US20180318359A1 - Methods and articles of manufacture for the treatment of animals - Google Patents

Methods and articles of manufacture for the treatment of animals Download PDF

Info

Publication number
US20180318359A1
US20180318359A1 US15/764,275 US201615764275A US2018318359A1 US 20180318359 A1 US20180318359 A1 US 20180318359A1 US 201615764275 A US201615764275 A US 201615764275A US 2018318359 A1 US2018318359 A1 US 2018318359A1
Authority
US
United States
Prior art keywords
amnion
injury
animal
manufacture
particulate mixture
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.)
Pending
Application number
US15/764,275
Inventor
Brandon Ames
Moises Barcelo-Fimbres
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.)
Anicell Biotech LLC
Original Assignee
Anicell Biotech LLC
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 Anicell Biotech LLC filed Critical Anicell Biotech LLC
Priority to US15/764,275 priority Critical patent/US20180318359A1/en
Assigned to ANICELL BIOTECH, LLC reassignment ANICELL BIOTECH, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BARCELO-FIMBRES, Moises, AMES, Brandon
Publication of US20180318359A1 publication Critical patent/US20180318359A1/en
Pending 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
    • 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
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/02Cosmetics or similar toiletry preparations characterised by special physical form
    • A61K8/0212Face masks
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/02Cosmetics or similar toiletry preparations characterised by special physical form
    • A61K8/0241Containing particulates characterized by their shape and/or structure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/98Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution of animal origin
    • A61K8/981Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution of animal origin of mammals or bird
    • A61K8/982Reproductive organs; Embryos, Eggs
    • 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
    • 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
    • A61K9/0048Eye, e.g. artificial tears
    • 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
    • A61K9/0048Eye, e.g. artificial tears
    • A61K9/0051Ocular inserts, ocular implants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • 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
    • A61L15/00Chemical aspects of, or use of materials for, bandages, dressings or absorbent pads
    • A61L15/16Bandages, dressings or absorbent pads for physiological fluids such as urine or blood, e.g. sanitary towels, tampons
    • A61L15/40Bandages, dressings or absorbent pads for physiological fluids such as urine or blood, e.g. sanitary towels, tampons containing ingredients of undetermined constitution or reaction products thereof, e.g. plant or animal extracts
    • 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
    • A61L15/00Chemical aspects of, or use of materials for, bandages, dressings or absorbent pads
    • A61L15/16Bandages, dressings or absorbent pads for physiological fluids such as urine or blood, e.g. sanitary towels, tampons
    • A61L15/42Use of materials characterised by their function or physical properties
    • A61L15/44Medicaments
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/26Mixtures of macromolecular compounds
    • 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
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/41Particular ingredients further characterized by their size
    • A61K2800/412Microsized, i.e. having sizes between 0.1 and 100 microns

Definitions

  • the present disclosure provides processed fetal tissues and cells suitable for promoting healing and methods of using these fetal tissues and cells to promote healing at the site of an injury.
  • Embodiments of the present invention feature methods and articles of manufacture that promote healing in animals.
  • the fetal tissue and cellular compositions disclosed herein have many advantages for use in promoting healing of injuries including immune privilege, an absence of associated ethical issues, and no requirement of invasive procedures for harvesting the cells and tissues.
  • the treatment regimen disclosed herein is remarkably effective at promoting the rapid healing of open wounds within one to two weeks.
  • One embodiment is directed to an article of manufacture.
  • the article comprises a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal.
  • the dried particulate mixture is capable of reconstitution to form a reconstituted medicament for administration to the subject animal to promote healing.
  • a therapeutically effective amount of the reconstituted medicament is applied by injecting at or around the periphery of an internal or external injury, or applied to the subject animal by way of dropper or spray, used as a wash, powder or dressing for wounds, surgically implanted, or the like to promote healing.
  • the treatment may also be applied by way of nebulization or oral.
  • applications to an eye of a subject animal feature drops.
  • one milliliter of reconstituted medicament is the amount of particulate matter obtained from the mechanically decellularized amnion of about 1.5 10 ⁇ 2 cm 3 to 5 10 ⁇ 2 cm 3 of amnion.
  • the mechanically decellularized amnion is filtered to contain particles of less than about 100 microns in diameter.
  • particulate suspension containing particles of less than about 20-100 microns in diameter is well suited for the treatment of the eye injury.
  • Embodiments of the present invention feature a particulate mixture comprising particles.
  • the particles have an approximate diameter of less than 500 ⁇ , or less than 400 ⁇ , or less than 300 ⁇ , or less than 200 ⁇ , or less than 150 ⁇ , or less than 100 ⁇ , or less 90 ⁇ , or less than 80 ⁇ , or less than 70 ⁇ , or less than 60 ⁇ , or less than 50 ⁇ , or less than 40 ⁇ , or less than 30 ⁇ , or less than 20 ⁇ , or less than 10 ⁇ .
  • the particles have a diameter of more than 10 ⁇ , or more than 20 ⁇ , or more than 30 ⁇ , or more than 40 ⁇ , or more than 50 ⁇ , or more than 60 ⁇ , or more than 70 ⁇ , or more than 80 ⁇ , or more than 90 ⁇ , or more than 100 ⁇ , or more than 200 ⁇ , or more than 300 ⁇ , or more than 400 ⁇ , or more than 500 ⁇ .
  • the particles have an approximate diameter of about 500 ⁇ , or about 400 ⁇ , or about 300 ⁇ , or about 200 ⁇ , or about 150 ⁇ , or about 100 ⁇ , or about 50 ⁇ , or about 25 ⁇ .
  • the article comprises a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal, and fetal cells obtained from one or more animals compatible with a subject animal to form a particulate cellular suspension medicament.
  • the particulate cellular suspension medicament is administered to a subject animal to promote healing.
  • a therapeutically effective amount of the particulate cellular suspension medicament can be injected around the periphery of an internal or external injury to promote healing, applied subject animal by way of dropper or spray, used as a wash, powder or dressing for wounds, surgically implanted, or the like.
  • applications to an eye of a subject animal feature drops.
  • a therapeutically effective amount of the particulate cellular suspension medicament comprises about 10 1 to 10 20 cells per mL of medicament. Other embodiments feature about 10 3 to about 10 7 cells per mL.
  • a further embodiment of the article further comprises a fetal tissue wrap.
  • the tissue wrap can comprise amnion tissue obtained from one or more animals compatible with a subject animal.
  • the wrap is constructed and arranged for placement in juxtaposition with a site of injury to promote healing.
  • the wrap comprising fetal tissue
  • one embodiment features a kit for the treatment of a subject animal.
  • One kit comprises a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal that can be capable of reconstitution to form a reconstituted medicament.
  • a further embodiment of the kit comprises fetal cells obtained from one or more animals compatible with a subject animal that are compatible with a particulate mixture obtained from the mechanical decellularization of amnion isolated from one or more animals.
  • the fetal cells are applied in cooperation or concurrently with the reconstituted medicament or form a combined medicament comprising a particulate cellular suspension and/or amniotic liquid medicament for administration to the subject animal to promote healing.
  • a further embodiment of the kit comprises a tissue wrap obtained from one or more animals compatible with a subject animal.
  • the tissue wrap comprises amnion tissue constructed and arranged for placement in juxtaposition with the site of injury.
  • the tissue wrap is applied in cooperation or concurrently with the reconstituted medicament or a combined medicament comprising a particulate cellular or acellular suspension medicament with or without mechanically decellularized amnion and/or amniotic liquid for administration to the subject animal to promote healing.
  • a further embodiment of the present invention is directed to a method of promoting healing in a subject animal.
  • One embodiment of the present method features the steps of administering to the site of an injury at least one of the group consisting of a reconstituted medicament, a reconstituted amnion suspension with or without cells and a tissue wrap all of which have been previously described.
  • a method comprises the step of applying a reconstituted medicament.
  • the reconstituted medicament is made from a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal.
  • the particulate cellular suspension medicament suspension comprises particles derived from the mechanical decellularization of amnion obtained from one or more animals compatible with the subject animal, and isolated amniotic fluid cells obtained from the one or more animals compatible with the subject animal.
  • the method further comprises the step of applying a tissue wrap to the site of the injury.
  • the wrap comprises amnion tissue compatible with the subject animal constructed and arranged for placement in juxtaposition with the site of injury.
  • the amnion tissue is air-dried for about 1 minute to about 48 hours or more. In another embodiment the amnion tissue is air-dried for about 1 hour to about 12 hours. In another embodiment, the amnion tissue is air-dried for about 1 hour to about 6 hours. In another embodiment, the amnion tissue is air-dried for about 1 hour to about 3 hours. In another embodiment, the amnion tissue is air-dried for about 1 hour to about 2 hours.
  • the site of injury is debrided prior to applying one or more of the group consisting of a cellular reconstituted suspension, or a reconstituted acellular suspension medicament and a tissue wrap.
  • a further embodiment is directed to a method of making a dried particulate mixture of mechanically decellularized obtained from one or more animals compatible with a subject animal.
  • the dried particulate mixture is capable of reconstitution to form a reconstituted medicament for administration to the subject animal to promote healing.
  • the method comprises the step of mechanically decellularizing amnion tissue to form particles capable of reconstitution.
  • a further embodiment is directed to a method of making a particulate cellular suspension medicament.
  • the method comprises the steps of providing a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal, and fetal cells obtained from one or more animals compatible with a subject animal to form a particulate cellular suspension medicament and forming a particulate cellular suspension.
  • the particulate cellular suspension medicament is administered to a subject animal to promote healing.
  • a further embodiment of the present invention features methods of making a tissue wrap, a particulate mixture medicament and a particulate cellular suspension medicament.
  • One embodiment of the method of making the tissue wrap comprises the steps of applying amnion tissue to a support to form a supported amnion. The supported amnion is next air dried to form the tissue wrap which is placed in a suitable containment means until applied to a site of injury.
  • FIG. 1 depicts a kit embodying features of the present invention
  • FIG. 2 depicts a tissue wrap embodying features of the present invention.
  • FIG. 3 shows a tissue wrap embodying features of the present invention in a container.
  • isolated cell refers to a cell that has been removed from its in-vivo location.
  • the term “decellularization” refers to a process that removes cells from a tissue while preserving the native ultrastructure and composition of the extracellular matrix (ECM).
  • ECM extracellular matrix
  • an amnion particulate mixture can be obtained by decellularizing a fetal tissue comprising amnion.
  • tissue decellularization There are a number of methods of decellularization of tissue known in the art, including, but not limited to, chemical agents, hypotonic and hypertonic solutions, detergents (e.g., Triton-X), alcohols, solvents (e.g., tributyl phosphate (TBP), biologic agents (e.g., collagenase, trypsin, lipase, nucleases, ⁇ -galactosidase), non-enzymatic agents (e.g., chelating agents such as EDTA or EGTA), physical agents (e.g., temperature, force and pressure, non-thermal irreversible, mechanical, electroporation (NTIRE) (see, for example, Crapo et al., Biomaterials.
  • detergents e.g., Triton-X
  • alcohols e.g., tributyl phosphate (TBP)
  • biologic agents e.g., collagenase, trypsin, lipase
  • one or a combination of the aforementioned methods may be used to decellularize a tissue.
  • methods that preserve the complex composition and three-dimensional ultrastructure of the extracellular matrix (ECM) are preferred.
  • a tissue is mechanically decellularized, e.g., by cryofractionation, a procedure in which a tissue is frozen and ground in a cryomill to produce a mixture of particles.
  • Such particles are obtained from the cryofractionation of about 0.5 cm 2 , or about 1 cm 2 , or about 1.5 cm 2 , or about 2 cm 2 , or about 2.5 cm 2 , or about 3 cm 2 , or about 3.5 cm 2 or about 4 cm 2 , or about 4.5 cm 2 to about 5 cm 2 of amnion or more.
  • the amnion can have a thickness of from about 500 ⁇ to 50 ⁇ , or from 400 ⁇ to about 50 ⁇ , or from about 300 ⁇ to 50 ⁇ , or from about 200 ⁇ to about 50 ⁇ , or from about 150 ⁇ to about 50 ⁇ , from about 100 ⁇ to about 50 ⁇ or from about 50 ⁇ to about 25 ⁇ or less.
  • the amnion has a thickness of about 500 ⁇ , or about 400 ⁇ , or about 300 ⁇ , or about 200 ⁇ , or about 150 ⁇ , or about 100 ⁇ , or about 50 ⁇ , or about 25 ⁇ or less.
  • 1 ⁇ refers to a micrometer or 10 ⁇ 6 meters.
  • amnion refers to a thin, cellular, extra-embryonic membrane that forms the inner membrane of a closed sac surrounding and protecting an embryo in reptiles, birds, and mammals.
  • the sac contains the fetus and amniotic fluid, in which the embryo is immersed, nourished and protected.
  • the amnion is a tough, transparent, nerve-free, and nonvascular membrane consisting of two layers of cells: an inner, single-cell-thick layer of ectodermal epithelium and an outer covering of mesodermal, connective, and specialized smooth muscular tissue.
  • the amnion expands to come in contact with the inner wall of the chorion creating the appearance of a thin wall of the sac extending from the margin of the placenta.
  • the amnion and chorion are closely applied, though not fused, to one another and to the wall of the uterus.
  • the fetal membranes are composed of two principal layers: the outer chorion that is in contact with maternal cells and the inner amnion that is bathed by amniotic fluid.
  • the amnion has multiple functions, e.g., as a covering epithelium, as an active secretary epithelium, and for intense intercellular and transcellular transport.
  • tissue refers to an aggregate of similar cells and associated extracellular matrix (ECM) forming a definite kind of organized material with a specific function, in a multicellular organism.
  • ECM extracellular matrix
  • an “amnion tissue” refers to the isolated cellular, extra-embryonic amnion membrane that is detached from the chorion.
  • the amnion tissue is air-dried.
  • the amnion is air-dried for about 60 to about 90 minutes or more at ambient temperature (i.e. about 18 to 24° C.).
  • a “particulate mixture” refers to the powder or particles obtained from the cryofractionation of amnion.
  • fetal tissue refers to extra-embryonic tissues including, but not limited to, amnion, chorion, yolk sac, the allantois, umbilical cord and/or fetal placenta (villous chorion).
  • fetal cells refers to cells resident in the extra-embryonic tissues including, but not limited to, amnion, chorion, yolk sac, the allantois, umbilical cord, fetal placenta (villous chorion) and/or amniotic fluid.
  • fetal cells refer to isolated fetal cells.
  • fetal cells refers to unfractionated cells of the amniotic fluid including epithelial and/or amniotic fluid or membrane-derived mesenchymal stem cells (see U.S. Patent Publication No. US 2013/0230924, which is incorporated by reference herein in its entirety).
  • injury means a pathological condition, such as, by way of example, without limitation, a wound, incision, a break in the skin, bone, tendon, ligament, muscle, neoplasia, eye, and soft tissues, an inflammation, infection, or other disease condition.
  • the term “promoting healing” refers to causing a favorable result compared to no treatment.
  • the favorable result comprises any one or more of the following such as reduction of scarring, reduction of inflammation, regrowth of normal tissue or growth of scar tissue, improved load bearing on a limb movement, closure of wound, reduction in infection and reduction in mortality associated with the underlying pathology.
  • compatible with a subject animal denotes the origin of the tissue as being from the same species or closely related species.
  • the term “compatible with a subject animal” refers to an xenograft, i.e., a tissue graft from different species.
  • the term “compatible with a subject animal” refers to allografts, i.e., a tissue from one individual to another of the same species with a different genotype.
  • an “animal” refers to living multi-cellular vertebrate organisms, a category that includes, for example, mammals, birds, reptiles, and amphibians.
  • the term mammal includes both human and non-human mammals.
  • the term “subject animal” includes both human and non-human subjects.
  • the term “subject animal” refers to an individual human or animal which has a site of injury or disease.
  • a non-human animal can refer to a mammal including, but not limited to, a domesticated animal such as a dog, a racing dog, sheep, a pig, a goat, cattle, a zebu, a cat, a guinea pig, a donkey, water buffalo, including “river buffalo” and “swamp buffalo”, a horse, a racing horse, a dromedary camel, a yak, a bactrian camel, a llama, an alpaca, a ferret, a mouse, a bali cattle, a gayal, a rabbit, a rat and a lab rat, a silver fox or a hedgehog.
  • a domesticated animal such as a dog, a racing dog, sheep, a pig, a goat, cattle, a zebu, a cat, a guinea pig, a donkey, water buffalo, including “river buffalo” and “swamp buffalo”,
  • a non-human animal can refer to mammals kept in zoos including, but not limited to, zebra, gazelle, wolves, wild swine (pigs & hogs), wild cattle, warthogs, vervet monkeys, two-toed sloths, tree pangolins, tigers, tapirs, tamandua or lesser anteaters, takins, sun bears, striped hyena, spotted hyena, spiral-horned antelope, somali wild ass, snow leopards, small cats, sloth bears, singing dogs, siamang, serval, sea lions, rock hyrax, rhinoceros, reindeer, red pandas, pygmy marmosets, pygmy hippopotamus, gravwalski's horses, pronghorns, prairie dogs, porcupines, polar bears, painted dogs, otters, oryx, orangutan,
  • a non-human animal can refer to mammals considered by the World Wildlife Fund to be endangered including, but not limited to, the amur leopard, black rhino, cross river gorilla, javan rhino, mountain gorilla, pangolin, saola, south china tiger, sumatran elephant, sumatran orangutan, sumatran rhino, sumatran tiger, vaquita, western lowland gorilla, yangtze finless porpoise, african wild dog, amur tiger, asian elephant, bengal tiger, black spider monkey, black-footed ferret, blue whale, bonobo, bornean orangutan, borneo pygmy elephant, chimpanzee, eastern lowland gorilla, fin whale, ganges river dolphin, giant panda, hector's dolphin, indian elephant, indochinese tiger, indus river dolphin, malayan tiger, north atlantic right whale, orangutan, sea lions
  • a non-human animal can refer to marsupials, including, but not limited to, wallabies, koalas, possums, opossums, kangaroos, bandicoots, wombats, bettongs, bilbys, quolls, quokkas and the Kenyan devil.
  • a liquid for reconstitution comprises a biocompatible solution such as normal saline, e.g. phosphate buffered saline (PBS) or amniotic fluid.
  • a preferred liquid for reconstitution is calcium-free sterile, non-pyrogenic isotonic solution suitable for intravenous administration.
  • one such liquid is sold under the trademark PlasmaLyte ATM in a single dose container for intravenous administration.
  • Each 100 mL contains 526 mg of Sodium Chloride, USP (NaCl); 502 mg of Sodium Gluconate (C 6 H 11 NaO 7 ); 368 mg of Sodium Acetate Trihydrate, USP (C 2 H 3 NaO 2 .3H 2 O); 37 mg of Potassium Chloride, USP (KCl); and 30 mg of Magnesium Chloride, USP (MgCl 2 .6H 2 O). It contains no antimicrobial agents. The pH is 7.4.
  • administering means applying or injecting or ingesting the medicament.
  • applying is used broadly and includes uses such as washes and implantation.
  • ECM extracellular matrix
  • Collagen is the most abundant structural protein in the connective tissue (ECM) and acts as a natural scaffold for cellular attachment in the body.
  • Amnion is an abundant source of collagen, as well as the other proteins, carbohydrates, lipids, hyaluronic acid, laminin, fibronectin, pluripotent mesenchymal stem cells (MSC) and other complex growth factors that are essential for fetal growth and development.
  • amnion has a complete lack of surface antigens, thus it does not induce an immune response when implanted into a ‘foreign’ body, which is in contrast to most other allograft implants.
  • Amnion also markedly suppresses the expression of the pro-inflammatory cytokines, IL-1 ⁇ and IL-1 ⁇ (Solomon et al., 2001, Br. J. Ophthalmol.
  • MMPs matrix metalloproteases
  • Amnion derived tissues are therefore immune-privileged and ideally suited for regenerative medicine applications.
  • Kit is an assembly of parts, materials, and compositions of matter packaged together to facilitate a treatment. Kits commonly comprise instructions for the use of the parts, materials and compositions.
  • Kit 11 has the following major elements: a first vial 15 , a second vial 17 , a container for a tissue wrap 21 , a syringe 23 , and instructions 25 .
  • the kit 11 is held in suitable packaging, as depicted, a box 27 .
  • suitable packaging may comprise any means for holding the collection of parts, materials and compositions.
  • the kit 11 for ocular treatments may comprise fibrin glue [not shown] for attaching the tissue wrap 23 to the eye.
  • the first vial 15 contains a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal that upon of reconstitution forms a reconstituted medicament.
  • the kit 11 may contain a vial containing such liquid for reconstitution [not shown] or the liquid for reconstitution may be derived from other sources.
  • the second vial contains fetal cells obtained from one or more animals compatible with a subject animal and compatible with a particulate mixture in the first vial 15 .
  • the fetal cells are applied in cooperation or concurrently with the reconstituted medicament or form a combined medicament comprising a particulate cellular suspension medicament for administration to the subject animal to promote healing.
  • the dried particulate mixture of the first vial 15 is reconstituted with or combined with the fetal cells of the second vial 17 , supplemented as needed with further liquid for reconstitution.
  • the dried particulate mixture can be reconstituted by suspension in a solution of 50% solution for reconstitution, such as PlasmaLyte ATM, and 50% amniotic fluid containing fetal cells.
  • the combined medicament is injected into or around the site of injury with syringe 23 .
  • the container for a tissue wrap 21 contains a tissue wrap derived from amnion tissue obtained from one or more animals compatible with a subject animal.
  • the tissue wrap comprises amnion tissue constructed and arranged for placement in juxtaposition with the site of injury.
  • the tissue wrap is applied in cooperation or concurrently with the reconstituted medicament or a combined medicament comprising a particulate cellular suspension medicament for administration to the subject animal to promote healing.
  • tissue wrap 31 is formed by affixing amnion tissue to a first support 33 on one side of the amnion and a second support on the other side of the amnion [not shown] and air drying the tissue for thirty minutes to three hours or more, and, most preferably, for about one hour.
  • the supports, of which first support 33 is depicted maintain the shape of the tissue during the drying process. The supports are preferably removed prior to placement of the tissue in container 21 .
  • Tissue wrap 31 is removed from the container and placed over the site of injury.
  • the dried particulate mixture is obtained from the mechanical decellularization or cryofractionation of about 1.5 10 ⁇ 2 cm 3 to 5 10 ⁇ 2 cm 3 of amnion/mL of reconstituted medicament and include particles greater than 20-100 microns in diameter.
  • the reconstituted medicament can be administered to the subject animal to promote the healing of superficial wounds.
  • a reconstituted medicament containing particles less than about 100 microns is suitable for treatment of eye injuries.
  • the kit 11 for the treatment of eye injuries comprises a dropper for administration.
  • the dried particulate mixture obtained from the cryofractionation comprises about 1.5 ⁇ 10 ⁇ 2 cm 3 to about 5 ⁇ 10 ⁇ 2 cm 3 of amnion/mL of reconstituted medicament.
  • the fetal cells can comprise amniotic fluid cells and the particulate matter can be filtered to contain particles that are less than 100 microns in diameter.
  • the fetal cells can have a concentration from 10 3 to 10 20 /mL mesyschimal and/or epithelial stem cells.
  • fetal cells can have a concentration of 10 3 to 10 12 /mL.
  • fetal cells can have a concentration of 10 4 to 10 12 /mL.
  • fetal cells can have a concentration of 10 4 to 10 11 /mL.
  • fetal cells can have a concentration of 10 4 to 10 10 /mL.
  • fetal cells can have a concentration of 10 4 to 10 9 /mL.
  • fetal cells can have a concentration of 10 4 to 10 8 /mL. In another embodiment, fetal cells can have a concentration of 10 4 to 10 7 /mL. In another embodiment, fetal cells can have a concentration of 10 4 to 10 6 /mL. In another embodiment, fetal cells can have a concentration of 10 3 to 10 6 /mL. In another embodiment, fetal cells can have a concentration of 10 3 to 10 7 /mL.
  • the fetal cells can have a concentration of about 0.8 ⁇ 10 6 to 1.2 ⁇ 10 6 cells/mL of the particulate cellular suspension.
  • the dried particulate mixture can contain particles obtained from the cryofractionation of from about 1.5 ⁇ 10 ⁇ 2 cm 3 to about 5 ⁇ 10 ⁇ 2 cm 3 amnion per mL of the particulate cellular suspension.
  • the particulate cellular suspension comprises a particulate matter obtained from the cryofractionation about 1.5 ⁇ 10 ⁇ 2 cm 3 to about 5 ⁇ 10 ⁇ 2 cm 3 amnion for every 10 6 plus or minus 2 ⁇ 10 5 amnion fluid cells.
  • Embodiments of the present invention are well suited for the treatment of open wounds, superficial and deep including such wounds that expose a movable body element of the animal's musculoskeletal system, for example, a ligament or tendon.
  • Such wounds can include ligaments and tendons that are lacerated.
  • the injury may comprise chondral and fascial pathologies, tendon and ligament pathologies, surface reconstruction or articular joint pathologies.
  • the chondral and fascial pathologies can include, for example, laminitis.
  • the tendon and ligament pathologies can include, for example, tendonitis, sport-induced superficial digital flexor tendon (SDFT) or suspensory ligament (SL) lesions.
  • the articular joint pathologies can include, for example, navicular disease, ringbone or arthritis.
  • the site of injury can also be an open wound that, in one example, is able to heal in about one to two weeks. In another example, healing of the site of injury reduces the risk of recurrence. In one example, the risk of recurrence of the injury can be reduced by about 5 fold.
  • This procedure defines the aseptic collection of amniotic material (amnion and amniotic fluid) for injection at the site of an injury.
  • the amnion container was picked up and sampled for Bioburden.
  • the amnion was aseptically transferred into the sterile field (laminar flow hood).
  • the amnion transport packaging previously disinfected, i.e. with 70% ETOH was opened.
  • a 50 mL sample of the Amnion Transport Solution was aseptically transferred into a 30 to 60 mL conical tube for pre-processing bioburden testing.
  • the vial was labeled with sample description, batch number, date and time and placed in a designated refrigerator.
  • the amnion from the incoming container was transferred into approximately 200 mL of Plasma Lyte-A in a sterile bioassay dish where it was gently rinsed. A piece of amnion was then spread evenly on a sterile cutting board carefully avoiding any overlaps. A record was made of the amnion preparation start time. Sterile gauze or laps were used to remove any remaining debris/blood from the surface of the amnion. The amnion was inverted and the surface of the opposite site was similarly washed. Any chorion was removed by blunt dissection to separate it from the amnion. After washing and cleaning, the amnion pieces were returned to the bioassay dish containing Plasma Lyte-A. Using a sterile scissors/scalpel, the amnion was cut into 2 to 10 sections. The approximate area of each piece (50-450 cm 2 ) was measured and recorded using a sterile stainless steel ruler.
  • the cleaned pieces of the amnion were placed back on the sterile cutting board and the amnion was spread out on the board taking care to not overlap.
  • a sterile nylon mesh was placed over the surface of the amnion again taking care to overlap them.
  • the amnion and mesh were then placed onto a sterile drying rack and allowed to air dry for a minimum of one hour. Start and stop times for drying were recorded.
  • a large sterile pan was first placed into the sterile field (laminar flow hood) and filled with cold packs from a ⁇ 80° C. freezer.
  • the aspiration containers with the amniotic fluid were disinfected with 70% ethanol (ETOH), inspected for integrity and placed on the cold packs in the laminar flow hood.
  • the source of the amniotic fluid was confirmed by looking at the Donor animal ID number.
  • the 2.0 mL of amniotic fluid was then aseptically pipeted into a 2 mL sterile microcentrifuge tube for bioburden testing.
  • the vial was labeled with the sample number, batch number, date and time and placed in the designated refrigerator.
  • Each amniotic fluid bottle was gently mixed and 1.0 mL of the fluid was collected using a 1 mL micropipette and transferred into a 2 mL Eppendorf microcentrifuge tube. A total of 50 ⁇ L of cell suspension was then added to 50 ⁇ L of trypan blue (0.4%) in an Eppendorf microcentrifuge tube and vortexed for 5 seconds. The sample was placed on a rack for 5 minutes.
  • a Neubauer chamber (hemocytometer) was rinsed with distilled water, and then sprayed with ETOH 70% and wiped clean and dried with paper towels. A cover slip was placed on the top of the micro-grids of the chamber. The trypan blue-cell suspension was gently mixed and used to fill both sides of the hemocytometer with 10 ⁇ L by capillary action. The cells were allowed to settle down for at least 30 seconds. The hemocytometer was placed under the microscope and all cells in the four 1 mm corner squares and one 1 mm center square were counted. For accuracy the total number of cells counted was greater than 100.
  • the cells were re-counted if >10% of the cells appeared clustered, by vigorously pipetting in the original cell suspension as well as in the trypan blue cell suspension mixture. Using a double cell counter, the number of viable and non-viable cells was determined. The cells in both chambers were counted and an average was calculated. For the trypan blue test, live cells did not take up the dye, whereas dead (non-viable) cells did. Thus non-viable cells stained blue and viable cells remained opaque. “Ghost” cells, which appeared as flattened pale blue cells were not counted.
  • the amnion was removed from the drying rack and transferred into the milling chambers having an impactor.
  • the milling chambers were placed into the Cryomill and cryofractionated using the following settings:
  • the milling chambers were allowed to warm to room temperature for approximately two hours. The start and stop times were recorded.
  • the amniotic fluid was aliquoted evenly into 50 mL sterile centrifuge tubes and centrifuged at 200-400 ⁇ g (1100-1600 rpm) for approximately 5-10 minutes at room temperature. The supernatant was then removed from each tube using a 25 mL sterile serological pipette. The amniotic liquid was kept in new container and the pellet was re-suspended in Plasma Lyte-ATM to a total volume of 25 mL in each tube. The re-suspended cells in any two different tubes were vortexed for approximately 3 to 5 sec and consolidated into a single tube prior to centrifugation at 200-400 ⁇ g (1100-1600 rpm) for approximately 5-10 minutes at ambient temperature. The preceding steps were repeated as necessary.
  • the supernatant from each tube was removed using a sterile pipette and the pellet was again re-suspended in a cell suspension solution (amniotic fluid and/or an isotononic solution, e.g., PlasmaLyte ATM) to bring the volume in each tube to about 10 mL and vortexed for approximately 3 to 5 seconds.
  • a cell suspension solution amniotic fluid and/or an isotononic solution, e.g., PlasmaLyte ATM
  • a 1 mL aliquot was removed and the cell count and viability was determined using the above-described trypan blue test.
  • red blood cells were present in the amniotic fluid cell suspension, they were removed using a RBC Lysing Solution.
  • amniotic fluid cells were then filtered through a 100 ⁇ m cell strainer, and re-suspended in PBS or PlasmaLyte ATM. The cells were again centrifuged for 10 minutes at 250 to 400 ⁇ g. The supernatant was decanted and the pellet was left in the 50 mL conical centrifuge tube.
  • cryovials that were previously labeled and their corresponding size of CoolCellTM freezer (CCF) racks were placed in the hood.
  • Cryostor 10TM is commercially available from Biolife Solutions.
  • the cell suspension solution and the amnion suspension solution were then combined into the cell suspension solution container. Using 50 mL serological pipettes, the solutions were homogenized several times. The container was again placed on the cold packs on the sterile field. Empty cryovials were placed in the CCF racks on cold packs and their caps were removed inside the hood (sterile field). The mix of cell/amnion suspension solution was pipeted into an empty Amnion Suspension container and a same volume of CryoStor 10TM was added and homogenized before being placed on the cold packs. A 50 mL pipette Combitip was fitted on to a repeat pipetor set to dispensing mode.
  • the fill volume was adjusted and 50 mL of the Cell/Amnion solution was aspirated and then adjusted to the desired dispense volume (1 or 2 mL).
  • the cryovials in the CCF were then filled and the vial caps were replaced securely and the rack of filled vials was placed on cold packs for QC inspection.
  • Cell freezing was achieved by cooling the cells at a cooling rate of 1° C. per minute from 4° C. to ⁇ 80° C., using a passive cooling controlled-rate freezer CoolCellTM (commercially available from Biocision.)
  • the cell suspension in cryoprotective freezing medium was aliquoted into each of the cryovials and the cells were gently mixed to maintain a homogeneous cell suspension.
  • the solid core of the CoolCell (black ring) at room temperature was seated in the bottom of the central cavity and the vials containing the cell suspension were placed in each well.
  • the lid of the CoolCellTM was fully sealed and the CoolcellTM was placed into a ⁇ 80° C. freezer for at least 4 hours prior to transfer on dry ice to long term storage. Cell viability and QA/QC were evaluated by thawing one vial after short term storage.
  • the amnion transport packaging previously disinfected, i.e. with 70% ethanol, methanol, etc.
  • the sterile field a laminar flow hood.
  • a sample of the Amnion Transport Solution was first transferred into a 50 mL conical tube for Bioburden testing. The vial was then labeled with sample description, batch number, date and time and placed in designated refrigerator.
  • Saline was aseptically added into a second receiving pan in the sterile field (i.e. laminar flow hood) and the amnion tissue was taken from incoming receiving pan to the second receiving pan containing the sterile saline. Any remaining blood was rinsed with sterile saline. After documenting the amnion preparation start time, sterile gauze or laps was used to remove any remaining debris/blood from the surface of the amnion. The amnion was then inverted and the other side was rinsed and washed. Any remaining chorion was removed by blunt dissection to separate it from the amnion. The amnion was kept wet with sterile saline.
  • the tissue was blocked off by cutting away any stringy ends and checked for holes or tears. After repositioning the amnion on the cutting board with the chorion side up, the approximate area of each piece of amnion was measured and recorded in cm 2 using a sterile stainless steel ruler.
  • the amnion (chorion side up) was covered with a sterile mesh (e.g., a nylon mesh) wetted with sterile saline.
  • a sterile mesh e.g., a nylon mesh
  • first steel mesh which is denoted by a first visible indicia, the color white
  • second steel mesh to be discussed below, which is denoted by a second visible indicia, the color blue. Any reference to the colors white or blue are directed to these visible indicia. Those skilled in the art will recognize that such visible indicia is matter of choice.
  • the mesh was cut to size and was allowed to slightly overlap the amnion. The mesh-covered amnion was then gently lifted, turned over and placed back on the cutting board.
  • Caution was used as to not disturb the mesh/amnion interface.
  • the newly exposed side of the amnion was wiped with sterile wipes or gauze pads to remove any remaining blood or small tissue particles.
  • the newly exposed side of the amnion was covered with a second steel mesh, BLUE sterile mesh (e.g., a nylon mesh) wetted with sterile saline. The mesh was cut to size and was allowed to slightly overlap the amnion.
  • BLUE sterile mesh e.g., a nylon mesh
  • the amnion tissue sandwiched between the white and blue mesh was placed on the drying rack where the amnion tissue was allowed to dry for 60 to 90 minutes at ambient temperature (65° to 70° F.). Caution was used as to not disturb the mesh/amnion interface. The amnion was kept unfolded and as flat as possible during this step. Additional drying racks were used as needed. The total drying time was recorded. The dried tissue was then removed from the rack(s) and laid flat on the cutting board, WHITE side up. The WHITE mesh was carefully removed from the entire sheet of amnion which was checked for holes or tears.
  • each section was cut using a scalpel or rotary cutting blade and the sizes and surface areas were recorded as 5 ⁇ 5 cm, 10 ⁇ 10 cm, round 15 mm diameter and round 22 mm diameter.
  • the sections can be cut and sized to fit particular needs and these sizes and shapes are only exemplary.
  • Pouches were transferred onto the sterile field (previously disinfected laminar flow hood). Using sterile forceps, each individual tissue membrane was inserted into the inner pouch. Large membranes were folded if necessary. The pouches were sealed with the dried amnion tissue on a mesh (see, for example, FIGS. 2 and 3 ) and inspected for a broken seal, impurities, and defects. Upon passing the inspection, the pouches were labeled with date and packaged in large pouches according to size and stored in the refrigerator or at room temperature. The donor ID, size, date, time, and initials were documented.
  • a container was 3 ⁇ 4 filled with hot tap water with a thermometer and cold water was added until a temperature of 37° C. was reached.
  • a vial of cryofractionated amnion with amniotic fluid cells was taken from the ⁇ 80° C. freezer. Holding the cap, the vial was partial immersed in the water bath for approximately 2 to 3 minutes with gentle agitation until the contents were melted. The vial was removed from the water bath and the exterior was wiped with sterile gauze saturated with 70% ethanol. The thawed contents were then ready for immediate use.
  • a horse with an injury was adequately restrained and sedated or anaesthetized before starting the procedure according to the veterinarian's directions.
  • Appropriate sedation or anesthesia caused minimal distress to the patient and prevented any long lasting discomfort.
  • Use of nose twitch was optionally required. In the case of young adults or unbroken horses, short-acting general anesthesia was required.
  • the site for the product implantation was processed as a surgically prepared area. After cleaning or clipping of any gross contamination, such as dust or mud, the complete the area was scrubbed with chlorhexidine for 5 to 7 minutes and then wiped down with alcohol swabs. Before proceeding, the area was cleaned until the alcohol swabs used on the scrubbed area were dirt-free.
  • Systemic antibiotics were administered prior to starting the procedure. However, in some instances, systemic antibiotics are not necessary.
  • the contents of the 2 mL vial were split into 3 to 4 doses (0.50 to 0.67 mL) and loaded into syringes (for example, 1 mL syringes) with sterile hypodermic needles (for example 22 gauge, 1.5 inch needles).
  • syringes for example, 1 mL syringes
  • sterile hypodermic needles for example 22 gauge, 1.5 inch needles.
  • the appropriate leg was resting or held up and restrained as needed.
  • the cryofractionated amnion and amniotic fluid cell mix was injected in close proximity to the lesion without penetrating the actual tendon, ligament or muscle.
  • the cellular particulate mixture was injected either peri-articularly or intra-articularly. After injection, the site was bandaged with sterile swabs and adhesive bandage.
  • Example 5 Implantation of Cryofractionated Amnion and Isolated Amniotic Fluid Cells in Combination with the Amnion Tissue Wrap
  • a kit comprising sterile hypodermic needles (18 and 23 gauge), syringes (3 to 5 mL), vials of thawed cryofractionated amnion and amniotic fetal cells as well as packaged amnion tissue wrap of the appropriate size were assembled (see FIG. 1 ) together with clippers, sterile gloves, 1% lidocaine, saline and sedatives and/or anesthetics.
  • a horse was treated at a veterinary clinic a severe leg severe injury caused by barbed wire.
  • the wound was initially pre-cleaned to remove dirt, scrubbed with chlorhexidine for 5 to 7 minutes and then wiped down with alcohol swabs.
  • the veterinarian aggressively debrided the wound. This step created a relatively clean wound bed and generated an inflammatory signal that induced the migration and proliferation of stem cells and growth factors (from the amniotic material and horse's own immune system). Systemic antibiotics were administered prior to starting the procedure.
  • the veterinarian determined the amount of amniotic material needed to treat the wound by measuring the maximal length, width and depth of the wound. Amniotic tissue wrap of the appropriate size was selected and applied on the wound. Treatment of an open wound with Amniotic tissue wrap and a cellular suspension of cryofractionated amnion and isolated amnion fluid cells.
  • Amount of cryofractionated amnion and isolated amnion fluid cells to be injected as a function of wound volume Amount of cryofractionated amnion/ Wound Area amnion fluid cell suspension to use ⁇ 12.5 cm 2 1.0 mL >12.5 cm 2 but ⁇ 25 cm 2 1.5 mL >25 cm 2 2.0 mL
  • cryofractionated amnion/amnion fluid cell suspension was injected into the pen-wound skin that appeared normal in texture, turgor, and free of infection using a 23-gauge needle on a 3 mL syringe.
  • the cell suspension was injected at a distance of approximately 5 mm from the wound edges and 10 to 15 mm deep at the 3-, 6-, and 9- and 12-o'clock positions (directly into the superficial fascia and the subcutaneous tissue of the wound), aiming the needle parallel to the wound margin at each location.
  • the cryofractionated amnion/amnion fluid cell suspension was mixed with 1% plain lidocaine in a 1:1 ratio. After the completion of the procedure, the surgical site was dressed with a nonporous dressing followed by application of a nonstick dressing and a dry sterile dressing of gauze.
  • the wound was assessed and showed striking improvement. Generally, treatment shows 2.5 times the rate of healing.
  • Corneal ulcers are the most common eye problem in horses.
  • a veterinarian located the foreign material and determined the wound severity (size of the corneal lesion) using visual inspection or fluorescein dye to highlight any subtle corneal defect or scratch.
  • the area around the eye was cleaned to remove debris or excessive liquid using sterile gauze.
  • amniotic tissue wrap ie. 15 or 22 mm diameter discs
  • the dehydrated amniotic tissue wrap was positioned over the tip of the index finger and applied directly to the eye surface while opening the eye lids with the other hand (if needed).
  • the membrane attached to the eye surface by hydrostatic tension may be used to assist to insure placement in challenging environments.
  • Example 7 Treatment of Eye Injuries with a Suspension of Cryofractionated Amnion
  • the eye surface and surrounding area was prepared as described in Example 6.
  • Eye drops were given either alone or in conjunction with the amniotic tissue wrap application described in Example 6.
  • Vials of cryofractionated amnion suspended in saline ie. PlasmaLyte ATM and/or Amniotic Fluid
  • saline ie. PlasmaLyte ATM and/or Amniotic Fluid
  • the vial was gently rotated upside down to mix solution.
  • 2 drops of the suspension were applied to the injured eye twice a day (4 drops total per day) over the membrane or ocular surface. Treatment lasted for a week or until the suspension was used up.
  • the suspension was pH balanced to prevent any irritation to the eye.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Dermatology (AREA)
  • Reproductive Health (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Transplantation (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Botany (AREA)
  • Birds (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Materials Engineering (AREA)
  • Pregnancy & Childbirth (AREA)
  • Gynecology & Obstetrics (AREA)
  • Gerontology & Geriatric Medicine (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Cosmetics (AREA)

Abstract

Methods are disclosed for the collection and processing of amniotic material in animals. These methods involve collection of amniotic material directly during parturition or caesarian section in animals for the processing of regenerative wound treatments and tissue repairs without culturing or utilization of any excess manipulation of tissue.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application 62/234,354, filed Sep. 29, 2015, entitled “Methods and Articles of Manufacture for the Treatment of Animals, the contents of which are incorporated herein by reference in their entirety.
  • STATEMENT REGARDING FEDERAL SPONSORSHIP
  • Inventions described herein were not conceived or reduced to practice with Federal sponsorship.
  • FIELD OF THE INVENTION
  • The present disclosure provides processed fetal tissues and cells suitable for promoting healing and methods of using these fetal tissues and cells to promote healing at the site of an injury.
  • BACKGROUND OF THE INVENTION
  • Wounds and injuries in animals and humans can sometimes be difficult to treat. Failure to adequately treat and promote healing can place the animal or individual in jeopardy with potential loss of function in an effected limb or amputation, or even loss of life.
  • It would be useful to have methods and articles of manufacture that promote healing in animals and humans. As used herein, unless the context requires otherwise, the term “animal: encompasses and includes humans.
  • SUMMARY OF THE INVENTION
  • Embodiments of the present invention feature methods and articles of manufacture that promote healing in animals.
  • The fetal tissue and cellular compositions disclosed herein have many advantages for use in promoting healing of injuries including immune privilege, an absence of associated ethical issues, and no requirement of invasive procedures for harvesting the cells and tissues. In addition, the treatment regimen disclosed herein is remarkably effective at promoting the rapid healing of open wounds within one to two weeks.
  • One embodiment is directed to an article of manufacture. The article comprises a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal. The dried particulate mixture is capable of reconstitution to form a reconstituted medicament for administration to the subject animal to promote healing.
  • By way of example, without limitation, a therapeutically effective amount of the reconstituted medicament is applied by injecting at or around the periphery of an internal or external injury, or applied to the subject animal by way of dropper or spray, used as a wash, powder or dressing for wounds, surgically implanted, or the like to promote healing. The treatment may also be applied by way of nebulization or oral. For example, without limitation, applications to an eye of a subject animal feature drops.
  • In one aspect, one milliliter of reconstituted medicament is the amount of particulate matter obtained from the mechanically decellularized amnion of about 1.5 10−2 cm3 to 5 10−2 cm3 of amnion. In one aspect, the mechanically decellularized amnion is filtered to contain particles of less than about 100 microns in diameter. In one aspect, particulate suspension containing particles of less than about 20-100 microns in diameter is well suited for the treatment of the eye injury.
  • Embodiments of the present invention feature a particulate mixture comprising particles. The particles have an approximate diameter of less than 500μ, or less than 400μ, or less than 300μ, or less than 200μ, or less than 150μ, or less than 100μ, or less 90μ, or less than 80μ, or less than 70μ, or less than 60μ, or less than 50μ, or less than 40μ, or less than 30μ, or less than 20μ, or less than 10μ. In other embodiments, the particles have a diameter of more than 10μ, or more than 20μ, or more than 30μ, or more than 40μ, or more than 50μ, or more than 60μ, or more than 70μ, or more than 80μ, or more than 90μ, or more than 100μ, or more than 200μ, or more than 300μ, or more than 400μ, or more than 500μ. In other embodiments, the particles have an approximate diameter of about 500μ, or about 400μ, or about 300μ, or about 200μ, or about 150μ, or about 100μ, or about 50μ, or about 25μ.
  • In certain embodiments, the article comprises a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal, and fetal cells obtained from one or more animals compatible with a subject animal to form a particulate cellular suspension medicament. The particulate cellular suspension medicament is administered to a subject animal to promote healing.
  • By way of example, without limitation, a therapeutically effective amount of the particulate cellular suspension medicament can be injected around the periphery of an internal or external injury to promote healing, applied subject animal by way of dropper or spray, used as a wash, powder or dressing for wounds, surgically implanted, or the like. For example, without limitation, applications to an eye of a subject animal feature drops.
  • A therapeutically effective amount of the particulate cellular suspension medicament comprises about 101 to 1020 cells per mL of medicament. Other embodiments feature about 103 to about 107 cells per mL.
  • A further embodiment of the article further comprises a fetal tissue wrap. The tissue wrap can comprise amnion tissue obtained from one or more animals compatible with a subject animal. The wrap is constructed and arranged for placement in juxtaposition with a site of injury to promote healing.
  • The wrap, comprising fetal tissue, can be air-dried for about 1 minute to about 48 or more. In another embodiment the wrap is air-dried for about 1 hour to about 12 hours. In another embodiment, the wrap is air-dried for about 1 hour to about 6 hours. In another embodiment, the wrap is air-dried for about 1 hour to about 3 hours. In another embodiment, the wrap is air-dried for about 1 hour to about 2 hours.
  • In another aspect of the invention, one embodiment features a kit for the treatment of a subject animal. One kit comprises a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal that can be capable of reconstitution to form a reconstituted medicament.
  • A further embodiment of the kit comprises fetal cells obtained from one or more animals compatible with a subject animal that are compatible with a particulate mixture obtained from the mechanical decellularization of amnion isolated from one or more animals. The fetal cells are applied in cooperation or concurrently with the reconstituted medicament or form a combined medicament comprising a particulate cellular suspension and/or amniotic liquid medicament for administration to the subject animal to promote healing.
  • A further embodiment of the kit comprises a tissue wrap obtained from one or more animals compatible with a subject animal. The tissue wrap comprises amnion tissue constructed and arranged for placement in juxtaposition with the site of injury. The tissue wrap is applied in cooperation or concurrently with the reconstituted medicament or a combined medicament comprising a particulate cellular or acellular suspension medicament with or without mechanically decellularized amnion and/or amniotic liquid for administration to the subject animal to promote healing.
  • A further embodiment of the present invention is directed to a method of promoting healing in a subject animal. One embodiment of the present method features the steps of administering to the site of an injury at least one of the group consisting of a reconstituted medicament, a reconstituted amnion suspension with or without cells and a tissue wrap all of which have been previously described.
  • For example, without limitation, in one embodiment, a method comprises the step of applying a reconstituted medicament. The reconstituted medicament is made from a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal.
  • Another method features the step of applying, by way of injection, a particulate cellular suspension medicament to the periphery of the injury. The particulate cellular suspension medicament suspension comprises particles derived from the mechanical decellularization of amnion obtained from one or more animals compatible with the subject animal, and isolated amniotic fluid cells obtained from the one or more animals compatible with the subject animal.
  • In one aspect, the method further comprises the step of applying a tissue wrap to the site of the injury. The wrap comprises amnion tissue compatible with the subject animal constructed and arranged for placement in juxtaposition with the site of injury. The amnion tissue is air-dried for about 1 minute to about 48 hours or more. In another embodiment the amnion tissue is air-dried for about 1 hour to about 12 hours. In another embodiment, the amnion tissue is air-dried for about 1 hour to about 6 hours. In another embodiment, the amnion tissue is air-dried for about 1 hour to about 3 hours. In another embodiment, the amnion tissue is air-dried for about 1 hour to about 2 hours.
  • In one aspect of the method, the site of injury is debrided prior to applying one or more of the group consisting of a cellular reconstituted suspension, or a reconstituted acellular suspension medicament and a tissue wrap.
  • A further embodiment is directed to a method of making a dried particulate mixture of mechanically decellularized obtained from one or more animals compatible with a subject animal. The dried particulate mixture is capable of reconstitution to form a reconstituted medicament for administration to the subject animal to promote healing. The method comprises the step of mechanically decellularizing amnion tissue to form particles capable of reconstitution.
  • A further embodiment is directed to a method of making a particulate cellular suspension medicament. The method comprises the steps of providing a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal, and fetal cells obtained from one or more animals compatible with a subject animal to form a particulate cellular suspension medicament and forming a particulate cellular suspension. The particulate cellular suspension medicament is administered to a subject animal to promote healing.
  • A further embodiment of the present invention features methods of making a tissue wrap, a particulate mixture medicament and a particulate cellular suspension medicament. One embodiment of the method of making the tissue wrap comprises the steps of applying amnion tissue to a support to form a supported amnion. The supported amnion is next air dried to form the tissue wrap which is placed in a suitable containment means until applied to a site of injury.
  • These and other features and advantages will be apparent upon viewing the Figures that are briefly described below and upon reading the detailed description that follows.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 depicts a kit embodying features of the present invention;
  • FIG. 2 depicts a tissue wrap embodying features of the present invention; and,
  • FIG. 3 shows a tissue wrap embodying features of the present invention in a container.
  • DETAILED DESCRIPTION
  • Unless otherwise defined herein, scientific and technical terms used herein have the meanings that are commonly understood by those of ordinary skill in the art. In the event of any latent ambiguity, definitions provided herein take precedence over any dictionary or extrinsic definition. Unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. The use of “or” means “and/or” unless stated otherwise. The use of the term “including”, as well as other forms, such as “includes” and “included”, is not limiting.
  • It is noted here that as used in this specification and the appended claims, the singular forms “a”, “an”, and “the” also include plural reference, unless the context clearly dictates otherwise.
  • The term “about” or “approximately” means within 10%, and more preferably within 5% (or 1% or less) of a given value or range.
  • As used herein, the term “isolated cell” refers to a cell that has been removed from its in-vivo location.
  • As used herein, the term “decellularization” refers to a process that removes cells from a tissue while preserving the native ultrastructure and composition of the extracellular matrix (ECM). For example, an amnion particulate mixture can be obtained by decellularizing a fetal tissue comprising amnion.
  • There are a number of methods of decellularization of tissue known in the art, including, but not limited to, chemical agents, hypotonic and hypertonic solutions, detergents (e.g., Triton-X), alcohols, solvents (e.g., tributyl phosphate (TBP), biologic agents (e.g., collagenase, trypsin, lipase, nucleases, α-galactosidase), non-enzymatic agents (e.g., chelating agents such as EDTA or EGTA), physical agents (e.g., temperature, force and pressure, non-thermal irreversible, mechanical, electroporation (NTIRE) (see, for example, Crapo et al., Biomaterials. 2011; 32(12): 3233-3243). In certain embodiments, one or a combination of the aforementioned methods may be used to decellularize a tissue. However, methods that preserve the complex composition and three-dimensional ultrastructure of the extracellular matrix (ECM) are preferred.
  • In one embodiment, a tissue is mechanically decellularized, e.g., by cryofractionation, a procedure in which a tissue is frozen and ground in a cryomill to produce a mixture of particles. Such particles are obtained from the cryofractionation of about 0.5 cm2, or about 1 cm2, or about 1.5 cm2, or about 2 cm2, or about 2.5 cm2, or about 3 cm2, or about 3.5 cm2 or about 4 cm2, or about 4.5 cm2 to about 5 cm2 of amnion or more. The amnion can have a thickness of from about 500μ to 50μ, or from 400μ to about 50μ, or from about 300μ to 50μ, or from about 200μ to about 50μ, or from about 150μ to about 50μ, from about 100μ to about 50μ or from about 50μ to about 25μ or less. In another embodiment, the amnion has a thickness of about 500μ, or about 400μ, or about 300μ, or about 200μ, or about 150μ, or about 100μ, or about 50μ, or about 25μ or less.
  • As used herein, 1μ refers to a micrometer or 10−6 meters.
  • The term “amnion” refers to a thin, cellular, extra-embryonic membrane that forms the inner membrane of a closed sac surrounding and protecting an embryo in reptiles, birds, and mammals. The sac contains the fetus and amniotic fluid, in which the embryo is immersed, nourished and protected. Typically, the amnion is a tough, transparent, nerve-free, and nonvascular membrane consisting of two layers of cells: an inner, single-cell-thick layer of ectodermal epithelium and an outer covering of mesodermal, connective, and specialized smooth muscular tissue. In the later stages of pregnancy, the amnion expands to come in contact with the inner wall of the chorion creating the appearance of a thin wall of the sac extending from the margin of the placenta. The amnion and chorion are closely applied, though not fused, to one another and to the wall of the uterus. Thus, at the later stage of gestation, the fetal membranes are composed of two principal layers: the outer chorion that is in contact with maternal cells and the inner amnion that is bathed by amniotic fluid. The amnion has multiple functions, e.g., as a covering epithelium, as an active secretary epithelium, and for intense intercellular and transcellular transport.
  • As used herein, the term “tissue” refers to an aggregate of similar cells and associated extracellular matrix (ECM) forming a definite kind of organized material with a specific function, in a multicellular organism.
  • As used herein, an “amnion tissue” refers to the isolated cellular, extra-embryonic amnion membrane that is detached from the chorion. In one embodiment, the amnion tissue is air-dried. In another embodiment, the amnion is air-dried for about 60 to about 90 minutes or more at ambient temperature (i.e. about 18 to 24° C.).
  • As used herein, a “particulate mixture” refers to the powder or particles obtained from the cryofractionation of amnion.
  • As used herein, the term “fetal tissue” refers to extra-embryonic tissues including, but not limited to, amnion, chorion, yolk sac, the allantois, umbilical cord and/or fetal placenta (villous chorion).
  • As used herein, the term “fetal cells” refers to cells resident in the extra-embryonic tissues including, but not limited to, amnion, chorion, yolk sac, the allantois, umbilical cord, fetal placenta (villous chorion) and/or amniotic fluid. In certain embodiments, the term “fetal cells” refer to isolated fetal cells.
  • In certain embodiments, the term “fetal cells” refers to unfractionated cells of the amniotic fluid including epithelial and/or amniotic fluid or membrane-derived mesenchymal stem cells (see U.S. Patent Publication No. US 2013/0230924, which is incorporated by reference herein in its entirety).
  • The term “injury” means a pathological condition, such as, by way of example, without limitation, a wound, incision, a break in the skin, bone, tendon, ligament, muscle, neoplasia, eye, and soft tissues, an inflammation, infection, or other disease condition.
  • The term “promoting healing” refers to causing a favorable result compared to no treatment. The favorable result comprises any one or more of the following such as reduction of scarring, reduction of inflammation, regrowth of normal tissue or growth of scar tissue, improved load bearing on a limb movement, closure of wound, reduction in infection and reduction in mortality associated with the underlying pathology.
  • The term “compatible with a subject animal” denotes the origin of the tissue as being from the same species or closely related species.
  • In other embodiments, the term “compatible with a subject animal” refers to an xenograft, i.e., a tissue graft from different species.
  • In another embodiment, the term “compatible with a subject animal” refers to allografts, i.e., a tissue from one individual to another of the same species with a different genotype.
  • As used herein, an “animal” refers to living multi-cellular vertebrate organisms, a category that includes, for example, mammals, birds, reptiles, and amphibians. The term mammal includes both human and non-human mammals. Similarly, the term “subject animal” includes both human and non-human subjects. The term “subject animal” refers to an individual human or animal which has a site of injury or disease.
  • As used herein, a non-human animal can refer to a mammal including, but not limited to, a domesticated animal such as a dog, a racing dog, sheep, a pig, a goat, cattle, a zebu, a cat, a guinea pig, a donkey, water buffalo, including “river buffalo” and “swamp buffalo”, a horse, a racing horse, a dromedary camel, a yak, a bactrian camel, a llama, an alpaca, a ferret, a mouse, a bali cattle, a gayal, a rabbit, a rat and a lab rat, a silver fox or a hedgehog.
  • In certain embodiments, a non-human animal can refer to mammals kept in zoos including, but not limited to, zebra, gazelle, wolves, wild swine (pigs & hogs), wild cattle, warthogs, vervet monkeys, two-toed sloths, tree pangolins, tigers, tapirs, tamandua or lesser anteaters, takins, sun bears, striped hyena, spotted hyena, spiral-horned antelope, somali wild ass, snow leopards, small cats, sloth bears, singing dogs, siamang, serval, sea lions, rock hyrax, rhinoceros, reindeer, red pandas, pygmy marmosets, pygmy hippopotamus, przewalski's horses, pronghorns, prairie dogs, porcupines, polar bears, painted dogs, otters, oryx, orangutan, okapi, ocelot, nubian ibex, nile lechwe, naked mole-rats, mountain lions (puma, cougar), monkeys, meerkat, mangabey, mandrill, lynx and bobcats, lions, leopards, lemur, jaguars, honey badgers (ratel), hippos, hamadryas baboons, guenon, guanaco, gorillas, giraffe, giant pandas, giant anteaters, gelada baboons, fossa, fishing cats, elephants, echidna, dhole, coquerel's sifaka, clouded leopards, chimpanzees, cheetahs, tigers, caracals, capybara, camels, brown bears, bonobos, binturongs, bat-eared fox, bats, armadillos, antelope, andean (spectacled) bears and agouti.
  • In certain other embodiments, a non-human animal can refer to mammals considered by the World Wildlife Fund to be endangered including, but not limited to, the amur leopard, black rhino, cross river gorilla, javan rhino, mountain gorilla, pangolin, saola, south china tiger, sumatran elephant, sumatran orangutan, sumatran rhino, sumatran tiger, vaquita, western lowland gorilla, yangtze finless porpoise, african wild dog, amur tiger, asian elephant, bengal tiger, black spider monkey, black-footed ferret, blue whale, bonobo, bornean orangutan, borneo pygmy elephant, chimpanzee, eastern lowland gorilla, fin whale, ganges river dolphin, giant panda, hector's dolphin, indian elephant, indochinese tiger, indus river dolphin, malayan tiger, north atlantic right whale, orangutan, sea lions, sei whale, snow leopard, Sri Lankan elephant, tigers and whales.
  • In certain embodiments, a non-human animal can refer to marsupials, including, but not limited to, wallabies, koalas, possums, opossums, kangaroos, bandicoots, wombats, bettongs, bilbys, quolls, quokkas and the Tasmanian devil.
  • The term “reconstituted” means that that an aqueous liquid is added to make the medicament. A liquid for reconstitution comprises a biocompatible solution such as normal saline, e.g. phosphate buffered saline (PBS) or amniotic fluid. A preferred liquid for reconstitution is calcium-free sterile, non-pyrogenic isotonic solution suitable for intravenous administration. For example, without limitation, one such liquid is sold under the trademark PlasmaLyte A™ in a single dose container for intravenous administration. Each 100 mL contains 526 mg of Sodium Chloride, USP (NaCl); 502 mg of Sodium Gluconate (C6H11NaO7); 368 mg of Sodium Acetate Trihydrate, USP (C2H3NaO2.3H2O); 37 mg of Potassium Chloride, USP (KCl); and 30 mg of Magnesium Chloride, USP (MgCl2.6H2O). It contains no antimicrobial agents. The pH is 7.4.
  • The term “administering” means applying or injecting or ingesting the medicament. The term “applying” is used broadly and includes uses such as washes and implantation.
  • Connective soft tissue defects or injuries often occur by damage to the extracellular matrix (ECM) that forms muscles, ligaments or tendons in mammals. Collagen is the most abundant structural protein in the connective tissue (ECM) and acts as a natural scaffold for cellular attachment in the body.
  • Amnion is an abundant source of collagen, as well as the other proteins, carbohydrates, lipids, hyaluronic acid, laminin, fibronectin, pluripotent mesenchymal stem cells (MSC) and other complex growth factors that are essential for fetal growth and development. In particular, amnion has a complete lack of surface antigens, thus it does not induce an immune response when implanted into a ‘foreign’ body, which is in contrast to most other allograft implants. Amnion also markedly suppresses the expression of the pro-inflammatory cytokines, IL-1α and IL-1β (Solomon et al., 2001, Br. J. Ophthalmol. 85 (4):444-9) and produces natural inhibitors of matrix metalloproteases (MMPs) expressed by infiltrating polymorphonuclear cells and macrophages (Hao et al., 2000, Cornea, 19 (3):348-52; Kim et al., 2000, Exp. Eye Res. 70 (3):329-37). Amnion also down-regulates TGF-β and its receptor expression by fibroblasts leading to the ability to modulate the healing of a wound by promoting tissue reconstruction. Furthermore, amnion has a broad spectrum of antimicrobial activity against bacteria, fungi, protozoa, and viruses for reduced risk of post-operative infection.
  • Amnion derived tissues are therefore immune-privileged and ideally suited for regenerative medicine applications.
  • A “kit” is an assembly of parts, materials, and compositions of matter packaged together to facilitate a treatment. Kits commonly comprise instructions for the use of the parts, materials and compositions.
  • Turning now to FIG. 1, a kit embodying features of the present invention, generally designated by the numeral 11 is depicted. Kit 11 has the following major elements: a first vial 15, a second vial 17, a container for a tissue wrap 21, a syringe 23, and instructions 25. The kit 11 is held in suitable packaging, as depicted, a box 27. Suitable packaging may comprise any means for holding the collection of parts, materials and compositions. For example, without limitation, bags, wraps, containers, ties and the like. The kit 11 for ocular treatments may comprise fibrin glue [not shown] for attaching the tissue wrap 23 to the eye.
  • The first vial 15 contains a dried particulate mixture of mechanically decellularized amnion obtained from one or more animals compatible with a subject animal that upon of reconstitution forms a reconstituted medicament. The kit 11 may contain a vial containing such liquid for reconstitution [not shown] or the liquid for reconstitution may be derived from other sources.
  • The second vial contains fetal cells obtained from one or more animals compatible with a subject animal and compatible with a particulate mixture in the first vial 15. The fetal cells are applied in cooperation or concurrently with the reconstituted medicament or form a combined medicament comprising a particulate cellular suspension medicament for administration to the subject animal to promote healing. In forming a combined medicament, the dried particulate mixture of the first vial 15 is reconstituted with or combined with the fetal cells of the second vial 17, supplemented as needed with further liquid for reconstitution. For example, the dried particulate mixture can be reconstituted by suspension in a solution of 50% solution for reconstitution, such as PlasmaLyte ATM, and 50% amniotic fluid containing fetal cells.
  • The combined medicament is injected into or around the site of injury with syringe 23.
  • The container for a tissue wrap 21 contains a tissue wrap derived from amnion tissue obtained from one or more animals compatible with a subject animal. The tissue wrap comprises amnion tissue constructed and arranged for placement in juxtaposition with the site of injury. The tissue wrap is applied in cooperation or concurrently with the reconstituted medicament or a combined medicament comprising a particulate cellular suspension medicament for administration to the subject animal to promote healing.
  • Turning now to FIG. 2 the container for tissue wrap 21 is depicted as a transparent bag through which the tissue wrap designated by numeral 31 can be seen. As seen in FIG. 3, tissue wrap 31 is formed by affixing amnion tissue to a first support 33 on one side of the amnion and a second support on the other side of the amnion [not shown] and air drying the tissue for thirty minutes to three hours or more, and, most preferably, for about one hour. The supports, of which first support 33 is depicted, maintain the shape of the tissue during the drying process. The supports are preferably removed prior to placement of the tissue in container 21.
  • Tissue wrap 31 is removed from the container and placed over the site of injury.
  • Returning now to FIG. 1, the dried particulate mixture is obtained from the mechanical decellularization or cryofractionation of about 1.5 10−2 cm3 to 5 10−2 cm3 of amnion/mL of reconstituted medicament and include particles greater than 20-100 microns in diameter. Upon reconstitution of the dried particulate mixture by suspension in a solution of amniotic fluid and/or PlasmaLyte A™, the reconstituted medicament can be administered to the subject animal to promote the healing of superficial wounds.
  • A reconstituted medicament containing particles less than about 100 microns is suitable for treatment of eye injuries. The kit 11 for the treatment of eye injuries comprises a dropper for administration.
  • The dried particulate mixture obtained from the cryofractionation comprises about 1.5×10−2 cm3 to about 5×10−2 cm3 of amnion/mL of reconstituted medicament.
  • The fetal cells can comprise amniotic fluid cells and the particulate matter can be filtered to contain particles that are less than 100 microns in diameter. The fetal cells can have a concentration from 103 to 1020/mL mesyschimal and/or epithelial stem cells. In another embodiment, fetal cells can have a concentration of 103 to 1012/mL. In another embodiment, fetal cells can have a concentration of 104 to 1012/mL. In another embodiment, fetal cells can have a concentration of 104 to 1011/mL. In another embodiment, fetal cells can have a concentration of 104 to 1010/mL. In another embodiment, fetal cells can have a concentration of 104 to 109/mL. In another embodiment, fetal cells can have a concentration of 104 to 108/mL. In another embodiment, fetal cells can have a concentration of 104 to 107/mL. In another embodiment, fetal cells can have a concentration of 104 to 106/mL. In another embodiment, fetal cells can have a concentration of 103 to 106/mL. In another embodiment, fetal cells can have a concentration of 103 to 107/mL.
  • In one example, the fetal cells can have a concentration of about 0.8×106 to 1.2×106 cells/mL of the particulate cellular suspension. The dried particulate mixture can contain particles obtained from the cryofractionation of from about 1.5×10−2 cm3 to about 5×10−2 cm3 amnion per mL of the particulate cellular suspension. In one example, the particulate cellular suspension comprises a particulate matter obtained from the cryofractionation about 1.5×10−2 cm3 to about 5×10−2 cm3 amnion for every 106 plus or minus 2×105 amnion fluid cells.
  • Embodiments of the present invention are well suited for the treatment of open wounds, superficial and deep including such wounds that expose a movable body element of the animal's musculoskeletal system, for example, a ligament or tendon. Such wounds can include ligaments and tendons that are lacerated.
  • For example, in the event the animal is a horse and the injury may comprise chondral and fascial pathologies, tendon and ligament pathologies, surface reconstruction or articular joint pathologies. The chondral and fascial pathologies can include, for example, laminitis. The tendon and ligament pathologies can include, for example, tendonitis, sport-induced superficial digital flexor tendon (SDFT) or suspensory ligament (SL) lesions. The articular joint pathologies can include, for example, navicular disease, ringbone or arthritis. The site of injury can also be an open wound that, in one example, is able to heal in about one to two weeks. In another example, healing of the site of injury reduces the risk of recurrence. In one example, the risk of recurrence of the injury can be reduced by about 5 fold.
  • Features of the present invention are further described with respect to the following Examples. These examples feature equine subjects and materials. However, materials derived from other animals species, for example, without limitation, canine, feline, bovine, porcine and other animal species materials and subjects can be prepared in a similar manner as outlined below. The equine materials have been utilized with avian, reptilian and other animal groups.
  • EXAMPLES Example 1: Amniotic Material Processing
  • This procedure defines the aseptic collection of amniotic material (amnion and amniotic fluid) for injection at the site of an injury.
  • Amnion Tissue
  • The amnion container was picked up and sampled for Bioburden. The amnion was aseptically transferred into the sterile field (laminar flow hood). The amnion transport packaging (previously disinfected, i.e. with 70% ETOH) was opened.
  • A 50 mL sample of the Amnion Transport Solution was aseptically transferred into a 30 to 60 mL conical tube for pre-processing bioburden testing. The vial was labeled with sample description, batch number, date and time and placed in a designated refrigerator.
  • (1) Amnion Preparation
  • The amnion from the incoming container was transferred into approximately 200 mL of Plasma Lyte-A in a sterile bioassay dish where it was gently rinsed. A piece of amnion was then spread evenly on a sterile cutting board carefully avoiding any overlaps. A record was made of the amnion preparation start time. Sterile gauze or laps were used to remove any remaining debris/blood from the surface of the amnion. The amnion was inverted and the surface of the opposite site was similarly washed. Any chorion was removed by blunt dissection to separate it from the amnion. After washing and cleaning, the amnion pieces were returned to the bioassay dish containing Plasma Lyte-A. Using a sterile scissors/scalpel, the amnion was cut into 2 to 10 sections. The approximate area of each piece (50-450 cm2) was measured and recorded using a sterile stainless steel ruler.
  • The cleaned pieces of the amnion were placed back on the sterile cutting board and the amnion was spread out on the board taking care to not overlap. A sterile nylon mesh was placed over the surface of the amnion again taking care to overlap them. The amnion and mesh were then placed onto a sterile drying rack and allowed to air dry for a minimum of one hour. Start and stop times for drying were recorded.
  • (2) Amniotic Fluid
  • (a) Amnion Fluid Preparation
  • A large sterile pan was first placed into the sterile field (laminar flow hood) and filled with cold packs from a −80° C. freezer. The aspiration containers with the amniotic fluid were disinfected with 70% ethanol (ETOH), inspected for integrity and placed on the cold packs in the laminar flow hood. The source of the amniotic fluid was confirmed by looking at the Donor animal ID number. The 2.0 mL of amniotic fluid was then aseptically pipeted into a 2 mL sterile microcentrifuge tube for bioburden testing. The vial was labeled with the sample number, batch number, date and time and placed in the designated refrigerator.
  • Using a 50 mL sterile disposable serological pipette all the remaining amniotic fluid was transferred into 1 liter sterile disposable bottles and placed on the cold pack. The total volume of the amniotic fluid and the color were recorded.
  • (3) Amniotic Fluid Cell Count and Determination of the Number of Viable Cells
  • Each amniotic fluid bottle was gently mixed and 1.0 mL of the fluid was collected using a 1 mL micropipette and transferred into a 2 mL Eppendorf microcentrifuge tube. A total of 50 μL of cell suspension was then added to 50 μL of trypan blue (0.4%) in an Eppendorf microcentrifuge tube and vortexed for 5 seconds. The sample was placed on a rack for 5 minutes.
  • A Neubauer chamber (hemocytometer) was rinsed with distilled water, and then sprayed with ETOH 70% and wiped clean and dried with paper towels. A cover slip was placed on the top of the micro-grids of the chamber. The trypan blue-cell suspension was gently mixed and used to fill both sides of the hemocytometer with 10 μL by capillary action. The cells were allowed to settle down for at least 30 seconds. The hemocytometer was placed under the microscope and all cells in the four 1 mm corner squares and one 1 mm center square were counted. For accuracy the total number of cells counted was greater than 100. The cells were re-counted if >10% of the cells appeared clustered, by vigorously pipetting in the original cell suspension as well as in the trypan blue cell suspension mixture. Using a double cell counter, the number of viable and non-viable cells was determined. The cells in both chambers were counted and an average was calculated. For the trypan blue test, live cells did not take up the dye, whereas dead (non-viable) cells did. Thus non-viable cells stained blue and viable cells remained opaque. “Ghost” cells, which appeared as flattened pale blue cells were not counted.
  • The number of cells was determined as follows: Each square represented a total volume of 0.1 mm3 or 1×10−4 mL (0.1 mm depth×1 mm width×1 mm height=0.1 mm3). The number of cells per mL was then deduced from the average viable cell count per 1 mm square×2×104. The total cell number was therefore equal to the number of cells per mL multiplied by the original volume of sample fluid. The cells were then diluted to the desired concentration.
  • Example 2: Aseptic Processing of the Amniotic Material
  • (1) Aseptic Cryofractionation of Amnion
  • After at least one hour, the amnion was removed from the drying rack and transferred into the milling chambers having an impactor. The milling chambers were placed into the Cryomill and cryofractionated using the following settings:
  • Number of Cycles: 4
  • Frequency 1/s: 10 CPS
  • Precooling Time: 10 minutes
  • Grinding Time: 4 minutes
  • Intermediate Cooling: 3 minutes
  • Once grinding was complete, the milling chambers were allowed to warm to room temperature for approximately two hours. The start and stop times were recorded.
  • Approximately 50 mL of the amnion suspension solution was dispensed into each milling chamber. The inside milling chamber and the impactor were rinsed with the solution multiple times until the ground amnion (dried particulate mixture) was re-suspended and collected in the bottom of the chamber. The impactor was removed using the magnet pen. The cryofractionated amnion solution was then transferred to the amnion suspension container and placed on cold packs in the sterile field and diluted to the desired amount.
  • (2) Aseptic Processing of Amniotic Fluid
  • The amniotic fluid was aliquoted evenly into 50 mL sterile centrifuge tubes and centrifuged at 200-400×g (1100-1600 rpm) for approximately 5-10 minutes at room temperature. The supernatant was then removed from each tube using a 25 mL sterile serological pipette. The amniotic liquid was kept in new container and the pellet was re-suspended in Plasma Lyte-A™ to a total volume of 25 mL in each tube. The re-suspended cells in any two different tubes were vortexed for approximately 3 to 5 sec and consolidated into a single tube prior to centrifugation at 200-400×g (1100-1600 rpm) for approximately 5-10 minutes at ambient temperature. The preceding steps were repeated as necessary.
  • The supernatant from each tube was removed using a sterile pipette and the pellet was again re-suspended in a cell suspension solution (amniotic fluid and/or an isotononic solution, e.g., PlasmaLyte A™) to bring the volume in each tube to about 10 mL and vortexed for approximately 3 to 5 seconds. A 1 mL aliquot was removed and the cell count and viability was determined using the above-described trypan blue test.
  • If red blood cells were present in the amniotic fluid cell suspension, they were removed using a RBC Lysing Solution. A 10× concentration was prepared as follows: NH4Cl (ammonium chloride)=8.02 gm+NaHCO3 (sodium bicarbonate)=0.84 gm adjusted to a total volume of 100 mL with Millipore filtered water. 10 mL of the 10× concentrate was added to 90 mL Millipore filtered water and refrigerated until use. The amount needed of Erythrolysis solution (15 mL per tube centrifuged) was removed from the refrigerator and kept for a period of 0.5 hours in the stabilization incubator. After centrifuging the amniotic fluid at 400×g for 10 minutes, the supernatant was removed and the pellet was re-suspended in Erythrolysis solution (minimum of 50 mL per tube). The contents of all the tubes were consolidated into one tube that was rocked for ˜10 minutes at room temperature until the liquid was clear red. The cells were again centrifuged for 5 minutes at 250 to 400×g. The supernatant was decanted. The pellet was washed with 50 mL of PBS or PlasmaLyte A™ before centrifugation again for 10 minutes at 250 to 400×g. The washing of the pellet was repeated as needed. The amniotic fluid cells were then filtered through a 100 μm cell strainer, and re-suspended in PBS or PlasmaLyte ATM. The cells were again centrifuged for 10 minutes at 250 to 400×g. The supernatant was decanted and the pellet was left in the 50 mL conical centrifuge tube.
  • (3) Procedure for Cryopreservation of Cryofractionated Amnion with Amnion Fluid Cells
  • Appropriate size cryovials that were previously labeled and their corresponding size of CoolCell™ freezer (CCF) racks were placed in the hood. CryoStor 10®, the Cell Suspension Solution and the Amnion Suspension Solution were also placed on cold packs in the hood. Cryostor 10™ is commercially available from Biolife Solutions.
  • The cell suspension solution and the amnion suspension solution were then combined into the cell suspension solution container. Using 50 mL serological pipettes, the solutions were homogenized several times. The container was again placed on the cold packs on the sterile field. Empty cryovials were placed in the CCF racks on cold packs and their caps were removed inside the hood (sterile field). The mix of cell/amnion suspension solution was pipeted into an empty Amnion Suspension container and a same volume of CryoStor 10™ was added and homogenized before being placed on the cold packs. A 50 mL pipette Combitip was fitted on to a repeat pipetor set to dispensing mode. The fill volume was adjusted and 50 mL of the Cell/Amnion solution was aspirated and then adjusted to the desired dispense volume (1 or 2 mL). The cryovials in the CCF were then filled and the vial caps were replaced securely and the rack of filled vials was placed on cold packs for QC inspection.
  • Cell freezing was achieved by cooling the cells at a cooling rate of 1° C. per minute from 4° C. to −80° C., using a passive cooling controlled-rate freezer CoolCell™ (commercially available from Biocision.)
  • The cell suspension in cryoprotective freezing medium was aliquoted into each of the cryovials and the cells were gently mixed to maintain a homogeneous cell suspension. The solid core of the CoolCell (black ring) at room temperature was seated in the bottom of the central cavity and the vials containing the cell suspension were placed in each well. The lid of the CoolCell™ was fully sealed and the Coolcell™ was placed into a −80° C. freezer for at least 4 hours prior to transfer on dry ice to long term storage. Cell viability and QA/QC were evaluated by thawing one vial after short term storage.
  • Example 3: Amniotic Tissue Wrap Preparation
  • After confirming the amniotic tissue source and donor mare ID and recording the time of receipt, the amnion transport packaging (previously disinfected, i.e. with 70% ethanol, methanol, etc.) was aseptically transferred into the sterile field (a laminar flow hood). A sample of the Amnion Transport Solution was first transferred into a 50 mL conical tube for Bioburden testing. The vial was then labeled with sample description, batch number, date and time and placed in designated refrigerator.
  • (1) Amniotic Membrane Wrap Preparation
  • Saline was aseptically added into a second receiving pan in the sterile field (i.e. laminar flow hood) and the amnion tissue was taken from incoming receiving pan to the second receiving pan containing the sterile saline. Any remaining blood was rinsed with sterile saline. After documenting the amnion preparation start time, sterile gauze or laps was used to remove any remaining debris/blood from the surface of the amnion. The amnion was then inverted and the other side was rinsed and washed. Any remaining chorion was removed by blunt dissection to separate it from the amnion. The amnion was kept wet with sterile saline. The tissue was blocked off by cutting away any stringy ends and checked for holes or tears. After repositioning the amnion on the cutting board with the chorion side up, the approximate area of each piece of amnion was measured and recorded in cm2 using a sterile stainless steel ruler.
  • The amnion (chorion side up) was covered with a sterile mesh (e.g., a nylon mesh) wetted with sterile saline. For the purpose of this discussion, there is a first steel mesh which is denoted by a first visible indicia, the color white, and a second steel mesh, to be discussed below, which is denoted by a second visible indicia, the color blue. Any reference to the colors white or blue are directed to these visible indicia. Those skilled in the art will recognize that such visible indicia is matter of choice. The mesh was cut to size and was allowed to slightly overlap the amnion. The mesh-covered amnion was then gently lifted, turned over and placed back on the cutting board. Caution was used as to not disturb the mesh/amnion interface. The newly exposed side of the amnion was wiped with sterile wipes or gauze pads to remove any remaining blood or small tissue particles. The newly exposed side of the amnion was covered with a second steel mesh, BLUE sterile mesh (e.g., a nylon mesh) wetted with sterile saline. The mesh was cut to size and was allowed to slightly overlap the amnion.
  • The amnion tissue sandwiched between the white and blue mesh was placed on the drying rack where the amnion tissue was allowed to dry for 60 to 90 minutes at ambient temperature (65° to 70° F.). Caution was used as to not disturb the mesh/amnion interface. The amnion was kept unfolded and as flat as possible during this step. Additional drying racks were used as needed. The total drying time was recorded. The dried tissue was then removed from the rack(s) and laid flat on the cutting board, WHITE side up. The WHITE mesh was carefully removed from the entire sheet of amnion which was checked for holes or tears. With the BLUE mesh side up on the cutting board, each section was cut using a scalpel or rotary cutting blade and the sizes and surface areas were recorded as 5×5 cm, 10×10 cm, round 15 mm diameter and round 22 mm diameter. Those skilled in the art will recognize that the sections can be cut and sized to fit particular needs and these sizes and shapes are only exemplary.
  • (2) Amniotic Membrane Wrap Pre-Packing
  • Pouches, Sealing Test and Labeling
  • The sterile field was set up for packaging including a sealer for packing pouches (Sealer settings: Temperature=177±9° C. (350±15° F.)). Three (3) empty pouches were sealed for visual inspection and retention and then labeled. Packing pouches are available from niumerous vendors. One suitable pouch is sold under the trandemark KAPAK™.
  • Amniotic Membrane Wrap Packing
  • Pouches were transferred onto the sterile field (previously disinfected laminar flow hood). Using sterile forceps, each individual tissue membrane was inserted into the inner pouch. Large membranes were folded if necessary. The pouches were sealed with the dried amnion tissue on a mesh (see, for example, FIGS. 2 and 3) and inspected for a broken seal, impurities, and defects. Upon passing the inspection, the pouches were labeled with date and packaged in large pouches according to size and stored in the refrigerator or at room temperature. The donor ID, size, date, time, and initials were documented.
  • Example 4: Implantation of Cryofractionated Amnion and Isolated Amniotic Fluid Cells
  • (1) Thawing Vials
  • A container was ¾ filled with hot tap water with a thermometer and cold water was added until a temperature of 37° C. was reached. A vial of cryofractionated amnion with amniotic fluid cells was taken from the −80° C. freezer. Holding the cap, the vial was partial immersed in the water bath for approximately 2 to 3 minutes with gentle agitation until the contents were melted. The vial was removed from the water bath and the exterior was wiped with sterile gauze saturated with 70% ethanol. The thawed contents were then ready for immediate use.
  • (2) Animal Restraint
  • A horse with an injury was adequately restrained and sedated or anaesthetized before starting the procedure according to the veterinarian's directions. Appropriate sedation or anesthesia caused minimal distress to the patient and prevented any long lasting discomfort. Use of nose twitch was optionally required. In the case of young adults or unbroken horses, short-acting general anesthesia was required.
  • (3) Implantation Procedure
  • The site for the product implantation was processed as a surgically prepared area. After cleaning or clipping of any gross contamination, such as dust or mud, the complete the area was scrubbed with chlorhexidine for 5 to 7 minutes and then wiped down with alcohol swabs. Before proceeding, the area was cleaned until the alcohol swabs used on the scrubbed area were dirt-free. Systemic antibiotics were administered prior to starting the procedure. However, in some instances, systemic antibiotics are not necessary.
  • The contents of the 2 mL vial were split into 3 to 4 doses (0.50 to 0.67 mL) and loaded into syringes (for example, 1 mL syringes) with sterile hypodermic needles (for example 22 gauge, 1.5 inch needles). The appropriate leg was resting or held up and restrained as needed. For injuries to a tendon, ligament or muscle the cryofractionated amnion and amniotic fluid cell mix was injected in close proximity to the lesion without penetrating the actual tendon, ligament or muscle. For injuries to articulation (i.e., joints) the cellular particulate mixture was injected either peri-articularly or intra-articularly. After injection, the site was bandaged with sterile swabs and adhesive bandage.
  • Example 5: Implantation of Cryofractionated Amnion and Isolated Amniotic Fluid Cells in Combination with the Amnion Tissue Wrap
  • (1) Animal Restrain
  • A horse was adequately restrained as described in Example 4.
  • (2) Materials
  • A kit comprising sterile hypodermic needles (18 and 23 gauge), syringes (3 to 5 mL), vials of thawed cryofractionated amnion and amniotic fetal cells as well as packaged amnion tissue wrap of the appropriate size were assembled (see FIG. 1) together with clippers, sterile gloves, 1% lidocaine, saline and sedatives and/or anesthetics.
  • (3) Wound Surface Preparation
  • A horse was treated at a veterinary clinic a severe leg severe injury caused by barbed wire. The wound was initially pre-cleaned to remove dirt, scrubbed with chlorhexidine for 5 to 7 minutes and then wiped down with alcohol swabs.
  • (4) Sharp surgical debridement:
  • The veterinarian aggressively debrided the wound. This step created a relatively clean wound bed and generated an inflammatory signal that induced the migration and proliferation of stem cells and growth factors (from the amniotic material and horse's own immune system). Systemic antibiotics were administered prior to starting the procedure.
  • (5) Treatment of an Open Wound with Amniotic Tissue Wrap
  • The veterinarian determined the amount of amniotic material needed to treat the wound by measuring the maximal length, width and depth of the wound. Amniotic tissue wrap of the appropriate size was selected and applied on the wound. Treatment of an open wound with Amniotic tissue wrap and a cellular suspension of cryofractionated amnion and isolated amnion fluid cells.
  • After the application of the amnion tissue wrap as described above, the approximate volume of the open wound volume was estimated and the amount of cryofractionated amnion and isolated amnion fluid cells to be injected were determined in accordance with guidelines shown in Table 3.
  • TABLE 3
    Amount of cryofractionated amnion and isolated amnion fluid cells to
    be injected as a function of wound volume.
    Amount of cryofractionated amnion/
    Wound Area amnion fluid cell suspension to use
    <12.5 cm2 1.0 mL
    >12.5 cm2 but <25 cm2 1.5 mL
      >25 cm2 2.0 mL
  • After debridement, the cryofractionated amnion/amnion fluid cell suspension was injected into the pen-wound skin that appeared normal in texture, turgor, and free of infection using a 23-gauge needle on a 3 mL syringe.
  • The cell suspension was injected at a distance of approximately 5 mm from the wound edges and 10 to 15 mm deep at the 3-, 6-, and 9- and 12-o'clock positions (directly into the superficial fascia and the subcutaneous tissue of the wound), aiming the needle parallel to the wound margin at each location. To minimize discomfort and to ensure an adequate spread of the amniotic material, the cryofractionated amnion/amnion fluid cell suspension was mixed with 1% plain lidocaine in a 1:1 ratio. After the completion of the procedure, the surgical site was dressed with a nonporous dressing followed by application of a nonstick dressing and a dry sterile dressing of gauze. Five to seven days after implant of the amnion allograft, the wound was redressed, and standard wound care consisting of saline wet to dry sterile gauze dressing was resumed. If the wound proved hard to heal, additional applications of the amnion tissue wrap and/or cryofractionated amnion/amnion fluid cell suspension were made using the same technique at 14- to 21-day intervals until the wound healed. Concomitant therapies were given i.e. wound offloading, and/or edema control as recommended by the veterinarian.
  • (6) Healing of Wound
  • The wound was assessed and showed striking improvement. Generally, treatment shows 2.5 times the rate of healing.
  • Example 6: Treatment of Eye Injuries with the Amnion Tissue Wrap
  • (1) Animal Restrain
  • A horse was adequately restrained as described in Example 4.
  • (2) Eye Surface Preparation
  • Corneal ulcers are the most common eye problem in horses. A veterinarian located the foreign material and determined the wound severity (size of the corneal lesion) using visual inspection or fluorescein dye to highlight any subtle corneal defect or scratch. The area around the eye was cleaned to remove debris or excessive liquid using sterile gauze.
  • (3) Application of the Amniotic Tissue Wrap
  • An appropriate size of amniotic tissue wrap (ie. 15 or 22 mm diameter discs) was selected. The dehydrated amniotic tissue wrap was positioned over the tip of the index finger and applied directly to the eye surface while opening the eye lids with the other hand (if needed). The membrane attached to the eye surface by hydrostatic tension (glue or sutures may be used to assist to insure placement in challenging environments.).
  • Example 7: Treatment of Eye Injuries with a Suspension of Cryofractionated Amnion
  • (1) Animal Restrain
  • A horse was adequately restrained as described in Example 4.
  • (2) Eye Surface Preparation
  • The eye surface and surrounding area was prepared as described in Example 6.
  • (3) Application of Eye Drops of Cryofractionated Amnion
  • Eye drops were given either alone or in conjunction with the amniotic tissue wrap application described in Example 6. Vials of cryofractionated amnion suspended in saline (ie. PlasmaLyte A™ and/or Amniotic Fluid). were thawed (36 to 37° C. or 97 to 98° F. water bath) for at least 5 min. The vial was gently rotated upside down to mix solution. 2 drops of the suspension were applied to the injured eye twice a day (4 drops total per day) over the membrane or ocular surface. Treatment lasted for a week or until the suspension was used up. The suspension was pH balanced to prevent any irritation to the eye.
  • The 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.
  • Any patent, patent application, publication, or other disclosure material identified in the specification is hereby incorporated by reference in its entirety and for all purposes to the same extent as if each such individual reference (e.g., patent, patent application, publication, or other disclosure material) was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Any material, or portion thereof, that is said to be incorporated by reference herein, but which conflicts with existing definitions, statements, or other disclosure material set forth herein, is only incorporated by reference to the extent that no conflict arises between that incorporated by reference material and the present disclosure material.
  • Other embodiments are within the following claims.

Claims (21)

1.-97. (canceled)
98. An article of manufacture comprising:
(i) a dried particulate mixture of mechanically decellularized fetal tissue obtained from one or more animals compatible with a subject animal, the dried particulate mixture is capable of reconstitution to form a reconstituted medicament for administration to the subject animal to promote healing; and
(ii) fetal cells obtained from one or more animals that are compatible with the dried particulate mixture,
wherein combining the dried particulate mixture and the fetal cells forms a medicament comprising a particulate cellular suspension for administration to the subject animal to promote healing,
wherein the subject animal is a non-human animal.
99. The article of manufacture of claim 98, wherein the fetal tissue and/or fetal cells comprises amnion or chorion.
100. The article of manufacture of claim 98, wherein the dried particulate mixture is obtained from the mechanical decellularization of about 1.5-5×102 cm3 of amnion/mL of reconstituted medicament.
101. The article of manufacture of claim 98, wherein the dried particulate mixture comprises particles greater than about 100 microns in diameter.
102. The article of manufacture of claim 98, wherein the dried particulate mixture contains particles of less than about 100 microns in diameter.
103. The article of manufacture of claim 98, wherein the particulate cellular suspension comprises about 0.8-1.2×106 amnion fluid fetal cells/mL of medicament.
104. The article of manufacture of claim 98, further comprising an air dried fetal tissue wrap obtained from one or more animals compatible with a subject animal, the fetal tissue wrap constructed and arranged for placement in juxtaposition with a site of injury to promote healing.
105. The article of manufacture of claim 98, wherein the fetal tissue comprises amnion and/or chorion.
106. The article of manufacture of claim 104, wherein the wrap is constructed and arranged for placing on the site of at least one of an eye injury or a corneal ulcer.
107. The article of manufacture of claim 98, wherein the healing occurs at a site of injury that exposes a movable body element of the animal's musculoskeletal system, wherein the movable body element is a muscle, joint, ligament, or tendon, wherein the injury comprises a laceration.
108. The article of manufacture of claim 107, wherein the healing is at a site of injury caused by a condition selected from the group consisting of chondral and fascial pathologies, tendon and ligament pathologies, surface reconstruction and articular joint path
109. A method for treating an injury in a subject animal, comprising the steps of: administering the article of manufacture of claim 98 to the periphery of an injury of a non-human animal, wherein the administering the particulate cellular suspension promotes healing of the injury,
wherein the administering comprises any one or more of an intravenous injection, an intra-articular injection, a subcutaneous injection, a superficial injection or a nebulization.
110. The method of claim 109, further comprising the step of applying an air dried fetal tissue wrap to the site of the injury.
111. The method of claim 109, wherein the injury is debrided prior to injecting.
112. The method of claim 109, wherein the injury is an eye injury.
113. A method, comprising:
(i) obtaining animal fetal tissue; and
(ii) decellularizing at least a portion of the animal fetal tissue to obtain a particulate mixture.
114. The method of claim 113, further comprising reconstituting the particulate mixture through adding a biocompatible solution to the particulate mixture, wherein adding the biocompatible solution creates a reconstituted particulate mixture comprising fetal cells, wherein the reconstituted particulate mixture is suitable for treating injuries to compatible injured animals.
115. The method of claim of claim 114, further comprising;
(i) filtering the reconstituted particulate mixture to increase the concentration of fetal cells; and
(ii) injecting the particulate mixture into an injured animal,
wherein the injection is made to a periphery of an injury of the injured animal.
116. The method of claim 115, wherein decellularizing at least a portion of the animal fetal tissue comprises obtaining extracellular matrix through mechanical decellularization comprising cryofractionation.
117. The method of claim 113, further comprising obtaining animal amnion fluid.
US15/764,275 2015-09-29 2016-09-29 Methods and articles of manufacture for the treatment of animals Pending US20180318359A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/764,275 US20180318359A1 (en) 2015-09-29 2016-09-29 Methods and articles of manufacture for the treatment of animals

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201562234354P 2015-09-29 2015-09-29
PCT/US2016/054475 WO2017059089A1 (en) 2015-09-29 2016-09-29 Methods and articles of manufacture for the treatment of animals
US15/764,275 US20180318359A1 (en) 2015-09-29 2016-09-29 Methods and articles of manufacture for the treatment of animals

Publications (1)

Publication Number Publication Date
US20180318359A1 true US20180318359A1 (en) 2018-11-08

Family

ID=58427940

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/764,275 Pending US20180318359A1 (en) 2015-09-29 2016-09-29 Methods and articles of manufacture for the treatment of animals
US15/473,513 Active 2037-11-02 US10993968B2 (en) 2015-09-29 2017-03-29 Methods and articles of manufacture for cosmetic results

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/473,513 Active 2037-11-02 US10993968B2 (en) 2015-09-29 2017-03-29 Methods and articles of manufacture for cosmetic results

Country Status (4)

Country Link
US (2) US20180318359A1 (en)
CA (1) CA3000401C (en)
MX (1) MX2018004015A (en)
WO (1) WO2017059089A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11369718B1 (en) * 2018-08-24 2022-06-28 Ariasa LLC Method and use of cryopreserved mesenchymal stem cells via liquid suspension from the amniotic sac for rejuvenation of the midface
CN112961775B (en) * 2021-03-10 2023-05-12 秦娟 Cell resuscitator
WO2023100067A1 (en) * 2021-11-30 2023-06-08 Riccio Michele Production process of a pulverized scaffold
WO2023205236A1 (en) * 2022-04-20 2023-10-26 Biostem Technologies, Inc. Multi-part processed human amniotic composition and methods of making and using thereof for treatment of peyronie's disease
US20230364154A1 (en) * 2022-05-12 2023-11-16 Anicell Biotech, Llc Methods and articles of manufacture for animal therapeutics

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014152234A1 (en) * 2013-03-15 2014-09-25 Anthrogenesis Corporation Improved method of making extracellular matrix compositions
US20170258727A1 (en) * 2011-06-10 2017-09-14 Tissuetech, Inc. Methods of processing fetal support tissues, fetal support tissue powder products, and uses thereof
US20180221418A1 (en) * 2012-08-15 2018-08-09 Mimedx Group, Inc. Micronized placental tissue compositions and methods of making and using the same

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030187515A1 (en) * 2002-03-26 2003-10-02 Hariri Robert J. Collagen biofabric and methods of preparing and using the collagen biofabric
AR047712A1 (en) * 2002-09-07 2006-02-15 Royal Veterinary College TREATMENT METHOD OF A NATURAL SOFT SKELETTIC TISSUE INJURY MANAGING A COMPOSITION OF MESENQUIMATOSE MOTHER CELLS
EP1858533A4 (en) * 2005-02-22 2009-09-09 Univ Johns Hopkins Med Use of amniotic fluid (af) in treating ocular disease and injury
WO2008060377A2 (en) * 2006-10-04 2008-05-22 Anthrogenesis Corporation Placental or umbilical cord tissue compositions
WO2009152205A1 (en) * 2008-06-10 2009-12-17 Meta Cosmetics, Llc. Topical compositions comprising imidazolidinedione analogs and their use to treat or prevent the appearance of skin wrinkling
US20110158958A1 (en) * 2009-12-07 2011-06-30 Sing George L Methods for treating ophthalmic disorders, diseases and injuries
EP2524034A1 (en) * 2010-01-14 2012-11-21 Organogenesis, Inc. Bioengineered tissue constructs and methods for producing and using thereof
SG189438A1 (en) * 2010-10-18 2013-05-31 Agency Science Tech & Res Use of exosomes to promote or enhance hair growth
US9132156B1 (en) * 2014-06-15 2015-09-15 Amnio Technology Llc Acellular amnion derived therapeutic compositions
US10736990B2 (en) * 2015-02-11 2020-08-11 Mimedx Group, Inc. Collagen and micronized placental tissue compositions and methods of making and using the same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170258727A1 (en) * 2011-06-10 2017-09-14 Tissuetech, Inc. Methods of processing fetal support tissues, fetal support tissue powder products, and uses thereof
US20180221418A1 (en) * 2012-08-15 2018-08-09 Mimedx Group, Inc. Micronized placental tissue compositions and methods of making and using the same
WO2014152234A1 (en) * 2013-03-15 2014-09-25 Anthrogenesis Corporation Improved method of making extracellular matrix compositions

Also Published As

Publication number Publication date
MX2018004015A (en) 2018-06-06
US10993968B2 (en) 2021-05-04
CA3000401A1 (en) 2017-04-06
US20170216195A1 (en) 2017-08-03
CA3000401C (en) 2022-09-27
WO2017059089A1 (en) 2017-04-06

Similar Documents

Publication Publication Date Title
CA3000401C (en) Articles of manufacture containing decellularized and reconstituted fetal tissue and methods of making the same
TW398982B (en) Grafts made from amniotic membrane; methods of separating, preserving, and using such grafts in surgeries
JP6755893B2 (en) Compositions and Methods for Treating and Preventing Tissue Injuries and Diseases
KR100999247B1 (en) Collagen biofabric and methods of preparation and use therefor
KR20160147058A (en) Therapeutic placental compositions, methods of making and methods of use
US20150010609A1 (en) Immunocompatible chorionic membrane products
Lim et al. Placental cells and tissues: the transformative rise in advanced wound care
Falcão et al. Effect of allogeneic mesenchymal stem cells (MSCs) on corneal wound healing in dogs
JP2017514507A (en) Immunocompatible chorionic product
JP2017514879A (en) Immunocompatible amniotic product
CN108938669A (en) A kind of stem cell ointment and preparation method thereof for treating skin injury
US11160272B2 (en) Methods for preserving tissue with medical grade honey
KR101816964B1 (en) Pharmaceutical adjuvant composition for treating damages of skin or blood vessel tissue
CN103301153B (en) Application of umbilical cord mesenchymal stem cell culture solution or product prepared from umbilical cord mesenchymal stem cell culture solution in preparation of medicament for treating skin wound
US11395837B2 (en) Placental tissue component compositions for treatment of skin defects and methods using same
CN110170072A (en) The preparation of cell fibronectin and the method for repairing dermal tissue
US20230364154A1 (en) Methods and articles of manufacture for animal therapeutics
JP7213479B2 (en) skin protectant
WO2022221852A1 (en) Multi-layer amniotic tissue grafts and uses thereof
Duddy Efficacy of a Liquid Amnion Allograft on Equine Healing
JP2024519731A (en) Allogeneic cultured keratinocyte products
JP2023502888A (en) Methods for Coating and Drying Heterogeneous Stem Cell-Derived Extracellular Vesicles
CN117425507A (en) Multi-layer amniotic tissue graft and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: ANICELL BIOTECH, LLC, ARIZONA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AMES, BRANDON;BARCELO-FIMBRES, MOISES;SIGNING DATES FROM 20180327 TO 20180411;REEL/FRAME:045554/0396

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCV Information on status: appeal procedure

Free format text: APPEAL BRIEF (OR SUPPLEMENTAL BRIEF) ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED