ear canal. Thus, in one embodiment, the invention provides a method of repairing a tympanic membrane comprising contacting the tympanic membrane with an umbilical cord biomaterial and an anti-infective agent, e.g., one of the anti-infective agents listed in Section 5.1.2, above. The anti-infective agent can be contacted either prior to, concurrently with, or subsequent to contacting the tympanic membrane with the umbilical cord biomaterial. The anti-infective agent can be present separate from, or as an integral part of, the biomaterial. For example, the anti-infective agent can be present on the surface of the biomaterial, or can be impregnated in the biomaterial. In a specific example, the anti-infective agent is an antibiotic, a bacteriostatic agent, antiviral compound, a virustatic agent, antifungal compound, a fungistatic agent, or an antimicrobial compound. In a specific embodiment, the anti-infective agent is ionic silver. In a more specific embodiment, the ionic silver is contained within a hydrogel. Ionic silver hydrogel is a preferred anti-infective agent because it is broad spectrum, with no known bacterial resistance; its application and removal are pain-free, and the hydrogel supports autolytic debridement. In a preferred embodiment, the umbilical cord biomaterial is impregnated with silver ions prior to application to the tympanic membrane. In another embodiment, the umbilical cord biomaterial is impregnated with silver ions after application of the biomaterial to the tympanic membrane, for example, by application of ear drops.
  • The invention further provides that the use of an umbilical cord biomaterial to repair a tympanic membrane deformity may be the sole treatment of the tympanic membrane, or may be in addition to another therapies or treatment used simultaneously in the course of treating or repairing a tympanic membrane. For example, the invention provides for the repair of a tympanic membrane comprising contacting the tympanic membrane with an umbilical cord biomaterial, and treating the tympanic membrane using an additional therapy not comprising contacting the tympanic membrane with an umbilical cord biomaterial, where the contacting and the additional therapy individually or together cause a measurable improvement in, maintenance of, or lessening of the worsening of, at least one aspect of a tympanic membrane deformity, as compared to a tympanic membrane not contacted with an umbilical cord biomaterial.
  • The invention further provides for the use of umbilical cord biomaterial to repair an ear condition in conjunction with repair of a tympanic membrane. For example, the umbilical cord biomaterial can be used to reconstruct or repair the outer or middle ear structures, including the auditory canal and middle ear chamber. The umbilical cord biomaterial, for example, may be used to repair or line the mastoid cavity, particularly where mastoid reconstruction is indicated in addition to tympanoplasty. In one embodiment, the umbilical cord biomaterial may be used to line the mastoid cavity where the mastoid cavity comprises exposed bone, that is, bone with no covering epithelial cell layer. In another embodiment, the umbilical cord biomaterial may be used as a oval window graft in stapes surgery, either alone or in conjunction with tympanoplasty or myringoplasty.
  • 5.2.3 Soft Tissue Repair
  • In another non-limiting embodiment, the invention further provides for the use of umbilical cord biomaterial to repair a soft tissue injury or defect in an individual. In one embodiment, the soft tissue defect is an abdominal wall defect. Such an abdominal wall defect, e.g., hernia, is repaired by suturing one or more sheets of umbilical cord biomaterial, either a single sheet or a laminate of sheets as disclosed elsewhere herein, either dried or hydrated, to the soft tissue such that the defect is repaired or ameliorated. In a specific embodiment, the soft tissue defect is a hernia or abdominal wall defect in which the abdominal wall allows exit of at least part of an organ from the abdominal cavity. In this embodiment, the defect, e.g., discontinuity or hernia, is preferably completely covered with one or more sheets of umbilical cord biomaterial, whether or not the discontinuity or hernia has been surgically closed. Typically, the umbilical cord biomaterial is sutured, stapled, or otherwise fastened to the defect such that repair is effected. In other specific embodiments, said soft tissue defect is a defect in the pelvic floor, an enteroceles, a rectoceles, or a cystoceles. In embodiments in which the abdominal wall defect is an opening of the abdominal wall to the exterior of the body, it is generally preferred that the defect be contacted with the mesothelial surface of the umbilical cord biomaterial.
  • In another specific embodiment, the defect is incontinence, and the umbilical cord biomaterial is used as an adjunct to a suburethral sling procedure to assist in the repair of gracilis muscle flaps sutured beneath the urethra.
  • In another specific embodiment, the soft tissue defect is a leg ulcer such as, e.g., a venous leg ulcer, arterial leg ulcer, diabetic ulcer or decubitus ulcer. Repair of a leg ulcer can comprise contacting a portion, or the entirety, of the leg ulcer with one or more pieces of umbilical cord biomaterial, either a single sheet or a laminate thereof, either dried or hydrated, such that the umbilical cord biomaterial becomes affixed to the leg ulcer. Typically, the biomaterial becomes affixed to the leg ulcer without fastening; however, the biomaterial can be sutured, stapled or glued to the skin surrounding the ulcer, or can be held in place by, e.g., a bandage or compression boot, or by any other method known to those of skill in the art.
  • In another specific embodiment, the soft tissue defect is a surgical adhesion. In a more specific embodiment, the surgical adhesion is an adhesion resulting from, e.g., gynecological surgery. It is estimated that approximately 97% of surgical patients develop adhesions after surgery and of these, between 5% and 8% develop complications. Without being bound by theory, the umbilical cord biomaterial would be an effective barrier to adhesions in that the amnion epithelial cells on one side of the biomaterials would occupy cell binding sites, making it difficult for host cells, such as fibroblasts, to attach and penetrate. Thus, the umbilical cord biomaterial can be used to prevent surgical adhesion by placing the biomaterial between two tissues that would ordinarily be expected to form a post-surgical adhesion.
  • In another specific embodiment, the soft tissue defect is a nasal septal perforation. The septal perforation may arise from any cause, e.g., inherited defect, trauma, drug use, etc. A piece of umbilical cord biomaterial, suitably shaped, can be placed along the septum in order to partially or completely occlude the perforation, and can be held in place by one or several sutures or tissue glue.
  • 5.2.4 Ocular Plugs
  • In another non-limiting embodiment, the invention further provides for the use of umbilical cord biomaterial in the formation of an ocular plug. An ocular plug at least partially, or, preferably, completely, occludes a hole in, e.g., the sclera, that is, e.g., caused by an injection, formed as a part of a surgical procedure to, e.g., allow insertion of a surgical tool into the lumen of the eye; caused by trauma; etc. Ocular plugs may be configured in any shape to accomplish the particular purpose at hand, e.g., occluding injection or ocular surgery-related holes in the sclera, prevention of leakage, drug delivery, anchoring of the plug, etc.
  • In one, preferred, embodiment, the invention provides an ocular plug that comprises a shaft attached to and extending from a cap. Typically, the cap is circular when viewed from the upper face. However, the cap may be oval, square, rectangular, polygonal, irregular, or may appear as a plurality of flanges extending substantially perpendicularly from the shaft. The upper face of the cap distal to the shaft, may be hemispherical, curved to a degree other than completely hemispherical, or may be substantially flat. Preferably, the surface of the upper face of the cap is shaped to approximate the curvature of the eye to promote comfort and reduce the possibility of inflammation or irritation associated with the eyelid moving over the face of the cap. The lower face of the cap proximal to the shaft may be substantially flat, but is preferably shaped to approximate the curvature of the eye. Preferably, the cap tapers towards the edges so that a smooth transition is made from sclera to cap when the eyelid passes over the cap. However, the cap need not taper from center towards the edges, and may have a discernibly blunt edge.
  • The cap is preferably of a sufficient diameter to promote seating and maintenance of position of the plug within the hole in the sclera, and to reduce the possibility of the shaft from passing completely through the sclera during or after insertion of the plug into the sclera. The outer diameter of the cap may be from 1-10 times the diameter of the shaft; preferably, the outer diameter of the cap is between 1-3 times the diameter of the shaft.
  • The shaft, as the remainder of the plug, may be configured to accomplish occlusion of an injection- or ocular surgery-related scleral hole. The shaft may be thin enough, for example, to occlude the hole made by a 33 gauge, or thinner, needle after intravitreous injection, or may be as thick as 1-2 mm in diameter, or more, to occlude holes created during, for example, macular hole surgery. The shaft may be of any size appropriate to occlude a particular discontinuity in the sclera. The shaft is preferably at least as long as a sclera is thick, but may be shorter than the thickness of a sclera, or may be longer. A typical sclera is 0.35-0.55 mm thick, but may be thicker or thinner. The thickness depends upon the particular individual, as well as the position of the discontinuity in the sclera; for example, the sclera tends to thin away from the iris and towards the retina. Where the shaft is longer than the thickness of a sclera, the shaft, when the plug comprising it is fully inserted, projects through the sclera an into the vitreous humor.
  • The surface of the shaft may be smooth or textured. For example, the surface of the shaft may be rough, ribbed or knurled so as to enhance contact between the plug and sclera, thereby reducing the potential for the plug to work its way out of the scleral hole. Particularly where the plug comprises a cap, the shaft may be ribbed or knurled directionally; that is, ribbed or knurled to promote insertion of the plug into the scleral hole and to discourage passage of the plug in the opposite direction, i.e., back out of the scleral hole.
  • In a preferred embodiment, the shaft is substantially cylindrical. In another embodiment, the shaft is substantially cylindrical along its entire length. In other embodiments, the shaft is ovoid, square, rectangular, square or rectangular with rounded edges, polygonal, or irregular in cross-section. In another embodiment, the shaft comprises a narrow portion and a wide portion. Typically, the shaft is attached to the cap through the narrow portion; the wide portion, distal to the cap, facilitates anchoring of the plug into the sclera. In one embodiment, the cross-sectional area of the wide portion is greater than that of the narrow portion. The wide portion of the shaft may be manufactured in a variety of configurations. For example, the shaft may flare. Such a flare may be substantially continuous along the length of the shaft, or may begin at any point along the length of the shaft. In another embodiment, the wide portion is a flange or protrusion from a portion of the main body of the shaft, e.g., from one side of the shaft. Such a flange or protrusion may have any shape that facilitates maintenance of the plug within the scleral hole while not substantially increasing the difficulty of insertion or the potential for scleral damage during insertion. In another embodiment, the wide portion comprises a flange or other protrusion that substantially encircles the shaft. For example, the wide portion may be an inverted cone or frustum, wherein the larger radius of the frustum is wider than the diameter of the shaft. In another embodiment, the wider portion of the shaft is a cylinder having a radius larger than the radius of the shaft. In another embodiment, the wider portion of the shaft has substantially the same cross-sectional shape as the shaft, but a cross-sectional area larger than the cross-sectional area of the shaft. There is no need, however, for the wide portion of the shaft to have a particular shape relative to the cross-sectional shape of the shaft, and the wide portion need not have the same cross-sectional shape as the shaft. In another embodiment, the shaft comprises a thread spirally disposed along a portion or all of the length of the shaft, so that the shaft of the plug functions as a screw. In this embodiment, the thread may proceed clockwise or counterclockwise along the shaft.
  • In another embodiment, the plug may be constructed so that the portion of the shaft distal to the cap comprises one or more flaps that may be folded against the shaft during insertion of the plug into the sclera, and which open, or fold away, from the shaft once the flap has been pushed completely through the sclera. The one or more flaps would act as an anchor.
  • In one embodiment, the wider portion of the shaft extends into the sclera itself, and serves as an anchor. In another embodiment, part or all of the wide portion of the shaft extends into the vitreous humor.
  • The end of the shaft distal to the cap may be flat, rounded, or tapered, or may be irregular. The surface of the end may be substantially perpendicular to the longitudinal axis of the shaft, or may be tilted, giving the end of the shaft a barbed appearance.
  • In one embodiment, the ocular plug comprises an opening extending at least the portion of the cap distal to the shaft, and, optionally, into the shaft. The opening can be used, for example, to receive a wire of fixed gauge. The wire is used to pick up the ocular plug and guide the ocular plug into a scleral discontinuity.
  • In another embodiment, the ocular plug does not comprise a cap. For example, the ocular plug may comprise a shaft only. In this embodiment, the shaft may be formed in any of the configurations as for the shafts of a plug with a cap as discussed above. For example, in its simplest form, the plug may simply be a cylinder, with a smooth, ribbed, knurled or textured surface, or may comprise one or more wide portions that can act as anchors. In one embodiment, the shaft (that is, the plug) comprises two wide portions. In a specific embodiment, the shaft is dumbbell-shaped. The dumbbell shape may be accomplished, for example, by thickening the ends of the shaft so that the change in thickness from center of the shaft to either end is continuous; alternatively, the change in thickness from center of the shaft to either end is discontinuous. Preferably, in this embodiment, the length of the shaft between the wide portions (e.g., the ends of the dumbbell) is at least the thickness of the sclera.
  • Ocular plugs may be pre-made to standard sizes, or may be custom-made to fill particular scleral holes or discontinuities, whether anticipated (as in the case of surgery) or unanticipated. In one embodiment, therefore, the invention provides an ocular plug, wherein said ocular plug has a shaft of a reproducible, standard size. The ocular plug may also be custom-made for a particular discontinuity. In specific embodiments, the standard or custom-made diameter size of a shaft for said ocular plug is a diameter sufficient to substantially occlude a scleral hole caused by passage of a 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12 or larger gauge needle. In other specific embodiments, the standard or custom-made diameter of a shaft for said ocular plug is about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5 millimeters, or wider. In other specific embodiments, the standard length of the shaft of said plug is 0.35, 0.40, 0.45, 0.50, 0.55, 0.60, 0.65, 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1.00, 1.05, 1.10, 1.15, 1.20, 1.25, 1.30, 1.35, 1.40, 1.45 or 1.50 millimeters.
  • Ocular plugs may be made from umbilical cord biomaterial by any method used to create or produce molded devices. Preferably, the umbilical cord biomaterial used to make an ocular plug is a dried, decellularized biomaterial.
  • Ocular plugs may be made, for example, by stamping the plugs from a sheet of the biomaterial using a shaped stamp. Alternatively, the plugs may be cut from a sheet of the biomaterial, or may be formed by removal of unwanted material from a block of the biomaterial. In a preferred embodiment, an ocular plug is formed using a mold. Where the ocular plug is formed using a mold, the biomaterial is preferably first made into a liquid, slurry, paste, or similar material amenable to forming in a mold.
  • In an exemplary embodiment of a method of making the ocular plug, umbilical cord biomaterial is first reduced to a collection of particles; that is, the biomaterial is micronized. The biomaterial may be micronized to a particle size of anywhere from about 1 micron to about 1 millimeter. Generally, the larger the particle size, the more porous the plug. Any method may be used to micronize the biomaterial, for example, ultrasound, physical shearing, homogenization, etc. Such micronization may be done dry (that is, by micronizing the biomaterial without any additional liquid), or may be done using a micronization liquid or carrier. If micronization with a liquid is performed, the liquid may be any physiologically acceptable liquid or solution that does not significantly degrade the biodegradable material, e.g., the tertiary structure of the proteins comprising the biomaterial. Typically, the ratio of biomaterial to liquid is 25 mg/ml to 300 mg/ml, but more or less of the biomaterial may be used. Determination that a desired particle size has been achieved may be accomplished by any means known in the art, e.g., microscopic examination, comparison to bead size standards, etc.
  • Once the desired micronized biomaterial is obtained, whether in wet or dry form, the micronized biomaterial is injected or otherwise forced into the mold and allowed to set. In one embodiment, the wet micronized biomaterial is forced into the mold and is then frozen, e.g., at a temperature of from −5° C. to −160° C. (though higher or lower temperatures would also work) for a time sufficient to allow ice crystals to form and grow, e.g., 2 hours to several days. The frozen plug is then freeze dried to substantial dryness, that is, to a water content of about 20% by weight or less. Preferably, any plugs formed using the biomaterial are freeze-dried to substantial dryness. Freeze-drying is particularly preferred as the process allows for the development of pores in the biomaterial constituting the plug. Plugs may also be heat-dried, but heat applied to dry the plugs is preferably not heat that would cause the breakdown of any component of the biomaterial. For example, in various embodiments, an ocular plug formed from biomaterial may be dried at about 70° C., about 65° C., about 60° C., about 55° C., about 50° C. or about 45° C., or less than about 70° C., less than about 65° C., less than about 60° C., less than about 55° C., less than about 50° C. or less than about 45° C.
  • Once the plugs are freeze-dried, or dried by other method, the biomaterial is preferably cross-linked to provide mechanical stability and integrity. Crosslinking may be accomplished by any method known in the art; particularly preferred are radiation, chemical, or heat crosslinking. Radiation crosslinking is preferred. Radiation used may be any known in the art to be useful for such a purpose, for example, electron-beam or e-beam radiation, gamma radiation or ultraviolet radiation. E-beam radiation is preferred. See, e.g., Odland, U.S. Pat. No. 5,989,498 “E-beam Sterilization of Biological Materials,” The intensity of radiation used may be that ordinarily used for the sterilization of medical instruments. The biomaterial may also be chemically crosslinked using any chemical crosslinking methodology known in the art, for example, thiol-thiol crosslinking, amide-amide crosslinking, amine-thiol crosslinking, amine-carboxylic acid and thiol-carboxylic acid crosslinking, etc., as appropriate for the material from which the plug is made. The plug may also be heat crosslinked, typically using a thermal dehydration process. Most preferably the heat used for such crosslinking does not significantly degrade or structurally weaken the material in any way. Plugs may be heat crosslinked, for example, by placing the plugs in a vacuum oven at 105° C. for 1-5 hours, or until the desired structural integrity or degree of crosslinking is achieved.
  • 5.2.5 Other Uses
  • The umbilical cord biomaterial can be used in any other medical application in which like materials are used, e.g., as a patch for wound repair.
  • In one embodiment, the umbilical cord biomaterial is used as a wrapping or covering for a replacement eye orb, e.g., a hydroxyapatite sphere.
  • In another embodiment, the umbilical cord biomaterial can be used to repair or ameliorate a cardiac defect. Such defects include, but are not limited to, cardiac wall defects, areas of necrosis due, e.g., to ischemia; repair of a cardiac valve; repair of patent foramen ovale, and the like. In another embodiment, the umbilical cord biomaterial is seeded or inoculated with cardiomyocytes. Immediately, or following culture to allow proliferation of the cardiomyocytes, the umbilical cord biomaterial is implanted into a cardiac defect as a living patch. In preferred embodiments, the umbilical cord biomaterial is placed so as to cover the defect completely.
  • 5.3 Kits
  • The present invention further provides kits comprising one or more pieces of umbilical cord membrane biomaterial. Preferably, each of the one or more pieces the biomaterial is individually sterilely wrapped, e.g., in a peel-pouch.
  • In a more specific embodiment, the kit comprises a piece of umbilical cord biomaterial that is at least about 2×8 cm. In another embodiment, said kit comprises a piece of umbilical cord biomaterial approximately the size of an eardrum. The kit may comprise one or more pieces of umbilical cord biomaterial and any other medical device, disposable or drug that would facilitate treatment of a disease, disorder or condition treatable using the biomaterial. Preferably, each piece of the umbilical cord biomaterial in the kit is provided as a single sheet or patch in a sterile container or wrapping separate from the remainder of kit contents. In another embodiment, the kit comprises two or more pieces of umbilical cord biomaterial, separately wrapped or contained. In another embodiment, said kit comprises a support for the umbilical cord biomaterial. In specific embodiments, the support may be a natural or a synthetic material. In other specific embodiments, said support is a plastic film, plastic sheet, or a stretchable plastic wrap. In another embodiment, said kit comprises one or more disposables, e.g., paper tissues or towels, mats, cotton swabs, plastic or rubber gloves, disposable forceps, or the like. In a specific embodiment, said disposables are bandages, means for sterilizing skin, swabs, gloves, or sterile sheets. In another embodiment, said kit comprises an anti-infective agent, for example, an antibiotic ointment, cream, or spray. In another embodiment, said kit comprises a piece of umbilical cord biomaterial and one or more wound healing agents. In a specific embodiment, said wound healing agent is PDGF, TGF, hyaluronic acid, fibrin, or fibronectin. In another embodiment, said kit comprises umbilical cord biomaterial and a means for applying compression to a part of the body. In a specific embodiment, of any of the kits above, the kit comprises an instruction sheet suitable for use by a non-medical end user; an instruction sheet suitable for use by an end user in a medical profession; or a materials safety data sheet; or a combination thereof.
  • 6. EXAMPLES 6.1 Example 1 Production of Umbilical Cord Biomaterial
  • The following example demonstrates one method of preparing umbilical cord biomaterial.
  • Materials and Equipment
  • The following items were obtained and, where appropriate, sterilized: human placenta (less than 48 hours old at the start of processing); surgical clamps/hemostats; scissors; scalpels; tweezers; Halsted mosquito; Adson bayonet forceps; grooved directors; cell scraper; autoclaved gauze; stainless steel rinsing trays; stainless steel cups; stainless steel processing trays. 0.9% NaCl solution; sterile water; specimen containers; personal protective equipment (including sterile and non-sterile gloves); certified clean room; decellularizing solution (0.5% deoxycholic acid solution); rocking platform (VWR Model 100); timer (VWR TRACEABLE° model); disinfected silicone grid; PVC wrap film; vacuum pump (Schuco-Vac 5711-130); heat dryer (BioRad Model 583); sterile cutting board; pouches for packaging (COT-360, 361, 362); stainless steel ruler; TRACEABLE° Digital Thermometer (Model 61161-364, Control Company); Accu-Seal Automatic Sealer (Accu-Seal, Model 630-1 B6 or 730-16B) with air compressor; and waterproof resealable bags (CCT-03S).
  • Procedure
  • A sterile field was set. The placenta was removed from the transport container and placed into a sterile stainless steel tray. Using surgical damps and scissors, the umbilical cord was cut off approximately 2 inches from the placental disc. The umbilical cord was rinsed with sterile 0.9% NaCl solution as many times as necessary to remove as much blood as possible; optionally, fingers were used to squeeze remaining blood from vessels. The umbilical cord was optionally placed in a separate sterile container cup prefilled with sterile 0.9% NaCl solution, if the cord did not have to be processed immediately. The harvested umbilical cord was placed in a refrigerator at 4° C. until use. The placental disk was placed back into the transport container to be utilized for other projects, or discarded.
  • The umbilical cord was processed as follows. The umbilical cord was removed from the specimen container, and squeezed to remove any remaining blood from vessels prior to introducing the umbilical cord to a processing tray. The umbilical cord was placed into a sterile stainless steel processing tray, and cut into segments 12 to 15 cm in length. The umbilical cord vein was then located for each segment, and canalized using an Adson bayonet forceps or grooved director. The vein and umbilical cord were then cut longitudinally, using scissors, until both the vein and umbilical cord were fully open. The umbilical cord, was placed on the processing tray with the opened vein side facing upward. The umbilical cord and vein were then bluntly dissected longitudinally between the vein wall and the umbilical cord wall with sterile tweezers or mosquito clamps. When both sides were separated, the vein was carefully removed. After vein removal, the two arteries were located and removed in the same manner. Depending on the purpose of the study, the resulting umbilical cord membrane (biomaterial) was placed in either in saline solution or in 1% deoxycholic acid solution (a decellularizing solution) and stored at 4° C. until serological testing results become available.
  • Storage and Quarantine of Umbilical Cord Membrane
  • The umbilical cord membrane, obtained as outlined above, was kept in sterile 0.9% saline solution or 1% deoxycholic acid solution for 10-20 days at 4° C. until serological test results, if ordered, were available. Saline solution, where used, was changed every 3 days. 1% deoxycholic acid solution, when used, was changed every 5 days.
  • Umbilical Cord Membrane Cleaning and Rinsing
  • A sterile field was set with a new set of sterilized trays as above. The Umbilical cord membrane was removed from the refrigerator and placed into a stainless steel processing tray. Sterile 0.9% saline solution is added to cover the bottom of the tray. All, or substantially all, residual deoxycholic solution, where used, was removed, and remaining cells and debris were removed from both sides of the tissue using a cell scraper and sterile tweezers. Sterile 0.9% saline solution was used as needed to aid in removal of the cells and debris. The umbilical cord membrane was rinsed three times in a separate stainless steel rinsing tray filled with sterile 0.9% saline solution. The saline solution was changed between each cleaning step. The umbilical cord membrane was then placed into a new sterile specimen container containing about 150 mL saline solution, and placed on a rocking platform for agitation for 5 minutes at setting #6. The scraping and rinsing steps were repeated once as necessary. The umbilical cord membrane was then placed into a sterile specimen container containing 150 mL sterile water, and placed on a rocking platform for agitation for 20 minutes at setting #6. This rinsing step was repeated three times.
  • Drying the Umbilical Cord Membrane
  • A TYVEK® sheet was placed onto a stainless steel processing tray. The cleaned umbilical cord membrane segments were removed from the specimen container one piece at a time, and excess fluid was gently squeezed out. The membrane segments were then placed on the surface of the TYVEK® sheet, epithelium side up, and gently stretched until flat. The membrane was then dried at about 50° C.±1.0° C. in a vacuum dryer. Sterile gauze was placed on the drying platform of the vacuum dryer, covering an area slightly larger than the area of the TYVEK® sheet. The total thickness of the gauze layer did not exceed the thickness of one folded 4×4 gauze. A sheet of silicone framing mesh was placed on top the gauze, smooth side up. The TYVEK® sheet with the tissue was then placed on the heat dryer platform on top of the silicone mesh. Another TYVEK® sheet was then placed on top of the tissue. A piece of PVC wrap film was then cut large enough to cover the entire drying platform, and pulled so that the film pulled tightly against the TYVEK® sheet (that is, was “sucked in” by vacuum) and so that there were no air leaks and no wrinkles over the tissue area). The vacuum pump was then set to approximately −22 inches Hg, and heat/vacuum drying was allowed to proceed for a total of about 120 minutes. Approximately 30-45 minutes into the drying process, the sterile gauze layer was replaced.
  • A new sterile field was set with a sterilized drying kit and cutting board. With the pump still running, the plastic film was removed from the TYVEK®, and the sheet and tissue were placed on a cutting board with the epithelium side of the tissue facing upward. The dried membrane (now umbilical cord biomaterial) was then gently removed from the TYVEK® sheet. The biomaterial segments were then cut with a scalpel into segments of a specified size, typically 2×2 cm or 1×1 cm. The dried, sized umbilical cord biomaterial was then placed and sealed into a peel-pouch package.
  • 6.2 Example 2 Production of Umbilical Cord Biomaterial Laminate
  • Objective: To increase the size of a sheet of umbilical cord biomaterial for hernia repair.
  • Materials and Methods: All cited dimensions are approximate. Umbilical cord membrane from a 23.5 hour-old placenta was collected and processed as in Example 1 up to the point of drying. The final size of the membrane was approximately 35 cm by 4 cm. The membrane was cut into three pieces approximately 10 cm long. The pieces were arranged so as to overlap by about 2 cm on the long edge, and were dried at 50° C. between two sheets of TYVEK®.
  • Results: The biomaterial comprising laminated membrane thus obtained was approximately 10 cm by 10 cm. The sections did not separate upon rehydration in saline for 72 hours.
  • Umbilical cord membrane can also be laminated by placing two or more pieces of the biomaterial, interior (of the umbilical cord) side down, on a substrate in a mounting frame. The laminated membrane is then placed in a gel dryer and dried to substantial dryness (≦about 20% water content by weight) to produce a laminated umbilical cord biomaterial.
  • Another method of constructing a thicker biomaterial is to laminate intact umbilical cord (including Wharton's jelly but lacking arteries and vein). The intact cord is then processed, e.g., by rinsing, soaking in a solution such as a buffered saline solution, e.g., phosphate buffered saline, or a mild ionic or nonionic detergent solution. The cord is dried in a vacuum dryer to create an intact, double-layer biomaterial. Two or more layers of this double-layer material can be laminated by layering the biomaterials and drying further in a heated vacuum dryer. The drying/dehydration process can be heat drying or any other processes as described below.
  • 6.3 Characterization of Dried Umbilical Cord Biomaterial
  • A study was undertaken to examine biomaterial made of heat dried human umbilical cord membrane (HUC) after sterilization by different doses of gamma irradiation. Samples of HUC were sterilized with 0, 20, 25, 30, or 40 kGy and then examined for water uptake (mass and thickness change), denaturation temperature, and tensile mechanical properties. HUC samples had been incubated for either 10 or 20 days in 1% D-cell (deoxycholic acid) solution during preparation.
  • 6.3.1 Water Uptake
  • Individual samples of HUC for each condition (n=3) were weighed on a microbalance. Samples were then incubated in 10 mL of phosphate buffered saline at 37° C. for 1 hour. Samples were removed from the PBS and blotted dry a minimum of three times with a KIMWIPE® tissue. The samples were again weighed on a microbalance. The percentage water uptake ([wet weight (Ww)−dry weight (Wd)]/Wd*100) and equilibrium water content ([Ww−Wd]/Ww*100) were calculated.
  • FIG. 1 summarizes the results of the rehydration for HUC incubated for 10 and 20 days. The initial water uptake of the control (nonsterilized samples) was much higher for HUC incubated for 20 days than for HUC incubated for 10 days, possibly due to the loosening of the membrane proteins by the detergent effect of the deoxycholic acid in the D-cell solution. Water uptake and equilibrium water content closely matched for the 10 and 20 days samples that were sterilized at all radiation doses. There was a linear decrease in the water uptake and the equilibrium water content of both sets of samples with increasing radiation dose. Even at the highest radiation dose, the membranes took up at least their own weight in water.
  • 6.3.2 Changes in Thickness
  • Individual dog bone shaped (see, e.g., FIG. 4) samples of HUC for each condition (n=8) were mounted in squares of vellum paper so that the membrane could be easily handled during and after hydration. The thickness was measured in three locations for each sample and averaged. Samples were then incubated in 10 mL of phosphate buffered saline at 37° C. for 1 hour. Thickness measurements were repeated after hydration.
  • Overall, the average dry thickness of the HUC was ˜70 μm, with the 10 and 20 day samples, having average thickness of 57 and 86 μm respectively (Table 1 and Table 2). There appeared to be no correlation between radiation dose and dry thickness of the HUC. After rehydration, there was a marked difference in thickness between the sterilized and non-sterilized samples. There was little difference in the rehydrated thickness between the 10 and 20 day samples. Without wishing to be bound by any theory or mechanism, the difference observed can be due to cross-linking of collagen molecules caused by irradiation. There appeared to be a decrease in the magnitude of the thickness change upon rehydration with increasing dose; this effect was more pronounced with the 10 day samples.
  • TABLE 1
    Changes in thickness of 10 day incubated HUC during hydration
    Dose increase
    (KGy) dry (um) SD wet (um) SD (%) SD
    0 61 23 243 82 316% 109% 
    20 44 10 107 20 147% 19%
    25 58 24 132 40 134% 28%
    30 67 22 125 45  87% 32%
    40 57 23 106 46  91% 36%
  • TABLE 2
    Changes in thickness of 20 day incubated HUC during hydration
    Dose increase
    (KGy) dry (um) SD wet (um) SD (%) SD
    0 88 22 279 51 240% 123% 
    20 76 22 149 26 106% 48%
    25 85 50 130 21  83% 61%
    30 61 23 138 51 125% 31%
    40 121 32 209 46  77% 36%
  • When the change in thickness of the membranes was compared to the water uptake (FIG. 2), there was a loose correlation between the amount of water taken up and the increase in thickness. The magnitude of the water uptake and the change in thickness both decreased with increasing gamma radiation dose. There was a stronger correlation between water uptake and thickness change for samples incubated for 10 days than those incubated for 20 days. This is due partially to the fact that the samples incubated for 20 days showed less difference between samples, but greater variability within a set of samples.
  • 6.3.3 Denaturation Temperature
  • Individual samples of HUC for each condition (n=3) were incubated in 10 mL of phosphate buffered saline at 37° C. for 1 hour. Samples were removed from the PBS and blotted dry a minimum of three times with a KIMWIPE® tissue. The samples were sealed in aluminum hermetic differential scanning calorimeter (DSC) pans and tested in a TA Instruments modulated DSC (Q1000) in standard mode from 5-110° C. at 10° C./min. TA Instruments' “Universal Analysis” software was used to calculate the onset and peak values of the denaturation point of the membranes.
  • FIG. 3 graphically summarizes the denaturation temperature results for the HUC samples. The results are identical for samples incubated for 10 and 20 days. Onset and peak temperatures were only a few degrees different for each of the samples, and both onset and peak temperatures decreased linearly with increasing radiation dose. There was very low variability in the results.
  • Time of incubation in D-cell solution did not affect the denaturation temperature. There was a linear decrease in the onset and peak denaturation temperatures with increasing radiation dose for both the 10 and 20 day samples. From the denaturation data, there appeared to be no difference between soaking HUC for 10 or 20 days in 1% D-cell solution.
  • 6.3.4 Tensile Properties
  • Individual dog bone shaped samples of human umbilical cord membrane for each condition (n=8) were mounted in squares of vellum paper so that the membrane could be easily handled during and after hydration. Samples were then incubated in 10 mL of phosphate buffered saline at 37° C. for 1 hour. The tensile properties of the membranes were evaluated based on American Society for Testing and Materials protocol D1708 (ASTM D1708); see FIG. 4 for more details of testing. Samples that had been cut along the long axis of the cord (longitudinal sections) and samples that had been cut perpendicular to the long axis (cross section) were tested where samples were available for each condition.
  • No differences were found between 10 and 20 day samples. Tests on the unsterilized human umbilical cord membrane determined the tissue to be anisotropic, with the tensile properties being different depending on the direction in which the samples were oriented (Table 3). Longitudinal samples had a higher stress at break and a higher modulus than cross section samples, and cross sectional samples had a greater range of values for the extension at break (how far the tissue stretched before breaking). In all cases, when the load was removed from the unsterilized samples, the tissue appeared by visual inspection to return to its original shape with no obvious distortions. Force was applied at a rate of 33 mm/min.
  • TABLE 3
    Tensile testing results for non-sterilized HUC (10 and 20 day samples)
    Stress Extension
    at break Modulus at break
    Orientation (MPa) (MPa) (%)
    Longitudinal 2.3-8.8 14.2-27.5 135-270
    Cross 0.1-3.5 0.5-8.9  60-480
  • While there were no differences between the tensile properties of the 10 and 20 day samples or the different radiation doses, there were striking differences in how unsterilized and sterilized samples failed. Nonsterilized samples failed by breakage of the sample, while the sterilized samples failed mostly by delamination of layers of tissue. Additionally, the sterilized tissue was noticeably deformed after the load had been removed.
  • The tensile properties of HUC most closely resembled skin (Table 4). This was true of both the sterilized and non-sterilized tissue.
  • TABLE 4
    Comparison of tensile properties of HUC (10 and 20 day samples)
    and other tissues
    Tensile Tensile Extension
    strength modulus at break
    (MPa) (MPa) (%)
    Tendon/ligament   100-2,000 50-150  5-50
    Articular cartilage 1-10
    Skin 10-40 2-20  50-200
    Compact bone 10,000-20,000 2-3
    HUC (long) non-sterile 2.3-8.8 14.2-27.5  135-270
    HUC (long) (sterile)  1.1-12.4 7.4-51.4  60-258
  • 6.3.5 Suture Pull-Out Strength
  • The umbilical cord biomaterial was determined to have a superior suture pull-out strength compared to dried human amniotic membrane. In a test similar to that described in Section 6.3.4, above, one short side of a 1×2 section of umbilical cord biomaterial was glued to vellum paper, and the other short side sutured to a second piece of vellum paper, as depicted in FIG. 5A. The pieces of vellum paper were held by grips, and a load was applied to the suture at a rate of about 12.7 mm/min. The umbilical cord biomaterial demonstrated an average pull-out resistance of about 1.4 Newtons (N), with a range of about 0.75 N to about 2.4 N, while the dried amniotic membrane demonstrated a pull-out resistance averaging about 0.3 N. See FIG. 5B.
  • 6.4 Example 4 Biocompatibility of Umbilical Cord Biomaterial
  • This study evaluated the host response to an implant made of umbilical cord biomaterial during absorption following subcutaneous implantation in a rat model.
  • 6.4.1 Materials and Methods
  • Test materials for implantation consisted of umbilical cord biomaterial or high density polyethylene (HDPE; control). Umbilical cord biomaterial was provided as dried umbilical cord membrane measuring approximately 1 cm×1 cm sections prepared in either 0.9% NaCl (Test article A; non-decellularized) or 1% deoxycholic acid (Test articles B and C; decellularized).
  • The 16 rats used in the experiments were ten week old male Rattus norvegicus strain H1A®:(SD)CVF® (Hilltop Lab Animals, Inc.). Animal weight at the time of implantation ranged from 344 grams to 392 grams. Maintenance of animals during the experiment conformed to Standard Operating Procedures based on the “Guide for the Care and Use of Laboratory Animals”.
  • On the day of the implant, each rat was identified and weighed. Groups of four animals were arbitrarily assigned to be terminated 1 week, 3 weeks, 6 weeks or eight weeks after implantation (Table 5).
  • TABLE 5
    Implantation of test articles:
    Bilateral
    Animal Implantation Termination
    Number Left Right Interval
    1 C A 1 week
    2 C A
    3 B A
    4 B A
    5 C A 3 weeks
    6 C A
    7 B A
    8 B A
    9 C A 6 weeks
    10 C A
    11 B A
    12 B A
    13 C A 8 weeks
    14 C A
    15 B A
    16 B A
  • For implantation, the animals were anesthetized by intraperitoneal injection of ketamine hydrochloride and xylazine (66 mg/kg and 9 mg/kg, respectively) dosed at 2.25 ml/kg. The implant region was scrubbed with a germicidal soap and wiped with 70% alcohol. Separate incisions were made on each side of the back through the skin and parallel to the lumbar region of the vertebral column. A pocket was formed by blunt dissection in the subcutaneous tissue on each side of the back. One section of the test material was implanted into each pocket such that it lay as flat in the pocket as reasonably possible. A nonsorbable suture was cut into approximately 1 cm length sections and placed at each test article implantation site as a location marker. One section of the negative control article (HDPE) was similarly implanted caudally to the sections of the test article. The skin was closed with wound clips.
  • Following implantation, the animals were observed daily for general health, and the incisions were examined for adverse reactions until wound clip removal. Detailed examinations for clinical signs of disease or abnormality were conducted weekly and at termination.
  • At 1, 3, 6 and 8 weeks after implantation, the designated animals were weighed and euthanized by carbon dioxide inhalation. Macroscopic observation of the viscera was conducted. The general appearance of the skin at the implantation sites was recorded. The implant sites were exposed by incision along the midline from the proximal to the distal end of the rat, and the skin was gently pulled away from the implantation site. The implanted materials were measured to the nearest millimeter (length and width) and the color and consistency of the surrounding tissue was documented. The sites were also photographed. The implant sites and any abnormal tissues were excised and preserved in 10% neutral buffered formalin (NBF) until further processing. After fixation, the implant sites and any abnormal tissues were histologically processed (embedded, sectioned and stained with hematosylin and eosin) for microscopic evaluation by a pathologist.
  • Implantation sites were evaluated to assess any change in the integrity of the form of the test material. The local tissue response was evaluated and compared to the reactions at the negative control article sited. The evaluation included characterization of the test material in regard to acute inflammation, chronic inflammation, granulation tissue formation, foreign body reaction, and foreign body giant cell formation. In addition, the formation and the thickness change of the fibrous capsule around the implants, the change in implants' characteristics at degradation (e.g., size and shape, formation of particles, fibers and amorphous gel, etc.) were also evaluated. Microscopic cellular changes were graded according to severity on a scale of 0 to 4.
  • 6.4.2 Results
  • Clinical observations. All animals appeared clinically normal throughout the study, Minor scabs or ulcerations were noted at the anesthetic injection sites; these areas resolved without treatment.
  • Body weight data. In general, all rats gained weight over the course of the study, and weight gains were considered acceptable.
  • Macroscopic observations. Generally, all animals, and the appearance of the skin, appeared macroscopically normal following termination. In general, the color and consistency of the tissue surrounding the implant appeared normal for all animals from three week termination interval. At the one week termination interval, some signs of surgical trauma was still evident, which is typical. For each animal, symptoms arising from implantation of the test articles (umbilical cord biomaterial) were mild enough that the biomaterial was considered a nonirritant.
  • Implant absorption. Implant size at the various termination points is presented in Tables 6 and 7. In several cases, the implant was found to have been completely absorbed by the host. Average implant sizes for the test articles at each of the terminations points is shown in Table 8.
  • TABLE 6
    Measurements of implanted materials at termination - left side
    Animal Length Presence/Absence
    Group # Site (mm) Width (mm) of Test Material
    1 Week 1 Test 9.8 7.8 Present
    Control 11.8 10.0 Present
    2 Test 8.6 8.9 Present
    Control 13.5 10.7 Present
    3 Test 10.5 10.9 Present
    Control 13.8 10.7 Present
    4 Test 10.0 8.8 Present
    Control 14.1 11.4 Present
    3 Weeks 5 Test 5.3 8.3 Present
    Control 11.0 13.6 Present
    6 Test 4.2 6.2 Present
    Control 12.2 15.6 Present
    7 Test 7.2 5.0 Present
    Control 10.9 13.1 Present
    8 Test 8.2 7.6 Present
    Control 11.4 14.0 Present
    6 Weeks 9 Test 6.9 6.9 Present
    Control 8.9 10.9 Present
    10 Test 5.9 4.6 Present
    Control 10.5 13.5 Present
    11 Test 6.1 6.2 Present
    Control 10.6 11.8 Present
    12 Test 6.2 6.9 Present
    Control 11.4 10.5 Present
    8 Weeks 13 Test 8.4 6.0 Present
    Control 12.1 12.1 Present
    14 Test NA NA Absent
    Control 14.8 12.0 Present
    15 Test NA NA Absent
    Control 12.7 14.0 Present
    16 Test 9.3 8.1 Present
    Control 12.5 13.5 Present
  • TABLE 7
    Measurements of implanted materials at termination - right side
    Animal Length Presence/Absence
    Group # Site (mm) Width (mm) of Test Material
    1 Week 1 Test 10.4 9.9 Present
    Control 12.2 12.2 Present
    2 Test 7.5 10.1 Present
    Control 12.1 11/2 Present
    3 Test 7.8 6.8 Present
    Control 10.8 11.6 Present
    4 Test 8.3 6.6 Present
    Control 11.7 12.2 Present
    3 Weeks 5 Test 5.5 8.0 Present
    Control 12.3 13.2 Present
    6 Test 6.1 5.5 Present
    Control 11.5 10.9 Present
    7 Test NA NA Absent
    Control 10.7 10.2 Present
    8 Test 7.6 5.5 Present
    Control 10.3 11.7 Present
    6 Weeks 9 Test NA NA Absent
    Control 10.6 9.5 Present
    10 Test NA NA Absent
    Control 10.4 14.9 Present
    11 Test NA NA Absent
    Control 11.4 10.5 Present
    12 Test NA NA Absent
    Control 10.2 12.7 Present
    8 Weeks 13 Test 7.8 5.6 Present
    Control 12.3 11.6 Present
    14 Test 11.2 5.9 Present
    Control 14.4 12.6 Present
    15 Test NA NA Absent
    Control 13.5 12.2 Present
    16 Test NA NA Absent
    Control 11.7 13.8 Present
  • TABLE 8
    Average length and width of test articles
    Average Length and Width of the Test Articles
    4830 4825 4838
    Length Width Length Width Length Width
    Interval (mm) (mm) (mm) (mm) (mm) (mm)
    1 weeks 9.2 8.4 10.3 9.9 8.5 8.4
    3 weeks 4.8 7.3 7.7 6.3 4.8 4.8
    6 weeks 6.4 5.8 6.2 6.6 Absorbed Absorbed
    8 weeks 4.2 3.0 4.7 4.1 4.8 5.8
  • Further results indicated that both decellularized and non-decellularized umbilical cord biomaterial have excellent biocompatibility. Animal responses to both biomaterials was similar to control USP grade HDPE. No fibrous tissue encapsulations developed during the course of the implantation study, whereas HDPE implants developed fibrosis at later stages of the study. While both decellularized and non-decellularized biomaterials were biodegradable, it was noted that implanted decellularized membranes lasted longer (e.g., about 6 to 8 weeks) than non-decellularized biomaterials (about 3 to 6 weeks).
  • EQUIVALENTS
  • The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims.
  • Various publications, patents and patent applications are cited herein, the disclosures of which are incorporated by reference in their entireties.

Claims (22)

1. An umbilical cord-derived biomaterial comprising an isolated mammalian umbilical cord membrane and Wharton's jelly, wherein said biomaterial comprises less than 20% water by weight.
2. The biomaterial of claim 1 that comprises at least one umbilical vessel.
3. The biomaterial of claim 1 that is decellularized.
4. The biomaterial of claim 1 comprising an exogenous bioactive molecule.
5. The biomaterial of claim 4, wherein said bioactive molecule is a cytokine or a growth factor.
6. The biomaterial of claim 4, wherein said bioactive molecule is an extracellular matrix protein.
7. The biomaterial of claim 6, wherein said extracellular matrix protein is collagen, fibronectin, elastin, vitronectin, or hyaluronic acid.
8. The biomaterial of claim 4, wherein said bioactive molecule is hyaluronic acid.
9. The biomaterial of claim 8, wherein said hyaluronic acid is crosslinked to said umbilical cord membrane.
10. The biomaterial of claim 1, comprising an exogenous polymer.
11. The biomaterial of claim 10, wherein said exogenous polymer is a synthetic biodegradable polymer or an anionic polymer.
12. The biomaterial of claim 4, wherein the bioactive molecule is an antibiotic, a hormone, a growth factor, an anti-tumor agent, an anti-fungal agent, an anti-viral agent, a pain medication, an anti-histamine, an anti-inflammatory agent, an anti-infective agent, a wound healing agent, a wound sealant, a cellular attractant, a scaffolding reagent, or a small molecule.
13. The biomaterial of claim 12, further comprising a hydrogel composition.
14. The biomaterial of claim 1, additionally comprising an exogenous stem cell.
15. The biomaterial of claim 3, additionally comprising an exogenous stem cell.
16. The biomaterial of claim 14, wherein said exogenous stem cell is a placental stem cell, a mesenchymal stem cell, an embryonic stem cell, or a somatic stem cell.
17. The biomaterial of claim 16, wherein said somatic stem cell is a neural stem cell, a hepatic stem cell, a pancreatic stem cell, an endothelial stem cell, a cardiac stem cell, or a muscle stem cell.
18. A laminate comprising a plurality of layers, wherein at least one of the layers comprises the biomaterial of claim 1.
19. A method of producing a biomaterial comprising isolating and decellularizing a composition comprising an umbilical cord membrane.
20. A biomaterial made by the method of claim 19.
21. A method of delivering a therapeutic agent to a subject comprising contacting the subject with the composition of claim 1, wherein said composition comprises a therapeutic agent.
22. A method of repairing a tympanic membrane having a deformity, comprising contacting said tympanic membrane with a biomaterial comprising an isolated mammalian umbilical cord membrane.
US13/333,172 2006-08-15 2011-12-21 Umbilical cord biomaterial for medical use Abandoned US20120114712A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/333,172 US20120114712A1 (en) 2006-08-15 2011-12-21 Umbilical cord biomaterial for medical use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US83813206P 2006-08-15 2006-08-15
US11/893,409 US8105634B2 (en) 2006-08-15 2007-08-15 Umbilical cord biomaterial for medical use
US13/333,172 US20120114712A1 (en) 2006-08-15 2011-12-21 Umbilical cord biomaterial for medical use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/893,409 Division US8105634B2 (en) 2006-08-15 2007-08-15 Umbilical cord biomaterial for medical use

Publications (1)

Publication Number Publication Date
US20120114712A1 true US20120114712A1 (en) 2012-05-10

Family

ID=38779720

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/893,409 Active 2030-11-05 US8105634B2 (en) 2006-08-15 2007-08-15 Umbilical cord biomaterial for medical use
US13/333,172 Abandoned US20120114712A1 (en) 2006-08-15 2011-12-21 Umbilical cord biomaterial for medical use

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/893,409 Active 2030-11-05 US8105634B2 (en) 2006-08-15 2007-08-15 Umbilical cord biomaterial for medical use

Country Status (2)

Country Link
US (2) US8105634B2 (en)
WO (1) WO2008021391A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8796315B2 (en) 2009-06-25 2014-08-05 Darlene E. McCord Methods for improved wound closure employing olivamine and human umbilical vein endothelial cells
US9144555B2 (en) 2012-11-30 2015-09-29 Darlene E. McCord Hydroxytyrosol and oleuropein compositions for induction of DNA damage, cell death and LSD1 inhibition
US10314688B2 (en) 2016-08-24 2019-06-11 Arthrex, Inc. Tissue use for repair of injury
US11511017B2 (en) 2019-03-12 2022-11-29 Arthrex, Inc. Ligament reconstruction

Families Citing this family (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US7311905B2 (en) * 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
KR101132545B1 (en) * 2001-02-14 2012-04-02 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
US7498171B2 (en) * 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
WO2004047770A2 (en) * 2002-11-26 2004-06-10 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
KR20070002067A (en) * 2004-03-26 2007-01-04 셀진 코포레이션 Systems and methods for providing a stem cell bank
US8182840B2 (en) 2005-09-27 2012-05-22 Tissue Tech, Inc. Amniotic membrane preparations and purified compositions and therapy for scar reversal and inhibition
US8187639B2 (en) 2005-09-27 2012-05-29 Tissue Tech, Inc. Amniotic membrane preparations and purified compositions and anti-angiogenesis treatment
EP2530145A1 (en) 2005-10-13 2012-12-05 Anthrogenesis Corporation Immunomodulation using placental stem cells
NZ567334A (en) * 2005-10-13 2012-08-31 Anthrogenesis Corp Production of oligodendrocytes from placenta-derived stem cells
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
EP1976978A2 (en) * 2005-12-29 2008-10-08 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US9598669B2 (en) 2005-12-29 2017-03-21 Anthrogenesis Corporation Composition for collecting placental stem cells and methods of using the composition
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
NZ606814A (en) 2006-10-23 2014-10-31 Anthrogenesis Corp Methods and compositions for treatment of bone defects with placental cell populations
KR20090109127A (en) * 2007-02-12 2009-10-19 안트로제네시스 코포레이션 Hepatocytes and chondrocytes from adherent placental stem cells? and cd34?, cd45? placental stem cell-enriched cell populations
NZ597779A (en) * 2007-02-12 2013-07-26 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
US20100172830A1 (en) * 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
WO2008151040A2 (en) * 2007-05-31 2008-12-11 Cook Biotech Incorporated Analgesic coated medical product
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
CA2736663C (en) 2007-09-07 2018-01-02 Surgical Biologics, Llc. Placental tissue grafts and improved methods of preparing and using the same
CN101978045A (en) * 2007-09-26 2011-02-16 细胞基因细胞疗法公司 Angiogenic cells from human placental perfusate
EP3524253A1 (en) 2007-09-28 2019-08-14 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
KR20210056449A (en) * 2007-11-07 2021-05-18 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
US9480549B2 (en) 2008-04-25 2016-11-01 Allosource Multi-layer tissue patches
US9358320B2 (en) * 2008-04-25 2016-06-07 Allosource Multi-layer tissue patches
EP3539380A3 (en) 2008-08-20 2019-12-18 Celularity, Inc. Improved cell composition and methods of making the same
KR20180108887A (en) * 2008-08-20 2018-10-04 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
CA2734446C (en) 2008-08-22 2017-06-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
CA2739166C (en) * 2008-10-10 2013-12-31 Julio Font Perez New biomaterial from wharton's jelly of the human umbilical cord
RU2562154C2 (en) * 2008-11-19 2015-09-10 Антродженезис Корпорейшн Amniotic adhesive cells
WO2010060031A1 (en) * 2008-11-21 2010-05-27 Anthrogenesis Corporation Treatment of diseases, disorders or conditions of the lung using placental cells
CA2767014C (en) 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
ES2767252T3 (en) * 2009-08-25 2020-06-17 Tissue Tech Inc Umbilical cord amniotic membrane products
ES2646750T3 (en) * 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
US20110212065A1 (en) * 2010-02-18 2011-09-01 Timothy Jansen Methods of manufacture of therapeutic products comprising vitalized placental dispersions
CN102198292B (en) * 2010-03-26 2013-06-26 卢世璧 Scaffolds of umbilical cord decellularized Wharton jelly for tissue engineering and preparation method thereof
WO2011120535A1 (en) 2010-03-30 2011-10-06 Histocell, S.L. New biomaterial from wharton's jelly umbilical cord
TWI578993B (en) 2010-04-07 2017-04-21 安瑟吉納西斯公司 Angiogenesis using placental stem cells
TW201138792A (en) 2010-04-08 2011-11-16 Anthrogenesis Corp Treatment of sarcoidosis using placental stem cells
US20130121972A1 (en) 2010-06-01 2013-05-16 Rouzbeh R. Taghizadeh Native wharton's jelly stem cells and their purification
US8840665B2 (en) 2010-06-11 2014-09-23 Liventa Bioscience, Inc. Method of tendon repair with amnion and chorion constructs
US20120010708A1 (en) * 2010-07-08 2012-01-12 AFcell Medical Amnion and chorion replacement cover and uses thereof in surgical repair of muscles
KR20130093091A (en) 2010-07-13 2013-08-21 안트로제네시스 코포레이션 Methods of generating natural killer cells
US9433490B2 (en) 2010-12-22 2016-09-06 University Of Florida Research Foundation, Inc. Multilayered implant materials derived from amniotic membrane, methods of making the multilayered implant materials, and method of using multilayered implant materials
EP2658557A1 (en) 2010-12-31 2013-11-06 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
KR102331661B1 (en) 2011-02-14 2021-11-25 미메딕스 그룹 인크. Micronized placental tissue compositions and methods for making and using the same
EP2675465B1 (en) 2011-02-14 2020-07-01 MIMEDX Group Inc. Tissue grafts modified with a cross-linking agent and method of making and using the same
AU2012217975B2 (en) * 2011-02-14 2015-11-19 Mimedx Group Inc. Micronized placental tissue compositions and methods for making and using the same
WO2012149486A1 (en) 2011-04-28 2012-11-01 Tissuetech, Inc. Methods of modulating bone remodeling
TWI602570B (en) 2011-06-01 2017-10-21 安瑟吉納西斯公司 Treatment of pain using placental stem cells
CA2837878A1 (en) 2011-06-10 2012-12-13 Tissuetech, Inc. Methods of processing fetal support tissues, fetal support tissue powder products, and uses thereof
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
US20130136773A1 (en) * 2011-09-30 2013-05-30 NuTech Spine, Inc. Expandable Placental Membrane and Methods of Making and Storing Same
EP2775928B1 (en) 2011-11-08 2019-02-20 Auxocell Laboratories Inc. Systems and methods for processing cells
EP2785360B1 (en) 2011-12-02 2019-06-19 MIMEDX Group Inc. Placental tissue grafts produced by chemical dehydration/freeze-drying and methods for making and using same
US9162011B2 (en) 2011-12-19 2015-10-20 Allosource Flowable matrix compositions and methods
EP2793745B1 (en) * 2011-12-22 2019-07-31 MIMEDX Group Inc. Cross-linked dehydrated placental tissue grafts and methods for making and using the same
US8961617B2 (en) 2012-03-08 2015-02-24 Liventa Bioscience, Inc. Amnion and chorion constructs and uses thereof in abdominal surgery
US9814802B2 (en) * 2012-04-30 2017-11-14 The University Of Kansas Method for promoting hair growth comprising implanting a tissue scaffold comprising CK-19 positive cells derived from Wharton's jelly mesenchymal stromal cells
US10851345B2 (en) * 2012-05-08 2020-12-01 Stem Cell Reserve Lp Stem cells and decellularized tissue matrix from cord tissue
US9295753B1 (en) 2012-07-02 2016-03-29 Celso Tello Amniotic membrane preparation and device for use as a lens or as a dressing for promoting healing
US9943551B2 (en) 2012-08-15 2018-04-17 Mimedx Group, Inc. Tissue grafts composed of micronized placental tissue and methods of making and using the same
US8904664B2 (en) * 2012-08-15 2014-12-09 Mimedx Group, Inc. Dehydration device and methods for drying biological materials
US11338063B2 (en) 2012-08-15 2022-05-24 Mimedx Group, Inc. Placental tissue grafts modified with a cross-linking agent and methods of making and using the same
EP2884944B1 (en) 2012-08-15 2020-10-07 MiMedx Group, Inc. Reinforced placental tissue grafts and methods of making and using the same
US20140248328A1 (en) * 2012-08-31 2014-09-04 Jennifer L. Wehmeyer Methods of treating amniotic membranes using supercritical fluids and compositions and apparatuses prepared therefrom
US9180145B2 (en) 2012-10-12 2015-11-10 Mimedx Group, Inc. Compositions and methods for recruiting and localizing stem cells
US8946163B2 (en) 2012-11-19 2015-02-03 Mimedx Group, Inc. Cross-linked collagen comprising metallic anticancer agents
US9827293B2 (en) 2013-01-17 2017-11-28 Mimedx Group, Inc. Non-surgical, localized delivery of compositions for placental growth factors
US10517931B2 (en) 2013-01-17 2019-12-31 Mimedx Group, Inc. Non-surgical, localized delivery of compositions for placental growth factors
US9655948B1 (en) 2013-01-17 2017-05-23 Mimedx Group, Inc. Non-surgical, localized delivery of compositions for placental growth factors
EP2945639B1 (en) 2013-01-18 2020-09-30 MIMEDX Group Inc. Methods for treating cardiac conditions
US10206977B1 (en) 2013-01-18 2019-02-19 Mimedx Group, Inc. Isolated placental stem cell recruiting factors
EP3622960A1 (en) 2013-02-05 2020-03-18 Celularity, Inc. Natural killer cells from placenta
US9427273B2 (en) * 2013-02-20 2016-08-30 BioDlogics, LLC Biologic balloon and method of use
US9498327B1 (en) 2013-03-05 2016-11-22 Biodlogics Llc Repair of tympanic membrane using human birth tissue material
US9855301B1 (en) 2013-03-13 2018-01-02 Biodlogics Llc Human birth tissue laminate and methods of use
WO2014164900A1 (en) 2013-03-13 2014-10-09 Allosource Fascia fibrous compositions and methods for their use and manufacture
US10029030B2 (en) 2013-03-15 2018-07-24 Mimedx Group, Inc. Molded placental tissue compositions and methods of making and using the same
EP2970882B1 (en) 2013-03-15 2018-11-28 AlloSource Cell repopulated collagen matrix for soft tissue repair and regeneration
EP2994147A4 (en) * 2013-05-10 2017-03-08 Cell-Innovations IP Pty Ltd Compositions and methods for the treatment of tinnitus
WO2015031681A1 (en) 2013-08-30 2015-03-05 Mimedx Group, Inc. Micronized placental compositions comprising a chelator
WO2015073381A1 (en) 2013-11-14 2015-05-21 Epic Medical Concepts & Innovations, Inc. Pneumatic somatosensory stimulation device and method
CA2932271C (en) 2013-12-06 2024-02-06 Allosource Methods of drying sheets of tissue
EP3094336A4 (en) 2014-01-17 2018-02-14 MIMEDX Group Inc. Method for inducing angiogenesis
AU2015264025A1 (en) * 2014-05-21 2016-12-01 Mimedx Group, Inc. Micronized Wharton's jelly
TW201603818A (en) 2014-06-03 2016-02-01 組織科技股份有限公司 Compositions and methods
US20160008410A1 (en) 2014-07-08 2016-01-14 Mimedx Group, Inc. Micronized wharton's jelly
USD748462S1 (en) 2014-08-11 2016-02-02 Auxocell Laboratories, Inc. Centrifuge clip
US9993748B2 (en) 2014-08-11 2018-06-12 Auxocell Laboratories, Inc. Centrifuge clip and method
JP7099822B2 (en) 2014-08-28 2022-07-12 ミメディクス グループ インコーポレイテッド Collagen-enhanced tissue graft
US9913466B2 (en) 2014-09-10 2018-03-13 Healthbanks Biotech Co. Ltd. Cryopreservation of umbilical cord tissue strips for cord tissue-derived stem cells
US10765705B2 (en) 2014-11-24 2020-09-08 Prime Merger Sub, Llc Visco-supplement compositions, and methods of use thereof
US20160243288A1 (en) 2015-02-23 2016-08-25 Tissuetech, Inc. Apparatuses and methods for treating ophthalmic diseases and disorders
CA2980316A1 (en) 2015-03-24 2016-09-29 Osiris Therapeutics, Inc. Compositions comprising meniscal tissues and uses thereof
JP2018516869A (en) 2015-05-20 2018-06-28 ティッシュテック,インク. Compositions and methods for preventing epithelial cell proliferation and epithelial-mesenchymal transition
WO2017053701A1 (en) 2015-09-23 2017-03-30 Osiris Therapeutics, Inc. Umbilical tissue compositions and methods of use
TW201733600A (en) 2016-01-29 2017-10-01 帝聖工業公司 Fetal support tissue products and methods of use
TWI586804B (en) * 2016-02-19 2017-06-11 生寶生物科技股份有限公司 Cryopreservation of umbilical cord tissue strips for cord tissue-derived stem cells
EP3474869A4 (en) * 2016-06-24 2020-01-08 Osiris Therapeutics, Inc. Human tissue derived compositions and uses thereof
EP3534981A4 (en) 2016-11-02 2020-06-24 Axogen Corporation Amnion tissue grafts and methods of preparing and using same
US10772986B2 (en) 2017-01-26 2020-09-15 Allosource Fascia fibrous compositions and methods for their use and manufacture
WO2018201116A1 (en) * 2017-04-29 2018-11-01 Aluta Biosciences Llc Umbilical cord implantable cover and method of making
WO2019035925A1 (en) * 2017-08-15 2019-02-21 Predictive Biotech, Inc. Composition and method for treating skin condition
FR3110384A1 (en) 2020-05-21 2021-11-26 Tbf - Genie Tissulaire suburretral band comprising a biological material
CN114099779A (en) * 2020-08-28 2022-03-01 傅毓秀 Waton's gel product capable of promoting bone regeneration
US20230149472A1 (en) * 2021-11-16 2023-05-18 AlphaLogix, LLC Canine-specific therapeutic compositions and methods of use
WO2024006343A1 (en) * 2022-06-29 2024-01-04 Biostem Technologies, Inc. Micronized compositions for wound healing prepared from intact human amnion-chorion tissue having an intact intermediate spongy layer positioned there between

Family Cites Families (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3157524A (en) * 1960-10-25 1964-11-17 Ethicon Inc Preparation of collagen sponge
US3800792A (en) * 1972-04-17 1974-04-02 Johnson & Johnson Laminated collagen film dressing
US4060081A (en) * 1975-07-15 1977-11-29 Massachusetts Institute Of Technology Multilayer membrane useful as synthetic skin
US4240794A (en) * 1979-06-25 1980-12-23 Beisang Arthur A Method of preforming vascular grafts of human and other animal origin
DE2943520C2 (en) * 1979-10-27 1982-05-19 Fa. Carl Freudenberg, 6940 Weinheim Process for the production of collagen sponge for medical or cosmetic purposes
US4361552A (en) * 1980-09-26 1982-11-30 Board Of Regents, The University Of Texas System Wound dressing
US4420339A (en) 1981-03-27 1983-12-13 Kureha Kagaku Kogyo Kabushiki Kaisha Collagen fibers for use in medical treatments
JPS58180162A (en) * 1982-04-19 1983-10-21 株式会社高研 Anti-thrombosis medical material
US4973493A (en) * 1982-09-29 1990-11-27 Bio-Metric Systems, Inc. Method of improving the biocompatibility of solid surfaces
US4599226A (en) * 1983-03-31 1986-07-08 Genetic Laboratories, Inc. Wound dressing comprising silver sulfadiazine incorporated in animal tissue and method of preparation
SU1286211A1 (en) 1983-04-18 1987-01-30 Кишиневский государственный медицинский институт Method of treatment of cicatrices
US4801299A (en) * 1983-06-10 1989-01-31 University Patents, Inc. Body implants of extracellular matrix and means and methods of making and using such implants
US4837285A (en) * 1984-03-27 1989-06-06 Medimatrix Collagen matrix beads for soft tissue repair
FR2563727B3 (en) * 1984-05-04 1987-05-07 Bontemps Raymond NOVEL PROCESS FOR THE PREPARATION OF SKIN GRAFT FROM A NATIVE CONNECTIVE TISSUE
US5230693A (en) * 1985-06-06 1993-07-27 Thomas Jefferson University Implantable prosthetic device for implantation into a human patient having a surface treated with microvascular endothelial cells
US5436135A (en) * 1985-09-02 1995-07-25 Pasteur Merieux Serums Et Vaccins New preparation of placenta collagen, their extraction method and their applications
US4772284A (en) * 1986-03-27 1988-09-20 Collagenix Corporation Breast prosthesis with improved biocompatibility and method of making the same
FR2613620B1 (en) 1987-04-13 1991-04-19 Krom Robert IMPLANT FOR HUMAN MEDICINE
DE3878909T2 (en) 1987-04-28 1993-08-19 Univ California DEVICE AND METHOD FOR PRODUCING A COMPOSITE HOUSING KIT.
US5036056A (en) * 1987-07-08 1991-07-30 Martin Kludas Methods for treating damaged corneal, uterine, or cartilage tissue
GB8803697D0 (en) * 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
EP0331786B1 (en) * 1988-03-11 1994-08-03 CHEMOKOL G.b.R. Ing.-Büro für Kollagenprodukte Method for the preparation of collagen membranes for haemostasis, wound treatment and implants
US5162430A (en) * 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
CA2015495A1 (en) * 1989-05-23 1990-11-23 David C. Muchow Modifying a membrane for use as a graft
US5141747A (en) * 1989-05-23 1992-08-25 Minnesota Mining And Manufacturing Company Denatured collagen membrane
US5116620A (en) * 1989-11-21 1992-05-26 Bruce A. Barber Antimicrobial wound dressing
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
CA2060223C (en) * 1991-02-12 1999-07-20 Clarence C. Lee Injectable medical lubricating fluid composition and method of use
NL9101149A (en) 1991-07-01 1993-02-01 Wilhelmus Hubertus Maria Van E Preparation for medical and/or cosmetic application with placental complex as the active ingredient, method for the production thereof, and method for medical application.
CA2071137A1 (en) * 1991-07-10 1993-01-11 Clarence C. Lee Composition and method for revitalizing scar tissue
US5428022A (en) * 1992-07-29 1995-06-27 Collagen Corporation Composition of low type III content human placental collagen
US5830548A (en) * 1992-08-11 1998-11-03 E. Khashoggi Industries, Llc Articles of manufacture and methods for manufacturing laminate structures including inorganically filled sheets
US5827641A (en) 1992-11-13 1998-10-27 Parenteau; Nancy L. In vitro cornea equivalent model
US6124259A (en) * 1993-01-28 2000-09-26 Celtrix Pharmaceuticals, Inc. Method for treating ophthalmic disorders with IGFBP
IT1263316B (en) * 1993-02-12 1996-08-05 Fidia Advanced Biopolymers Srl MULTILAYER NON WOVEN FABRIC IN WHICH ONE OF THE LAYERS IS ESSENTIALS ESSENTIALS FROM HYALURONIC ACID ESTERS
US5698579A (en) 1993-07-02 1997-12-16 Celgene Corporation Cyclic amides
US5607590A (en) * 1993-08-06 1997-03-04 Shimizu; Yasuhiko Material for medical use and process for preparing same
US5523291A (en) 1993-09-07 1996-06-04 Datascope Investment Corp. Injectable compositions for soft tissue augmentation
GB9400163D0 (en) * 1994-01-06 1994-03-02 Geistlich Soehne Ag Membrane
EP0746268B1 (en) 1994-02-18 2003-10-01 Organogenesis Inc. Bioremodelable collagen graft prosthesis
US5656478A (en) * 1994-02-25 1997-08-12 The Regents Of The University Of California Smooth muscle tissue formation in vivo using cultured smooth muscle cells combined with an extracellular matrix
US5595571A (en) * 1994-04-18 1997-01-21 Hancock Jaffe Laboratories Biological material pre-fixation treatment
US5723010A (en) * 1995-03-31 1998-03-03 Toyo Boseki Kabushiki Kaisha Medical device and method for producing the same
JPH09122227A (en) * 1995-10-31 1997-05-13 Bio Eng Lab:Kk Medical material and manufacture thereof
JPH09122225A (en) * 1995-10-31 1997-05-13 Bio Eng Lab:Kk Raw membrane material for medical material and manufacture thereof
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
EP0907721A1 (en) * 1996-05-28 1999-04-14 Brown University Research Foundation Hyaluronan based biodegradable scaffolds for tissue repair
WO1997048405A1 (en) 1996-06-21 1997-12-24 Jury Evgenievich Belyaev Semi-finished product for producing drug bases, bases obtained using the same and variants, and drugs obtained using these bases and variants
US6281230B1 (en) * 1996-07-24 2001-08-28 Celgene Corporation Isoindolines, method of use, and pharmaceutical compositions
US5635517B1 (en) * 1996-07-24 1999-06-29 Celgene Corp Method of reducing TNFalpha levels with amino substituted 2-(2,6-dioxopiperidin-3-YL)-1-oxo-and 1,3-dioxoisoindolines
EP0925294B3 (en) 1996-07-24 2018-07-04 Celgene Corporation Substituted 2(2,6-dioxopiperidin-3-yl)phthalimides and -1-oxoisoindolines and method of reducing tnf-alpha levels
HU228769B1 (en) 1996-07-24 2013-05-28 Celgene Corp Substituted 2(2,6-dioxopiperidin-3-yl)phthalimides and -1-oxoisoindolines and their use for production of pharmaceutical compositions for mammals to reduce the level of tnf-alpha
US5798368A (en) * 1996-08-22 1998-08-25 Celgene Corporation Tetrasubstituted 2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolines and method of reducing TNFα levels
CN1263731C (en) * 1996-08-12 2006-07-12 赛尔金有限公司 Novel immunotherapeutic agents and their use in production of cytokine levels
US5814328A (en) * 1997-01-13 1998-09-29 Gunasekaran; Subramanian Preparation of collagen using papain and a reducing agent
US6152142A (en) * 1997-02-28 2000-11-28 Tseng; Scheffer C. G. Grafts made from amniotic membrane; methods of separating, preserving, and using such grafts in surgeries
US5993844A (en) * 1997-05-08 1999-11-30 Organogenesis, Inc. Chemical treatment, without detergents or enzymes, of tissue to form an acellular, collagenous matrix
US6379323B1 (en) * 1997-05-09 2002-04-30 Acoustic Technologies, Inc. Bio erodable myringotomy tube
EP1028737B1 (en) * 1997-07-03 2007-04-04 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US6143315A (en) * 1997-07-24 2000-11-07 Wang; Ming X. Biochemical contact lens for treating injured corneal tissue
US5932205A (en) * 1997-07-24 1999-08-03 Wang; Ming X. Biochemical contact lens for treating photoablated corneal tissue
US5955476A (en) * 1997-11-18 1999-09-21 Celgene Corporation Substituted 2-(2,6-dioxo-3-fluoropiperidin-3-yl)-isoindolines and method of reducing inflammatory cytokine levels
US5874448A (en) * 1997-11-18 1999-02-23 Celgene Corporation Substituted 2-(2,6 dioxo-3-fluoropiperidin-3-yl)-isoindolines and method of reducing TNFα levels
US6371992B1 (en) * 1997-12-19 2002-04-16 The Regents Of The University Of California Acellular matrix grafts: preparation and use
US6113932A (en) * 1998-03-02 2000-09-05 Children's Hospital Medical Center Nontoxic vernix compositions and method of producing
JP4695259B2 (en) * 1998-03-16 2011-06-08 セルジーン コーポレイション 2- (2,6-Dioxopiperidin-3-yl) isoindoline derivatives, their preparation and their use as inflammatory cytokine inhibitors
US6432710B1 (en) * 1998-05-22 2002-08-13 Isolagen Technologies, Inc. Compositions for regenerating tissue that has deteriorated, and methods for using such compositions
ES2292245T3 (en) * 1998-05-29 2008-03-01 Osiris Therapeutics, Inc. MESENCHIMATIC MOTHER CELLS CD45 + AND / OR FIBROBLASTO + HUMANAS.
US6110196A (en) 1998-06-17 2000-08-29 Edwards; Stuart D. Apparatus and method for tympanic membrane tightening
US6734018B2 (en) * 1999-06-07 2004-05-11 Lifenet Process for decellularizing soft-tissue engineered medical implants, and decellularized soft-tissue medical implants produced
US6093417A (en) * 1999-01-11 2000-07-25 Advanced Medical Instruments Composition to treat ear disorders
JP2000237300A (en) 1999-02-23 2000-09-05 Djk Research Center:Kk Hollow yarn with fine diameter, its hollow yarn bundle and manufacture thereof
CN1342146A (en) * 1999-03-18 2002-03-27 塞尔基因公司 Substituted 1-oxo-and 1,3-dioxoisoindolines and their use in pharmaceutical compositions for reducing inflammatory cytokine levels
JP2003508127A (en) 1999-08-27 2003-03-04 デパートメント オブ ナショナル ディフェンス Hydrogel wound dressing containing liposome-encapsulated therapeutic agent
US6300315B1 (en) * 1999-08-28 2001-10-09 Ceramedical, Inc. Mineralized collagen membrane and method of making same
DE19964143C2 (en) * 1999-09-11 2001-07-12 Keiper Gmbh & Co Vehicle seat with package position
US6312474B1 (en) * 1999-09-15 2001-11-06 Bio-Vascular, Inc. Resorbable implant materials
US6432712B1 (en) * 1999-11-22 2002-08-13 Bioscience Consultants, Llc Transplantable recellularized and reendothelialized vascular tissue graft
US7004977B2 (en) 1999-11-24 2006-02-28 A Enterprises, Inc. Soft tissue substitute and method of soft tissue reformation
US6428802B1 (en) * 1999-12-29 2002-08-06 Children's Medical Center Corp. Preparing artificial organs by forming polylayers of different cell populations on a substrate
US6376244B1 (en) * 1999-12-29 2002-04-23 Children's Medical Center Corporation Methods and compositions for organ decellularization
US6479064B1 (en) * 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
US6866686B2 (en) * 2000-01-28 2005-03-15 Cryolife, Inc. Tissue graft
JP2003535620A (en) * 2000-02-18 2003-12-02 リジェネレーション テクノロジーズ インク. Transplant tissue implanted with growth factors and other additives
DE60018480T2 (en) 2000-03-09 2006-02-16 Syntacoll Ag MULTILAYER COLLAGEN MATRIX FOR TISSUE CONSTRUCTION
GB2360789A (en) 2000-03-30 2001-10-03 Christopher Mason Method of producing tissue structures
US7713544B2 (en) 2000-07-28 2010-05-11 Emory University Biological component comprising artificial membrane
WO2002014480A2 (en) * 2000-08-16 2002-02-21 Duke University Decellularized tissue engineered constructs and tissues
EP1320390A2 (en) 2000-09-18 2003-06-25 Organogenesis Inc. Bioengineered flat sheet graft prosthesis and its use
US6458810B1 (en) * 2000-11-14 2002-10-01 George Muller Pharmaceutically active isoindoline derivatives
US7311905B2 (en) * 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
IL156303A0 (en) * 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US20030045552A1 (en) 2000-12-27 2003-03-06 Robarge Michael J. Isoindole-imide compounds, compositions, and uses thereof
US7091353B2 (en) * 2000-12-27 2006-08-15 Celgene Corporation Isoindole-imide compounds, compositions, and uses thereof
EP1362095B1 (en) * 2001-02-14 2015-05-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
WO2002067812A2 (en) * 2001-02-23 2002-09-06 University Of Massachusetts Tympanic membrane patch
JP5024694B2 (en) 2001-06-21 2012-09-12 株式会社ビーエムジー Radiation sterilizable medical material and its use
US7067492B2 (en) 2001-09-06 2006-06-27 Omnio Ab Method of promoting healing of a tympanic membrane perforation
AU2002362932B2 (en) * 2001-10-18 2008-06-19 Lifecell Corporation Remodeling of tissues and organs
US20030161818A1 (en) * 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US20030187515A1 (en) * 2002-03-26 2003-10-02 Hariri Robert J. Collagen biofabric and methods of preparing and using the collagen biofabric
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
EP1538913A2 (en) 2002-04-12 2005-06-15 Celgene Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
CN1668733A (en) * 2002-05-30 2005-09-14 细胞基因公司 Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
UA83504C2 (en) * 2003-09-04 2008-07-25 Селджин Корпорейшн Polymorphic forms of 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione
US7775965B2 (en) * 2004-03-09 2010-08-17 The Board Of Regents Of The University Of Oklahoma Decellularized grafts from umbilical cord vessels and process for preparing and using same
US7244759B2 (en) * 2004-07-28 2007-07-17 Celgene Corporation Isoindoline compounds and methods of making and using the same
CN101237898A (en) * 2005-06-10 2008-08-06 细胞基因公司 Human placental collagen compositions, processes for their preparation, methods of their use and kits comprising the compositions
CN101252957A (en) * 2005-06-30 2008-08-27 人类起源公司 Repair of tympanic membrane using placenta derived collagen biofabric
WO2007009062A2 (en) * 2005-07-13 2007-01-18 Anthrogenesis Corporation Treatment of leg ulcers using placenta derived collagen biofabric
WO2007009061A2 (en) * 2005-07-13 2007-01-18 Anthrogenesis Corporation Ocular plug formed from placenta derived collagen biofabric
SG184751A1 (en) * 2005-10-26 2012-10-30 Jan-Eric W Ahlfors Acellular bioabsorbable tissue regeneration matrices produced by incubating acellular blood products
US20080044848A1 (en) * 2006-06-09 2008-02-21 Heidaran Mohammad A Placental niche and use thereof to culture stem cells

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8796315B2 (en) 2009-06-25 2014-08-05 Darlene E. McCord Methods for improved wound closure employing olivamine and human umbilical vein endothelial cells
US9018237B2 (en) 2009-06-25 2015-04-28 Darlene E. McCord Methods for improved wound closure employing olivamine and endothelial cells
US9144555B2 (en) 2012-11-30 2015-09-29 Darlene E. McCord Hydroxytyrosol and oleuropein compositions for induction of DNA damage, cell death and LSD1 inhibition
US9662302B2 (en) 2012-11-30 2017-05-30 Darlene E. McCord Hydroxytyrosol and oleuropein compositions for induction of DNA damage, cell death and LSD1 inhibition
US10231939B2 (en) 2012-11-30 2019-03-19 Darlene E. McCord Hydroxytyrosol and oleuropein compositions for induction of DNA damage, cell death and LSD1 inhibition
US10314688B2 (en) 2016-08-24 2019-06-11 Arthrex, Inc. Tissue use for repair of injury
US10987209B2 (en) 2016-08-24 2021-04-27 Arthrex, Inc. Tissue use for repair of injury
US11918453B2 (en) 2016-08-24 2024-03-05 Arthrex, Inc. Tissue use for repair of injury
US11511017B2 (en) 2019-03-12 2022-11-29 Arthrex, Inc. Ligament reconstruction

Also Published As

Publication number Publication date
WO2008021391A1 (en) 2008-02-21
US8105634B2 (en) 2012-01-31
US20080069895A1 (en) 2008-03-20

Similar Documents

Publication Publication Date Title
US8105634B2 (en) Umbilical cord biomaterial for medical use
US7928280B2 (en) Treatment of leg ulcers using placenta derived collagen biofabric
US20070038298A1 (en) Repair of tympanic membrane using placenta derived collagen biofabric
US20070021762A1 (en) Ocular plug formed from placenta derived collagen biofabric
US20080131522A1 (en) Use of placental biomaterial for ocular surgery
US8071135B2 (en) Placental tissue compositions
KR100999247B1 (en) Collagen biofabric and methods of preparation and use therefor
ES2437865T3 (en) Composition in sheet form
ES2759445T3 (en) Tissue Matrix Forming Procedure
US20120010708A1 (en) Amnion and chorion replacement cover and uses thereof in surgical repair of muscles
US20120035744A1 (en) Amnion and chorion constructs and uses thereof in joint repair
JPWO2006075602A1 (en) Sheet-like composition using amniotic membrane and method for producing the same
US20120035743A1 (en) Amnion and chorion constructs and uses thereof in minimally invasive surgeries
US9480549B2 (en) Multi-layer tissue patches
Gindraux et al. Human amniotic membrane: clinical uses, patents and marketed products
US20130236506A1 (en) Amnion and chorion constructs and uses thereof in ob-gyn surgery
CN108295313B (en) Regenerative repair type lacrimal passage stent and preparation method thereof
US10583219B1 (en) Multilayer bioabsorbable construct and methods of use
RU2269308C1 (en) Method for testicular prosthetics at monorchism and anorchism
Wainwright Cardiac reconstruction with organ specific extracellular matrix

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: HLI CELLULAR THERAPEUTICS, LLC, A DELAWARE LIMITED

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ANTHROGENESIS CORPORATION, A NEW JERSEY CORPORATION, D/B/A CELGENE CELLULAR THERAPEUTICS;REEL/FRAME:039411/0443

Effective date: 20160229

AS Assignment

Owner name: CELULARITY BIOSOURCING, LLC, NEW JERSEY

Free format text: CHANGE OF NAME;ASSIGNOR:HLI CELLULAR THERAPEUTICS, LLC;REEL/FRAME:044707/0351

Effective date: 20170606

AS Assignment

Owner name: CELULARITY, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CELULARITY BIOSOURCING, LLC;REEL/FRAME:044098/0717

Effective date: 20171103