WO1998012274A1 - Methods and devices for preparing protein concentrates - Google Patents

Methods and devices for preparing protein concentrates Download PDF

Info

Publication number
WO1998012274A1
WO1998012274A1 PCT/US1997/016897 US9716897W WO9812274A1 WO 1998012274 A1 WO1998012274 A1 WO 1998012274A1 US 9716897 W US9716897 W US 9716897W WO 9812274 A1 WO9812274 A1 WO 9812274A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogel
composition
protein
kit
fibrinogen
Prior art date
Application number
PCT/US1997/016897
Other languages
French (fr)
Inventor
Chandrashekar Pathak
Stephen C. Rowe
Original Assignee
Chandrashekar Pathak
Rowe Stephen C
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
Priority to AU46486/97A priority Critical patent/AU4648697A/en
Application filed by Chandrashekar Pathak, Rowe Stephen C filed Critical Chandrashekar Pathak
Publication of WO1998012274A1 publication Critical patent/WO1998012274A1/en
Priority to US10/010,715 priority patent/US7009034B2/en
Priority to US10/068,807 priority patent/US6887974B2/en
Priority to US10/293,453 priority patent/US7211651B2/en
Priority to US10/364,592 priority patent/US7057019B2/en
Priority to US11/293,892 priority patent/US7332566B2/en
Priority to US11/725,751 priority patent/US7605232B2/en
Priority to US12/069,821 priority patent/US7592418B2/en
Priority to US12/156,085 priority patent/US8003705B2/en
Priority to US12/496,060 priority patent/US20090324721A1/en
Priority to US12/555,970 priority patent/US8557535B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/75Fibrinogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • 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
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • 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
    • 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
    • A61L24/00Surgical adhesives or cements; Adhesives for colostomy devices
    • A61L24/04Surgical adhesives or cements; Adhesives for colostomy devices containing macromolecular materials
    • A61L24/10Polypeptides; Proteins
    • A61L24/106Fibrin; Fibrinogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/30Extraction; Separation; Purification by precipitation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L89/00Compositions of proteins; Compositions of derivatives thereof
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06KGRAPHICAL DATA READING; PRESENTATION OF DATA; RECORD CARRIERS; HANDLING RECORD CARRIERS
    • G06K7/00Methods or arrangements for sensing record carriers, e.g. for reading patterns
    • G06K7/10Methods or arrangements for sensing record carriers, e.g. for reading patterns by electromagnetic radiation, e.g. optical sensing; by corpuscular radiation
    • G06K7/10544Methods or arrangements for sensing record carriers, e.g. for reading patterns by electromagnetic radiation, e.g. optical sensing; by corpuscular radiation by scanning of the records by radiation in the optical part of the electromagnetic spectrum
    • G06K7/10821Methods or arrangements for sensing record carriers, e.g. for reading patterns by electromagnetic radiation, e.g. optical sensing; by corpuscular radiation by scanning of the records by radiation in the optical part of the electromagnetic spectrum further details of bar or optical code scanning devices
    • G06K7/10831Arrangement of optical elements, e.g. lenses, mirrors, prisms
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06KGRAPHICAL DATA READING; PRESENTATION OF DATA; RECORD CARRIERS; HANDLING RECORD CARRIERS
    • G06K2207/00Other aspects
    • G06K2207/1016Motor control or optical moving unit control
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit

Definitions

  • the technical field of this invention is the preparation of concentrated protein compositions.
  • Background of the Invention Methods of preparing concentrated protein compositions from initial dilute protein compositions find use in a variety of different industries, including the chemical, biological, academic research, biotechnological and medical industries.
  • "Fibrin Sealants” also known as fibrin gels or fibrin glues
  • fibrin gels or fibrin glues are a type of blood derived composition used in the medical industry which are prepared through methods of concentrating blood plasma proteins that have been developed for use as tissue adhesives. drug delivery vehicles and the like, .although such compositions are not yet FDA approved in the United States due to concerns over blood borne contaminants, such compositions are marketed in Europe and elsewhere throughout the world.
  • a typical commercial fibrin glue kit consists of a iai ol Kophili/ed concentrated human fibrinogen. prepared from pooled human donor blood, that also contains fibronectin. Factor XIII and reduced amounts of piasminogcn The concentrate, also known as cryoprecipitate.
  • the second component of the adhesive system is a lyophilized bovine thrombin solution which is reconstituted with a calcium chloride solution
  • the formulation may also contain additional components like a fibnonolysis inhibitor
  • the reconstituted solutions are mixed and used as a surgical adhesive system
  • the most common method used for the preparation of the fibrinogen component of the above described kits is cryoprecipation In cryoprecipitation. Fresh blood plasma is frozen at -80 °C for at least 6 to 12 h The temperature of the frozen plasma is then raised to around 0-4 °C.
  • Microencapsulated drug particles and similarly protected pharmaceutically active agents are described in: Kissel et al.. J. Controlled Release ( 1991 ) 16:27; Tabata et al.. Pharmaceutical Research (1993) 10:487: EP A 83303606.4; U.S. Patent No. 5.143.662: Mathiowitz & Langer. J. Controlled Release ( 1987) 5: 13: Nihant et al.. J.
  • an initial protein comprising composition such as whole blood or plasma
  • a non-protein denaturant hydrogel under conditions sufficient for a substantial portion of the water present in the initial composition to be absorbed by the hydrogel.
  • the resultant protein rich phase is separated from the swollen hydrogel phase to produce the protein concentrate.
  • the subject methods find use in a variety of applications, particularly in the preparation of fibrinogen rich compositions from whole blood or derivatives thereof, where the resultant fibrinogen compositions find use in a variety of applications, as fibrin sealant tissue adhesives. as drug delivery vehicles, as sources of growth factors to promote wound healing, and the like.
  • FIG. 1 provides a schematic representation of the inventive method of preparing fibrinogen rich compositions.
  • Fig. 2 provides a schematic representation of the preparation of a drug loaded fibrin gels according to the subject invention.
  • vial A contains controlled release microspheres encapsulated with bioactive compounds and calcium salt.
  • Vial B contains all the components of a fibrin glue system (i.e. fibrinogen. thrombin etc.) except calcium, in a lyophilized form.
  • Vial A and B are reconstituted with sterile saline solution and used as a fibrin glue system.
  • a and B arc mixed and applied in situ in a surgical field using a suitable surgical device.
  • Fig. 3 provides a graphical representation of the water absorptive properties of a PEG 20.000 diacrylate hydrogel.
  • Fig. 4 provides a graphical representation of the protein concentration over time in a protein comprising aqueous composition contacted with a hydrogel according to the subject invention.
  • Fig. 5 provides a schematic representation of various polymeric crosslinking agents of the subject invention.
  • ( «w) represents a biodegradable component such as polyhydroxy acids, polylactones and their copolymers, or synthetic peptide sequences which are cleaved by enzymes inside the human body
  • (mm) represents a reactive functional group such as carbodiimiddazole, aldehyde, epoxide, n-hydroxysuccinimide and the like
  • ( — ) represents a biocompatible inert component. such as polyethylene glycol. dextran.
  • polyvinyl alcohol: (/Hum ) represents a copolymer of trimethylene carbonate and lactones or a synthetic peptide sequence which is cleavable by human enzymes.
  • Figs. 7(a) to (e) depicts a second embodiment of a device for producing protein concentrates from a physiological fluid according to the subject invention.
  • the blood composition prior to use, may be screened for the presence of one more pathogens, e.g. AIDS. Hepatitis B. etc.
  • One or more components may be added to the blood composition, such as anticoagulants, and the like.
  • the blood composition may be derived from a pooled supply of blood from a variety of different hosts, i.e. a pooled donor supply, or from the host in which the ultimate protein concentrate is to be empioyed. i.e. an autologous source.
  • a protein concentrate e.g. a fibrinogen rich composition
  • an initial blood composition as described above.
  • a protein concentrate e.g. a fibrinogen rich composition
  • the full scope of the invention also encompasses the preparation of protein concentrates from non-blood compositions, as described above.
  • the hydrogel with which the blood composition is combined in the subject methods may comprise one or more different hydrogels in combination, where usually no more than four different hydrogels. and more usually no more than three hydrogels will be used together, where a combination of different hydrogels may be employed for greater control over the types of components of the blood composition that are absorbed by the hydrogel and the nature of the resultant fibrinogen rich composition.
  • a hydrogel blend may be employed that combines a hydrogel with a first particular cut-off value, as described in greater detail below, and a second hydrogel that selectively removes a particular component of the initial fluid, e.g. albumin.
  • Hydrogels employed in the subject methods are compositions that are capable of absorbing water from an aqueous composition with which they are contacted to swell in size and increase in mass, where the increase in mass will typically be at least 10 and up to 1000 fold or greater than the dry mass of the hydrogel.
  • the hydrogels will be hydrogels which do not denature or modulate the conformation of proteins with which they come into contact, i.e. the hydrogels employed in the subject invention are non-protein denaturant hydrogels.
  • Preferred are hydrogels that are highly resistant to protein adsorption.
  • Hydrogels suitable for use in the subject methods will be biocompatible, by which is meant that they are suitable for contact with a blood derived composition that is to be introduced into a mammalian host. i.e. they will not leach toxic or unwanted substances into the blood composition upon contact.
  • the hydrogel will be one that does not substantially change the pH of the blood composition with which it is contacted and is sterile in nature.
  • the hydrogel composition that is employed may be in a variety of configurations, including particles, beads, rods, sheets, irregular shapes and the like, where those shapes with greater surface area to total mass ratios are preferred, at least in certain embodiments.
  • the porosity of the hydrogel which is dependent on the amount or degree of crosslinking present in the hydrogel. also affects the absorption characteristics of the hydrogel. For example, where absorption of water and small molecules is desired, a hydrogel with a high degree of crosslinking and a consequent small average porosity will be employed.
  • hydrogels that have a low degree of crosslinking and a consequent large average porosity will be employed.
  • hydrogels that have a molecular weight "cut-off absorption point i.e. a molecular weight limitation in excess of which components are not absorbed
  • cut-off absorption point i.e. a molecular weight limitation in excess of which components are not absorbed
  • hydrogels of particular interest are for use in certain embodiments are those having molecular weight cutoffs of: 100,000 daltons (where one desires to retain immunoglobulins); 150,000 daltons (where the removal of immunoglobulins is desired); 15.000 daltons (where one desires to retain growth factors in the protein concentrate); etc.
  • the hydrogel can be modified to provide for specific binding of one or more of the components of the blood composition to the surface of the hydrogel.
  • the hydrogel can be modified to comprise an albumin specific binding reagent, such as Cibacron blue, as described in Keogh & Eaton, J. Lab. & Clin. Med. (1994) 124:537.
  • an albumin specific binding reagent such as Cibacron blue
  • Other means of tailoring the hyrdogels to further control the nature of the protein concentrate produced by the subject method include using hydrogels comprising agents that act as water absorbents and/or precipitants, where such agents include ethanol. PEG 400, phosphate buffer and the like.
  • hydrogel compositions employed in the subject methods can be prepared by methods known to those skilled in the art. where specific methods for producing a number of different hydrogels which are suitable for use in the subject can be found in the Experimental Section infra.
  • suitable hydrogels or precursors thereof may be purchased from various commercial sources, where such sources include: Shearwater. BASF. Polysciences and the like.
  • hydrogels suitable for use in the subject methods are described in U.S. Patent No " s.: 5,626.863; 5,573,934; 5.567,435; 5,529,914; 5. 14,380; 5.476.909; 5.041 ,292. the disclosures of which are herein inco ⁇ orated by reference.
  • the hydrogels described in U.S. Patent No. 5,410,016 are the hydrogels described in U.S. Patent No. 5,410,016. the disclosure of which is herein inco ⁇ orated by interest.
  • Specific hydrogels of interest include: cross-linked poly(ethylene glycol diacrylate); cross-linked poly(ethylene oxide)-(polypropylene oxide)-poly(ethylene oxide) diacrylate; and the like.
  • the blood composition is contacted with the hydrogel under conditions sufficient for at least some of. and in many cases a substantial amount of. at least the water component of the blood composition to be absorbed from the blood composition into the hydrogel to produce a fibrinogen rich composition and a swollen hydrogel.
  • substantial amount of water is meant at least about 10 %. usually at least about 50 % and more usually at least about 90 %-95% of the initial water present in the blood composition.
  • Any convenient means of contacting the blood composition with the hydrogel may be employed, such as placing both components into the same container, mixing or agitating the two components to combine them, and the like.
  • the amount of hydrogel that is contacted with the blood composition will be sufficient to absorb the desired amount of water and other components from the blood composition.
  • a dry hydrogel composition Where a dry hydrogel composition is employed. 1 gram of dry hydrogel material will be used for fluid volume ranging from about 5 to 200 ml. usually from about 10 to 150 ml. For a partially hydrated hydrogel composition (where partially hydrated means a hydrogel composition comprising from about 20 to 50% polymer), one gram of hydrogel will be used for a volume of fluid ranging from about 1 to 10 ml. usually from about 2 to 3 ml. The two components will be maintained in contact for sufficient time for the desired amount of water (as well as other desired components) to be absorbed by the hydrogel. The amount of time for which contact is maintained between the hydrogel and the composition will vary depending on the particular nature of the hydrogel. i.e. composition, degree of hydration and surface area.
  • a longer period of time may be required when partially hydrated hydrogels are employed, where a shorter period of time may be sufficient when completely dehydrated and/or an excess of hydrogel is employed.
  • contact will be maintained for at least about 30 min and will generally not be maintained for more than 200 hours, where embodiments in which contact does not have to be maintained for more than about 48 hours are preferred.
  • the pH will be maintained at from 6.5 to 1 1. usually from about 6.5 to 7.5 and more usually from about 7 to 7.5.
  • the temperature of the abso ⁇ tion mixture will be maintained at from about 0 to 40. usually from about 0 to 30 and more usually from about 4 to 30 °C.
  • the subject methods of preparing protein concentrate compositions may be used in conjunction with one or more additional protein concentration and/or separation techniques, e.g. chromatography. precipitation with a precipitation agent, and the like, where such methods are known to those of skill in the art.
  • the process may further comprise the addition of one or more additional volumes of water to the protein concentrate following the initial abso ⁇ tion of water and components from the initial fluid. For example, a first volume of serum could be contacted with a hydrogel and reduced to 0.1 of its initial volume following abso ⁇ tion of water by the hydrogel. A volume of water could then be added to the reduced volume and abso ⁇ tion be allowed to occur for a second time. whereby further components are removed from the concentrate. This process could be repeated a plurality of times, as desired.
  • the fibrinogen rich phase is then removed from the swollen hydrogel to yield the subject fibrinogen rich composition.
  • Removal of the fibrinogen rich phase from the swollen hydrogel may be performed using any convenient technique or protocol.
  • the fibrinogen rich may be removed from the swollen hydrogel by aspirating the fibrinogen rich phase from the hydrogel surface.
  • the fibrinogen rich phase may be removed from the swollen hydrogel through centrifugation. which separates the two phases.
  • Other techniques that may be employed include: decanting, filtration, simpie mechanical separation, and the like. The above process may be repeated one or more additional times to obtain a final protein rich composition of desired characteristics. Separation yields a fibrinogen rich composition.
  • the resultant fibrinogen rich compositions may be substantially free of one or more low molecular weight components naturally present in the intial blood composition, such as albumin, growth factors, and the like.
  • the resultant fibrinogen rich compositions will be substantially free of components having a weight that does not exceed 100.000 daltons.
  • fibrinogen rich compositions with reduced albumin concentration i.e. compositions that have reduced albumin concentrations over that which would occur during the concentration process.
  • the subject fibrinogen rich composition may be used as produced above or modified through the addition of one or more different agents that modulate the chemical and/or physical nature of the composition.
  • Additional agents which may added to the composition include: proteins associated with coagulation, e.g. Factor II. fibronectin and the like; viscosity modifiers, such as collagen, sodium hyaluronate. and the like; antioxidants. such as hydroquinone. vitamin E. and the like; buffering agents, such as HEPES and the like; processing aids, antifibrinolytic agents, platelet activating agents, wound healing agents, and the like.
  • Visualization agents i.e. agents that may help a surgeon see those tissues to which the fibrin glue has been applied
  • specific visualization agents of interest are those that provide for color contrast with the background tissue, with blue and green being preferred colors, where specific agents include: indocyanine green, FD& C no. 1. FD & C no. 6. eosin. fluorescein. and the like.
  • the subject method of preparing the fibrinogen rich composition may be performed at both the laboratory scale and scaled up to produce large amounts of fibrinogen rich composition, e.g. from pooled plasma.
  • the process of the invention may be conducted using standard laboratory and medical equipment.
  • whole blood may be withdrawn from a mammalian host into a syringe containing an anticoagulant and emptied into a sterile centrifuge tube containing the absorbable hydrogel. e.g. hydrogel beads.
  • the hydrogel is then allowed to swell in the presence of whole blood for a period of time sufficient for the desired amount of protein concentration to occur, as described above.
  • the resultant concentrated plasma i.e.
  • fibrinogen rich phase is then separated from swollen hydrogel and other cellular material, e.g. red blood cells, by any convenient means, e.g. centrifugation and aspiration using a blunt needle.
  • swollen hydrogel and other cellular material e.g. red blood cells
  • any convenient means e.g. centrifugation and aspiration using a blunt needle.
  • Such laboratory scale processes find particular use in situations where one wishes to prepare an autologous tissue adhesive, e.g. where the adhesive is prepared from a patient ' s own blood prior to, or even during, a surgical operation.
  • pooled blood plasma which may be screened for viruses such as Hepatitis B. and AIDS, may be transferred to a batch reactor containing sterile absorbable hydrogel beads, e.g.
  • the subject fibrinogen rich compositions find use in a variety of applications, as components of tissue adhesives. in drug delivery vehicles, hemostatic agents, wound healing agents, and the like.
  • tissue adhesives the subject compositions will be used in combination with a coagulation component, usually a solution of thrombin and calcium ions, where upon combination of the composition with the coagulation component a fibrin sealant composition is produced which sets into a biocompatible proteinaceous matrix, i.e. a fibrin gel or sealant.
  • a coagulation component usually a solution of thrombin and calcium ions
  • fibrin sealants may be prepared from the subject compositions by applying them to a tissue repair site (i.e. a tissue site to which adherence of a second tissue is desired) either simultaneously or sequentially with a thro bin calcium ion setting composition, where suitable thrombin/calcium compositions are readily available from commercial sources and known to those of skill in the art.
  • tissue repair site i.e. a tissue site to which adherence of a second tissue is desired
  • a thro bin calcium ion setting composition where suitable thrombin/calcium compositions are readily available from commercial sources and known to those of skill in the art.
  • the two components described above may simply be applied sequentially or simultaneously to the tissue repair site via a needle or syringe or other application system. In certain embodiments, it is preferred to apply the components sequentially so as to "prime" the tissue, which results in improved tissue adhesive results.
  • a first component of the fibrin glue e.g. the crosslinker or thrombin.
  • the fibrinogen components which may include additional crosslinker/thrombin.
  • Fibrin sealants as prepared above with subject compositions find use as tissue adhesives in a variety of clinical applications, where such applications are reviewed in Schlag & Redl. Fibrin Sealant in Operative Surgery (1986) Vol. 1-7. and include: cardiovascular surgery, orthopaedic surgery, neurosurgery, ophthalmic surgery, general surgery and traumatology, plastic reconstruction and maxillofacial surgery, otorhinolaryngology. and the like.
  • the fibrin sealant may comprise a visualization agent (e.g.
  • the visualization agent including those described above, may be present in one or both of. but usually one of. the components of the fibrin sealant, i.e. it may be present in the fibrinogen rich component (where it may have been introduced to the fibrinogen rich composition after its production or introduced into the initial fluid from which the fibrinogen rich compositions is produced (with the amount used selected in view of the abso ⁇ tion of agent by the hydrogel)) and/or the thrombin calcium ion component.
  • the subject fibrinogen rich compositions according to the subject invention may also be used in a fibrin gel or sealant or matrix for biologically active agent or drug delivery.
  • Active agents of interest which may be delivered with fibrin sealant compositions prepared as described above include: proteins, carbohydrates, nucleic acids, and inorganic and organic biologically active molecules, where specific biologically active agents include but are not limited to: enzymes, antibiotics, antineoplastic agents, local anesthetics, hormones, antiangiogenic agents, antibodies. neurotransmitters. psychoactive drugs, drugs affecting reproductive organs, and oligoncucleotides such as antisense oligonucleotides.
  • the active agent or agents are present in a separate phase from the fibrin gei which protects the fibrin gel while it is setting from adverse effects of the active agent and/or modulates the release kinetics of the active agent from the gel.
  • Biodegradable vehicles in which the active agent may be present include encapsulation vehicles, such as microparticles, microspheres, microbeads, micropeliets and the like, where the ac e agent is encapsulated in a bioerodible or biodegradable polymer such as polyanhyd ⁇ de, polyglycolic acid, polylactic acid, polyorthocarbonate. polycaprolactone, polyt ⁇ methylene carbonate or their copolymers. caging or entrapping molecules, such as cyclodext ⁇ ns and the like, etc Biodcgradeable vehicle protected active agents are particularly preferred where the active agent is an antibiotic, such as gentamycin. tetracyicine. and the like
  • the amount of fibrin sealant and dosage of agent introduced into the host will necessarily depend upon the particular drug and condition being treated Administration may be by any convenient means, such as svnnge. cannula. trochar. and the like
  • Albumin rich compositions can be used in the preparation of albumin tissue adhesives or glues analogous to fibrin glues I or albumin glues, the albumin present in the albumin rich phase can be crosslinked using a number of different cross-linking means, including the use of chemical crosslinking agents, such as polyaldehydes. thermal crosslinking agents, such indocyanine green in combination with light (e g laser emitting at 780 nm). and the like. See U.S. Patent No. 5.583,1 14, the disclosure of which is herein inco ⁇ orated by reference.
  • a water soluble, biodegradeable synthetic cross-linkers will find use in the preparation of protein gels, such as fibrin glues and albumin glues mentioned above, or other compositions in which polymeric compounds are cross-linked, as described herein and in other applications known to those of skill in the art.
  • the water soluble, biodegradeable synthetic cross-linkers will be multi-functional, where by multifunctional is meant that the cross-linkers will be other than monofunctional, where illustrative multifunctional cross-linkers are difunctional, trifunctional, tetrafunctional. pentafunctional. hexafunctional.... up to "'n " '-functional. where n is an integer representing the number of different end cap functional regions present on the polymeric crosslinkers.
  • the water soluble, biodegradable polymeric crosslinkers will have a core that is a biologically inert polymeric unit; (b) an extension at each end of the core that is a biodegradable polymeric unit: and (c) an end cap on each extension that is a reactive moiety.
  • the biologically inert polymeric unit or region that makes up the core of the compound will be an inert polymeric block that is biocompatible. where the block may or may not be biodegradable.
  • the core is a polymeric region that is water soluble, where preferred polymers from which the core may be derived include: polvethers. e.g. polyakyleneoxides such as poly(ethylene glycol). poly( ethylene oxide). poly(ethylene oxide )-co-(poly(propyleneoxide)block copolymers: poly(vinyl alcohol); poly(vinyl pyrrolidone). poly(amino acids), poly(ethyloxzoline). dextran and the like, where polyethers. and more particularly polyoxyalkylenes.
  • the biodegradable extension component of the subject crosslinkers will be a component that degrades under physiological conditions into non-toxic products, where the biodegradable extension component will generally be hydrolyzable.
  • Hydrolyzable components of interest include: polymers, copolymers and oligomers of: glycolide. dl-lacide. d-lactide. 1-lactide. caprolactone. dioxanone and trimethylene carbonate or their copolymers and the like.
  • polylinkers with enzymatically hydrolyzable biodegradable extension components e.g. regions cleavable by metalloproteinases and collagenases.
  • biodegradable regions include polyhydroxyacids. polyorthocarbonates, polyanhyd ⁇ des. polylactones. polyaminoacids and polyphosphates and the like
  • the size or length of biodegradable region may be varied to in a number of ways For example, by using a crosslinker based on glycohde in the crosslinking of proteins, the resultant crosslinked protein degrades much faster than crosslinked proteins based on polycaprolactone cross- linkers Thus, by choosing appropriate biodegradable polymer regions in the crosslinker.
  • a suitable degradation profile of the resultant crosslinked protein can be obtained with difunctional cross-linkers, peptidic biodegradable regions are preferred
  • the reactive moiety that is the end-cap on each of the extensions is an activated functional group which provides for covalent bonding to proteins under in vivo conditions without free radical initiation
  • Such groups include carbodiimidazole. sulfonyl chloride, chlorocarbonates. n-hydroxysucc ⁇ n ⁇ md ⁇ l ester, succinimidyl ester, epoxides. aryl hahdes. sulfasuccinimidyl esters, maleimides.
  • FIG 5 provides further description of such polymeric cross-linkers
  • the biodegradable region of the crosslinker is represented by ( « * ⁇ « ⁇ * ).
  • the activated reactive end-group is represented by ( ⁇ » ), and the inert, biocompatible block is represented by (- )
  • Structure A shows a linear water soluble biodegradable polymer end-capped with two reactive functionalities like carbodiimidazole (CDI)
  • the linear water soluble region is a polyalkylene oxide, preferably polyethv lene gl> col which is extended with the biodegradable region which is a copolymer or homopolymer of tnmethylene carbonate This polymer is then terminated with CDI Structure B is a branched or star shaped t ⁇ functional biodegradable polymer which has inert polymer at the center
  • the inert polymer is extended with oligomeric biodegradable extensions which are then terminated by reactive functional end group Structures C and D show a multibranched
  • the resultant gel degrades due to hydrolysis of the biodegradable polymer such as polytrimethylene carbonate polymer inside the crosslinker.
  • the crosslinking density of the resultant network can be controlled by the overall molecular weight of the structure. A lower molecular weight such as 600 will give much higher crosslinking density as compared to a higher molecular weight crosslinker such a with molecular weight 10000 daltons.
  • the high molecular weight linear crosslinker is preferred in obtaining elastic gels.
  • the reaction between proteins and crosslinkers can be carried out directly on tissues and used as tissue glue.
  • the tertafuctional structures are obtained by reacting the polyalkylene oxide copolymer such as Tetronic 908 (obtained from BASF co ⁇ oration) with caprolactone in presence of stannous octoate.
  • the reaction is carried out in melt at 180 °C for 6 hours under nitrogen atmosphere.
  • the molar ratio of caprolactone to Tetronic 908 is kept to 12. which maintains water solubility of Tetronic 908-caprolactone copolymer in water.
  • the polymer is activated with CDI and used in crosslinking reaction with proteins.
  • the protein concentrates prepared by the subject methods also find use in the preparation of novel hydrogel wound dressings.
  • hydrogel wound dressings from the subject protein concentrates, such as the fibrinogen rich compositions discussed above, where the protein concentrate serves as the "aqueous composition component.”
  • the protein concentrates are combined with a macromonomer component and a cross-linking agent under conditions sufficient to produce a hydrogel comprising the protein concentrate composition interspersed throughout the cross-linked gel network.
  • Macromonomers that find use in the preparation of hydrogel wound dressings are non-toxic, water soluble, non-ionic macromonomers that comprise a polymerizable group, preferably an addition polymerizable group, where macromonomers which can be crosslinked via free radical polymerization are preferred.
  • water soluble polymers having unsaturated polymerizable groups such as acrylate. methacrylate, itaconate, and the like find use.
  • Water soluble polymers of interest which can be modified with unsaturated polymerizable groups include: polvethers. such as polyalkyleneoxide polymers and copolymers. polyethyleneoxide. polyethylene glycol. poiyethyleneoxide-polypropyleneoxide block, random or graft copolymers; polyvinyl alcohol; polyvinyl pyrrolidinone: and the like, where polyether polymers or derivatives thereof are particularly preferred as macromonomers. If desired, two or more macromonomers can be copolymerized to obtain suitable hydrogel properties.
  • other pharmaceutically acceptable catalysts and cocatylysts can be added to the macromonomer solution to accelerate the polymerization speed and/or to improve its shelf life.
  • small amounts of vinyl pyrrolidinone (concentration around 1 -10 micrometers per ml) can be added while using eosin- triethanol photoinitiating system.
  • Inhibitors such as hydroquinone may be added to prevent premature polymerization of macromonomer during its storage.
  • the wound dressings described may be prepared with bioactive compounds such as antibiotics to reduce the bacterial infection.
  • agents that may be included in the composition to modulate the ultimate properties of the hydrogel wound dressing prepared therefrom include proteinaceous polymers, such as collagen, other polymeric compounds, such as hyaluronic acid (including derivatives thereof), dextran and the like.
  • proteinaceous polymers such as collagen
  • other polymeric compounds such as hyaluronic acid (including derivatives thereof), dextran and the like.
  • hydrogel wound dressing composites which include, in addition to the hydrogel, at least one of collagen and hyaluronic acid, where in such composites.
  • PEG is the preferred hydrogel.
  • Hydrogel wound dressings produced in accordance with the present invention can be prepared with desired physical and chemical properties by choosing specific structural features, such as nature of polymerizable group of the macromonomer. the number of polymerizable groups present per macromonomer chain, the chain length and chemical structure of the macromonomer. where the modulation of such parameters to obtain a hydrogel wound dressing of desired characteristics is within the skill of those in the art.
  • kits for preparing the subject fibrinogen rich compositions kits for preparing the wound dressings and kits for preparing fibrin sealants from the subject fibrinogen rich compositions
  • Kits for preparing the fibrinogen rich compositions will comprise at least a a hydrogel composition and instructions for preparing the fibrinogen rich composition according to the methods of the subject invention, where the instructions may be present in the kit as an insert, inco ⁇ orated into the containers and/or packaging of the kit, and the like
  • Kits for preparing the fibrinogen rich compositions may further comprise anticoagulants, e g heparin.
  • kits for preparing a fibrin sealant according to the subject invention will comprise at least a fibrinogen rich composition prepared according to the subject invention and a thrombin calcium setting component present in separate containers
  • the kit mav further comprise a device for delivery the fibrin sealant to a tissue repair site as described above
  • the kits may also comprise an visualization agent and/or an active agent, preferably present in a biodegradable vehicle
  • kits for preparing fibrin sealants will also typically comprise instructions for preparing the fibrin sealant, which instructions mav be present as a package insert and/or associated with the containers present in the kit and/or packaging of the kit
  • the container will further comprise at least one opening for introducing the initial composition into the container and removing the resultant protein concentrate from the container.
  • the devices will comprise a contacting means for ensuring fluid transfer between the fluid introduced into the container and the hydrogel present therein.
  • the contacting means is a simple as a polymeric interface layer that is resistant to protein abso ⁇ tion adso ⁇ tion.
  • the contacting means may comprise a filtering means, such as a glass filter or membrane, which is impermeable to the hydrogel. Representative devices according to the subject invention are further described in terms of the figures. In Figure 6 is represented a first device embodiment according to the subject invention.
  • Device 10 is a plastic/glass bottle 12 (could also be a bag) containing hydrogel beads 14 separated from the remainder of the bottle interior by filter mesh 18 which is permeable to liquids but not to the hydrogel beads (i.e. the contacting means).
  • the bottle has a narrow neck 20 that has a volume measurement printed thereon 22.
  • plasma is added to the hydrogel beads and the swelling process is started.
  • the bottle is inverted every 10 minutes to separate the concentrated (the filter prevents the hydrogel from entering the narrow neck region. In the narro neck region (upon inversion), the volume of plasma is measured. When the desired volume is reached, the concentrated plasma is removed using a suitable technique.
  • Fig. 7 depicts a second embodiment of a device according to the subject invention.
  • Cylindrical device 30 has entry port 32 for the introduction of fluids into the device.
  • the entry port has a seal (e.g. silicon such as those employed on blood banking blood bags) through which fluids can be sterily introduced into the device or a removable lid (not shown) which can be used in an asceptic technique.
  • Areas 34 contain hydrogel which can absorb fluid and smaller molecules.
  • the hydrogel might have a molecular weight cutoff of about 80.000 so albumin would be absorbed into the hydrogel.
  • the hydrogel compartment would not change physical volume, but has room for the hydrogel to swell and expand. However, other embodiments in which the physical volume of the hydrogel compartment is expandable are envisioned.
  • Collection compartment 36 is a clyindrical shaped cup (though other shaped compartments are also contemplated. such as rectangular etc.) that has walls made of material that does not absorb protein and is also impermeable to fluid.
  • Exit port 38 allows for the sterile removal of fluid, i.e. a septum that can be punctured with a needle.
  • the top layer of the hydrogel compartment which is a material resistant to protein adso ⁇ tion/abso ⁇ tion (i.e. the contacting means).
  • the top layer could be fabricated from an appropriate polymeric material, e.g. PEG. or be a membrane which allows for passage of water and other small molecules, such as those used in the purification industry.
  • a membrane provides greater flexibility in the nature of the hydrogel that may be employed, as direct contact of the protein components of the fluid and the hydrogel can be prevented by the membrane. Accordingly, the hydrogel in such devices need not necessarily be a hydrogel that is non-denaturing, as that term is employed above.
  • Fig. 7(e) provides a top view of the device.
  • Figs. 7 (b) to 7 (d) show the device in use.
  • fluid 40 is introduced through port 32. Water and other small molecules are absorbed into hydrogel 34. As the fluid is absorbed, the level of the fluid falls until it reaches the top of the collection compartment 36. Concentrate is then removed from the collection compartment through exit 38.
  • the device is configured with dimensions chosen based on the intended nature of use. For example, in the preparation of a fibrinogen rich composition from plasma, in many situations the device will have dimensions sufficient to hold up to 1 unit of blood. Where desired, the entire device can be rotated at low ⁇ m (usually less than 500) to facilitate mixing of the fluid and reduction of fouling by proteins of the top or interface layer of the hydrogel or the membrane 39 covering the hydrogel component. Furthermore, coatings or active coatings or gel microspheres with active coatings could be placed on the periphery of the device (on the inside of the outside wall of the device above the hydrogel compartment) which would specifically absorb albumin, other proteins or specific cell types for cell purification.
  • the membrane 39 over the fluid absorbing component 34 is properly selected, a wider range of swelling polymers may be employed as the hydrogel component as mentioned above. Accordingly, polymer compositions such as those found in disposable diapers to absorb urine may be employed. Alternatively, the device could operate with hydrogel beads that are excluded from the collection cup 36 by having a means over the collection cup.
  • Polyethylene glycols ( Merck, mol. wt 20000 and BDH mol. wt 6000) were used as received.
  • Pluronic® and Tetronic® polyols were purchased from BASF co ⁇ oration.
  • Photoinitiator Darocure -W 2959 was purchased from Ciba Geigy.
  • the biodegradable polymers like polylactic acid, polyglycolic acid are purchased from Polysciences. All other reagents, solvents are of reagent grade and are purchased from commercial sources such as Fluka. Aldrich and Sigma. Small laboratory equipment was purchased from Fisher or Cole-Parmer.
  • PEG 10000 diacrylate is prepared by reacting 30 grams of PEG molecular weight 10.000 Da with 1.82 g of triethyl amine and 1.61 g of acryloyl chloride. Polyethyleneoxide (PEO) molecular weight 35.000 Da (30 g) is reacted with 0.52 g of triethyl amine and 0.46 g of acryloyl chloride to get a corresponding diacrylate derivative.
  • PEO polyethyleneoxide
  • Some polyethylene glycol based macromonomers may also be purchased using commercial sources such as Sartomer. Polysciences.
  • PEO content molecular weight 12500 daltons. purchased from BASF co ⁇ oration
  • the polymeric diol is then transferred to a 3 neck reaction flask equipped with nitrogen inlet and thermometer. 500 ml toluene. 3.3 ml of triethylamine amine and 1.9 ml of acryloyl chloride are added to the reaction mixture under dry nitrogen atmosphere. The reaction mixture is stirred overnight and filtered to remove triethylamine hydrochloride. The filtrate is added to 3000 ml hexane to precipitate the diacrylate derivative of Pluronic F127.
  • the macromonomer is purified by several dissolution-precipitation steps from THF-hexane solvent-nonsolvent system. Finally the diacrylate is dried under vacuum at 40 °C to a constant weight.
  • Other derivatives of Pluronics with different arrangement of PEO-PPO blocks and with different HLB values can also be acrylated in a similar manner.
  • the filtered sterile solution is filled into 50 ml plastic syringe (wrapped in aluminum foil for protection against light) with 22 gauge needle.
  • a 100 ml of sterile mineral oil which is constantly being stirred using a magnetic stir bar is exposed to green light source (American Argon ion laser. Model 905 emitting at 532 nm. 100 mW/c ⁇ r).
  • green light source American Argon ion laser. Model 905 emitting at 532 nm. 100 mW/c ⁇ r.
  • the macromonomer solution is flushed out of needle and the droplets are collected into sterile mineral oil which is under green light irradiation. After 5 minutes of irradiation of monomer solution to green light, the photopolymerized hydrogel beads are separated from mineral oil by filtration.
  • hydrogel beads are further washed with sterile hexane to remove traces of mineral oil.
  • the hydrogels beads are then stored in sterile saline solution for 5 hours to remove initiator fragments, unreacted or uncrosslinked macromonomer etc.
  • the washed beads are then isolated by filtration and lyophilized under sterile conditions.
  • thermosensitive macromonomer 30 g of Tetronic 908 polyol is dissolved in 400 ml dry benzene. 100 ml of benzene is distilled to remove traces of water from the polyol. The solution is cooled to 30 "C and 1.45 g triethylamine and 1 .90 g acryloyl chloride are added. The reaction mixture is refluxed for 1 h under argon atmosphere. It is then cooled and then filtered to removed triethylamine hvdrochloride. The filtrate is then added to 2000 mi hexane to precipitate the polymer. The polymer is purified by several precipitations from THF- hexane solvent-nonsolvent system. Further solvent removal/drying is achieved by vacuum drying overnight at 60 °C.
  • FIG. 1 Preparation with Hydrogel Beads Blood plasma is concentrated with hydrogel beads using a method analogous to that described in example DI . above. The process is shown schematically in Fig. 1.
  • dry or partially dry hydrogel beads are placed in a sterile tube as shown in Fig. 1 A.
  • a blood plasma solution is transferred to the tube as shown in Fig. B.
  • the mixture is allowed to remain in contact for a sufficient period of time for the hydrogel beads to selectively absorb water from the plasma, whereby a fibrinogen rich composition is produced, as shown in Fig. IC.
  • a small amount of plasma is taken out periodically and is analyzed for total protein concentration using a biurette method, the results of which are shown in Fig. 4.
  • polyethylene glycol having a molecular weight 2000 is dried at 90- 100 °C in a glass sealing tube.
  • the tube is then cooled and transferred inside an air bag where 12.24 g of trimethylene carbonate and 50 mg of stannous octoate are added to the tube.
  • the glass tube is then sealed under nitrogen atmosphere and heated with stirring at 155 °C and maintained at this temperature for 6 hours.
  • the polyethylene glycol-polytrimethylene carbonate polymer is cooled and recovered by breaking the giass sealing tube. It is further purified by several precipitations from toluene-hexane solvent-nonsolvent system.
  • the product is dried in vacuum at 40 °C and used immediately in the activation reaction.
  • Tetronic 908-caprolactone polyol( PCLP)-30 g of Tetronic 908 is charged in a dry 3 neck flask equipped with magnetic stirrer and vacuum inlet. The flask is then heated in a silicone oil bath at 100 °C for 12 hours to dry the Tetronic 908. The flask is cooled to room temperature and 1 .642 g of caprolactone and 0.02 g of stannous 2-ethylhexanoate are added to the flask. The tlask is heated to 180 U C for 6 hours under nitrogen atmosphere. The reaction is product is then dissolved in 200 ml dry toluene (warming of toluene accelerates dissolution).
  • the toluene solution is added to 2,000 ml dry heptane with constant stirring.
  • the product is isolated by filtration. Further purification is accomplished by precipitation of toluene solution of PCLP in heptane.
  • the product is dried in vacuum at 40 U C and used immediately in the activation reaction.
  • Tetronic 908-caprolactone copolymer 30 g is Tetronic 908-caprolactone copolymer is dissolved in 400 ml dry benzene. About 100 ml of benzene is distilled off and the solution is cooled to 50 °C. 2.24 g of carbodiimidazole is added to reaction mixture . The mixture is refluxed for 2 h under nitrogen atmosphere . At the end of 2 hour period, the solution cooled added to 4,000 ml hexane to precipitate the polymer. It is further purified by repeated (3 times) precipitation from toluene-hexane system. The polymer is dried under vacuum at 40 °C.
  • Tetronic 908-caprolactone succinate prepared by method described above is dissolved in 100 ml dry methylene chloride.
  • the mixture is cooled to 0 °C in ice bath and 0.5 g of 4-dimthylaminopyridine and 1 g of dicyclohexylcarbodiimide (DCC) are added.
  • DCC dicyclohexylcarbodiimide
  • the mixture is stirred at 0 °C for 6 hour and filtered.
  • the filtrate is then added to 2.000 ml dry hexane to precipitate the activated succinimydyl ester.
  • the product is isolated by filtration, dried under vacuum and stored under argon at 4 "C.
  • the CDI/succinimydil activated crosslinker polymers as prepared above are dissolved in aqueous buffer solution and reacted with albumin rich solutions to form a gel.
  • PEG 10.000 diacrylate synthesized by a procedure described in example Bl . is dissolved in 20 grams of phosphate buffer solution ( pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH : PO , 8.0 g/L of NaCl and 1.15 g/L Na,HPO,).
  • phosphate buffer solution pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH : PO , 8.0 g/L of NaCl and 1.15 g/L Na,HPO,).
  • a photoinitiator solution 300 mg of Darocur® 2959, Ciba Geigy, dissolved in 700 mg ethanol
  • the aqueous macromonomer solution containing photoinitiator is sterile filtered using 50 ml syringe and 0.2 mm syringe filter. About 10-12 ml of sterile solution is transferred into transparent plastic mold (10 cm x 5 cm x 2 mm). The solution is then exposed to long wavelength UV light (Blak Ray light source, model 3-100A. Flood 365 nm. intensity 30 mW/cm 2 ) for 5 minutes. The polymerized, flexible gel is removed from the mold. This sterile gel can be directly applied over the wound.
  • PEG 10.000 diacrylate synthesized by a procedure described in example . is dissolved in 12 grams of phosphate buffer solution ( pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH.PO,, 8.0 g/L of NaCl and 1.15 g/L NaTlPO,).
  • phosphate buffer solution pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH.PO,, 8.0 g/L of NaCl and 1.15 g/L NaTlPO,).
  • photoinitiator solution 300 mg of Darocur® 2959. Ciba Geigy, dissolved in 700 mg ethanol
  • the aqueous macromonomer solution containing photoinitiator is sterile filtered using 50 ml syringe and 0.2 mm syringe into a sterile amber colored 25 ml glass bottle.
  • the solution is lyophilized while maintaining sterility to remove water.
  • the vial is capped with a sterile rubber septum.
  • the lyophilized macromonomer powder containing photoinitiator is used in making 'in situ' formable wound dressings. 4 Autologus or single donor single donor blood or blood components encapsulated wound dressing
  • Platelets may be activated using thrombin or suitable activating agent before or after the encapsulation process.
  • the activated platelets release platelets derived growth factors which are then released in controlled manner by the hydrogel matrix.
  • a sterile amber colored glass vial containing 3 grams macromonomer and photoinitiator (similar to described in example 6) is dissolved in 12 ml tissue culture medium (for example Dulbecco's modification of Eagle's medium, a medium suitable for culturing human foreskin fibroblasts).
  • tissue culture medium for example Dulbecco's modification of Eagle's medium, a medium suitable for culturing human foreskin fibroblasts.
  • the macromonomer solution in tissue culture medium is then sterile filtered into a plastic mold of desired shape for example 10 cm x 5 cm x 2 mm size and exposed to long UV light as mentioned in previous examples.
  • the polymerized hydrogel is then removed from the mold and used as wound dressing.
  • PEG diacrylate synthesized by a procedure described in example BI . is dissolved in 12 grams of phosphate buffer solution ( pH 7.2. 0.2 g/L KCl. 0.2 g/L of KH.POj, 8.0 g/L of NaCl and 1.15 g/L Na.HPO, and 0.5% sodium hyaluronate) .
  • phosphate buffer solution pH 7.2. 0.2 g/L KCl. 0.2 g/L of KH.POj, 8.0 g/L of NaCl and 1.15 g/L Na.HPO, and 0.5% sodium hyaluronate
  • photoinitiator solution 300 mg of Darocur® 2959. Ciba Geigy, dissolved in 700 mg ethanol
  • the aqueous macromonomer solution containing photoinitiator and hyaluronic acid is sterile filtered using 50 ml syringe and 0.2 ⁇ m syringe filter. About 10-12 mL of sterile solution is transferred into transparent plastic mold (cavity size 10 cm x 5 cm x 2 mm). The solution is then exposed to long wavelength UV light (Blak Ray light source, model 3-100A, Flood 365 nm. intensity 30 mW/cnr) for 5 minutes. The polymerized, flexible gel is removed from the mold. This sterile gel can be directly applied over the wound.
  • 100 mg of sterile collagen lyophilized powder is then added to the macromonomer solution.
  • the dispersion is well mixed and then transferred into transparent plastic mold (cavity size 10 cm x 5 cm x 2 mm).
  • the dispersion is then exposed to long wavelength UV light (Blak Ray light source, model 3- 100A. Flood 365 nm. intensity 30 mW/cnr) for 5 minutes.
  • the polymerized, flexible gel is then removed from the mold and used as poiyalkyleneoxide-collagen hydrogei wound dressing.
  • thermosensitive macromonomer 10 grams of thermosensitive macromonomer (synthesized as described in example
  • the filtered cold solution is directly applied over wound surface or injected into a wound cavity
  • the cold solution conforms to the wound geometry or cavity and the body temperature causes physical gelation of the macromonomer
  • the physically gelled solution is then irradiated with long UV light to crosslink the macromonomer solution
  • the light induces chemical crosslinking and forms a chemically crosslinked hydrogel which has good absorptive and mechanical properties
  • the polymerized gel is also non adherent to the tissue and can be easily removed from the wound site

Abstract

Methods and compositions are provided for preparing protein concentrates from protein comprising aqueous compositions. In the subject methods, an initial protein comprising aqueous composition, such as whole blood or a derivative thereof, is contacted with a non-protein denaturant hydrogel under conditions sufficient for a substantial amount of water present in the composition to be absorbed by the hydrogel, resulting in the production of a protein concentrate, such as a fibrinogen rich composition. Of particular interest is the use of the subject methods to prepare fibrinogen rich compositions, where such compositions produced according to the subject invention are useful in fibrin sealants, drug delivery vehicles and in a number of other diverse applications.

Description

METHODS AND DEVICES FOR PREPARING PROTEIN CONCENTRATES
CROSS-REFERENCE TO RELATED APPLICATIONS This application is a continuation-in-part of U.S. application serial no.:
60/026.536 filed September 23. 1996; U.S. application serial no. : 60/039.904 filed March 4. 1997 and U.S. application serial no.: 60/040.417 filed March 13. 1997, the disclosures of which are herein incoφorated by reference.
INTRODUCTION
Technical Field
The technical field of this invention is the preparation of concentrated protein compositions. Background of the Invention Methods of preparing concentrated protein compositions from initial dilute protein compositions find use in a variety of different industries, including the chemical, biological, academic research, biotechnological and medical industries. For example. "Fibrin Sealants" (also known as fibrin gels or fibrin glues) are a type of blood derived composition used in the medical industry which are prepared through methods of concentrating blood plasma proteins that have been developed for use as tissue adhesives. drug delivery vehicles and the like, .although such compositions are not yet FDA approved in the United States due to concerns over blood borne contaminants, such compositions are marketed in Europe and elsewhere throughout the world. A typical commercial fibrin glue kit consists of a iai ol Kophili/ed concentrated human fibrinogen. prepared from pooled human donor blood, that also contains fibronectin. Factor XIII and reduced amounts of piasminogcn The concentrate, also known as cryoprecipitate. is reconstituted with a reconstituting solution and warmed to 37 °C The second component of the adhesive system is a lyophilized bovine thrombin solution which is reconstituted with a calcium chloride solution The formulation may also contain additional components like a fibnonolysis inhibitor The reconstituted solutions are mixed and used as a surgical adhesive system The most common method used for the preparation of the fibrinogen component of the above described kits is cryoprecipation In cryoprecipitation. fresh blood plasma is frozen at -80 °C for at least 6 to 12 h The temperature of the frozen plasma is then raised to around 0-4 °C. resulting in the formation of a precipitated supernatant that contains fibπnogen and Factor XIII, i e a cryoprecipitate The cryoprecipitated suspension is then recovered. Another method described in the literature is the use of common non-toxic organic/inorganic compounds such as ethanol, polyethylene glycol. poly(vιnyl alcohol). 1 -6-hexanoιc acid and ammonium sulfate as precipitating agents
The above methods of preparing the fibrinogen containing component of fibrin glue compositions are time consuming and complex Furthermore, in approaches such as cryoprecipitation. special equipment like a refrigerated centrifuge, is often required Finally, different methods of precipitation produce fractions with different adhesive and physical characteristics which can adversely affect the ultimate adhesive product Accordingly, there is a continued need for the development of new methods for preparing concentrated protein compositions, and particularly fibrinogen rich fractions from blood compositions Ideally, such methods would be relatively simple and rapid, require minimal handling of the plasma and not include a cryoprecipitation step, and provide serum concentrates suitable tor use in fibrin glue s\ stems, in wound healing promotion systems, in drug delivery, and in tissue regeneration 1-urthermore. such methods would ideally be suitable tor use in the preparation ot autologous serum concentrates that eliminate pathogen transmission risk present in serum concentrates prepared from pooled donor sources Also of interest would be the development of a simple method capable of efficiently producing concentrated protein compositions from large volumes of initial fluid, e.g. pooled human serum in emergency surgery situations. Also of interest would be the development of devices for use in performing the subject methods. Relevant Literature
Fibrin sealants and methods for their production, as well as clinical applications thereof, are reviewed in David H. Sierra. "'Fibrin Sealant Adhesive
Systems: A Review of Their Chemistry, Material Properties and Clinical
Applications." J. Biomaterials Applications ( 1993) 7:309-352. Other references of interest include: Sierra & Feldman. J. Applied Biomaterials (1992) 3:147-151 : U.S.
Pat. No. 5,405,607: U.S. Pat. No. 5.030,215: and U.S. Pat. No. 5,395.923.
Devices for preparing and administering a fibrin sealant to facilitate tissue repair are described in: US Pat. No. 4.874.368; US Pat. No.4.631.055; US Pat.
No.4.735.616; US Pat. No.4,359.049; US Pat. No.4.978.336: US Pat. No.5.1 16.315; US Pat. No.4.902.281 : US Pat. No.4.932.942; WO 91/09641. and Tange. R.A.. Fibrin
Sealant in Operative Medicine: Otolaryngology-Vol. 1 ( 1986).
Microencapsulated drug particles and similarly protected pharmaceutically active agents are described in: Kissel et al.. J. Controlled Release ( 1991 ) 16:27; Tabata et al.. Pharmaceutical Research (1993) 10:487: EP A 83303606.4; U.S. Patent No. 5.143.662: Mathiowitz & Langer. J. Controlled Release ( 1987) 5: 13: Nihant et al.. J.
Colloid & Interface Science (1995) 173:55: and Irwin et al. Pharmaceutical Research
( 1994) 1 1 : 1968.
Hydrogels and methods for their preparation are reported in: U.S. Patent Nos.:
5.626.863; 5,573.934: 5.567.435; 5,410.016: 5.529.914; 5.514.380; 5.476.909; 5.041.292. 5,583.1 14: as well as in Walter et al.. J. Macromol. Sci.-Phys. ( 1994) B33
(3&4):267; Pathak et al.. J. Am. Chem. Soc. (1992) 1 14: 831 1 : Sawhney et al..
Macromolecules (1993) 26:581 : Keogh & Eaton. J. Laboratory & Clinical Med.
( 1994) 124:4:537; and Reddi et al.. Macromolecular Reports ( 1995) A32:789.
SUMMARY OF THE INVENTION
Methods and devices for the preparation of protein concentrates, as well as novel protein concentrate compositions prepared thereby, products derived therefrom
-j - and applications in which the products find use. are provided. In the subject methods, an initial protein comprising composition, such as whole blood or plasma, is contacted with a non-protein denaturant hydrogel under conditions sufficient for a substantial portion of the water present in the initial composition to be absorbed by the hydrogel. Following absorption, the resultant protein rich phase is separated from the swollen hydrogel phase to produce the protein concentrate. The subject methods find use in a variety of applications, particularly in the preparation of fibrinogen rich compositions from whole blood or derivatives thereof, where the resultant fibrinogen compositions find use in a variety of applications, as fibrin sealant tissue adhesives. as drug delivery vehicles, as sources of growth factors to promote wound healing, and the like.
BRIEF DESCRIPTION OF THE FIGURES Fig. 1 provides a schematic representation of the inventive method of preparing fibrinogen rich compositions. Fig. 2 provides a schematic representation of the preparation of a drug loaded fibrin gels according to the subject invention. In this figure, vial A contains controlled release microspheres encapsulated with bioactive compounds and calcium salt. Vial B contains all the components of a fibrin glue system (i.e. fibrinogen. thrombin etc.) except calcium, in a lyophilized form. Vial A and B are reconstituted with sterile saline solution and used as a fibrin glue system. A and B arc mixed and applied in situ in a surgical field using a suitable surgical device.
Fig. 3 provides a graphical representation of the water absorptive properties of a PEG 20.000 diacrylate hydrogel.
Fig. 4 provides a graphical representation of the protein concentration over time in a protein comprising aqueous composition contacted with a hydrogel according to the subject invention.
Fig. 5 provides a schematic representation of various polymeric crosslinking agents of the subject invention. In this figure. («w) represents a biodegradable component such as polyhydroxy acids, polylactones and their copolymers, or synthetic peptide sequences which are cleaved by enzymes inside the human body; (mm) represents a reactive functional group such as carbodiimiddazole, aldehyde, epoxide, n-hydroxysuccinimide and the like; ( — ) represents a biocompatible inert component. such as polyethylene glycol. dextran. polyvinyl alcohol: (/Hum ) represents a copolymer of trimethylene carbonate and lactones or a synthetic peptide sequence which is cleavable by human enzymes.
Fig. 6 depicts a first embodiment of a device for producing protein concentrates from a physiological fluid according to the subject invention.
Figs. 7(a) to (e) depicts a second embodiment of a device for producing protein concentrates from a physiological fluid according to the subject invention.
DESCRIPTION OF THE SPECIFIC EMBODIMENTS Methods and devices for the preparation of protein concentrates, as well as novel protein concentrate compositions prepared thereby and products derived therefrom, are provided. In the subject methods, an initial protein comprising composition, such as whole blood or plasma, is contacted with a non-protein denaturant hydrogel under conditions sufficient for a substantial portion of the water present in the initial composition to be absorbed by the hydrogel. Following absorption, the resultant protein rich phase is removed from the swollen hydrogel to produce the protein concentrate, where the term "removed" is employed in a broad sense to mean that the swollen hydrogel and the protein rich phase are isolated from one another, where removal can be accomplished via active separation, e.g. by centrifugation, or passively, e.g. in those embodiments where the hydrogel and protein rich phase separate by themselves, where the protein rich phase is subsequently isolated from the hydrogel through simple aspiration or other analogous technique. By selecting an appropriate hydrogel. the nature of the resultant protein concentrate may be controlled. The subject methods find use in a variety of applications, particularly in the preparation of fibrinogen rich compositions from whole blood or derivatives thereof (where the initial blood composition may be a pooled donor source or an autologous source, where the resultant fibrinogen compositions find use in a variety of applications, fibrin sealant tissue adhesives. drug delivery vehicles, and the like. In further describing the subject invention, the methods for preparing the subject protein concentrates will first be described in greater detail, followed by a discussion of the resultant concentrates themselves, representative applications in which they find use and kits and devices suitable for use in their preparation. Before the subject invention is further described, it is to be understood that the invention is not limited to the particular embodiments of the invention described below, as variations of the particular embodiments may be made and still fall within the scope of the appended claims. It is also to be understood that the terminology employed is for the purpose of describing particular embodiments, and is not intended to be limiting. Instead, the scope of the present invention will be established by the appended claims.
It must be noted that as used in this specification and the appended claims, the singular forms "a.'* ς"an." and "'the" include plural reference unless the context clearly dictates otherwise. Unless defined otherwise all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs.
In practicing the subject methods, an initial protein comprising composition is contacted with a hydrogel under conditions sufficient for at least a portion of at least the water present in said blood composition to be absorbed by the hydrogel. The initial protein containing composition may be any aqueous composition that comprises one or more proteins of interest, where such compositions include both naturally occurring compositions, such as physiologically derived fluids, e.g. blood, plasma, urine, cerebrospinal fluid, tears, saliva, milk, peritoneal cavity fluid and the like; and synthetically prepared compositions, e.g. tissue culture medium, and the like.
Physiological fluids of interest may be obtained from a variety of hosts, including cows, sheep, pigs. deer, humans and the like. For example, the subject methods can be used to produce enriched protein compositions from cow or sheep milk, where the cow or sheep may be a transgenic animal engineered to produce milk containing a recombinant protein of interest.
Of particular interest is the preparation of protein concentrates from initial blood compositions. The blood composition employed in the subject methods will typically be derived from a mammalian source, where suitable sources include cows, sheep, pigs. deer, humans and the like, where humans may be the preferred source depending on the intended use of the composition. The blood composition may be whole blood or a blood product, i.e. a whole blood derivative, where whole blood is subjected to one or more filtration steps, e.g. removal of red blood cells, and the like. In other words, blood compositions that may be used in the subject invention include both whole blood and blood derivatives, e.g. plasma, platelet containing serum, and the like, where such blood compositions may be obtained from their hosts and produced according to methods known in the art. The blood composition, prior to use, may be screened for the presence of one more pathogens, e.g. AIDS. Hepatitis B. etc. One or more components may be added to the blood composition, such as anticoagulants, and the like. The blood composition may be derived from a pooled supply of blood from a variety of different hosts, i.e. a pooled donor supply, or from the host in which the ultimate protein concentrate is to be empioyed. i.e. an autologous source.
For clarity and ease of understanding, the invention is further described below in terms of the preparation of a protein concentrate, e.g. a fibrinogen rich composition, from an initial blood composition as described above. However, in so describing the invention, it should be understood that the full scope of the invention also encompasses the preparation of protein concentrates from non-blood compositions, as described above.
The hydrogel with which the blood composition is combined in the subject methods may comprise one or more different hydrogels in combination, where usually no more than four different hydrogels. and more usually no more than three hydrogels will be used together, where a combination of different hydrogels may be employed for greater control over the types of components of the blood composition that are absorbed by the hydrogel and the nature of the resultant fibrinogen rich composition. For example, a hydrogel blend may be employed that combines a hydrogel with a first particular cut-off value, as described in greater detail below, and a second hydrogel that selectively removes a particular component of the initial fluid, e.g. albumin. Hydrogels employed in the subject methods are compositions that are capable of absorbing water from an aqueous composition with which they are contacted to swell in size and increase in mass, where the increase in mass will typically be at least 10 and up to 1000 fold or greater than the dry mass of the hydrogel. Importantly, the hydrogels will be hydrogels which do not denature or modulate the conformation of proteins with which they come into contact, i.e. the hydrogels employed in the subject invention are non-protein denaturant hydrogels. Preferred are hydrogels that are highly resistant to protein adsorption.
Hydrogels suitable for use in the subject methods will be biocompatible, by which is meant that they are suitable for contact with a blood derived composition that is to be introduced into a mammalian host. i.e. they will not leach toxic or unwanted substances into the blood composition upon contact. Preferably, the hydrogel will be one that does not substantially change the pH of the blood composition with which it is contacted and is sterile in nature.
Suitable hydrogels include macromolecular and polymeric materials into which water and small molecules can easily diffuse and include hydrogels prepared through the cross linking, where crosslinking may be either through covalent. ionic or hydrophobic bonds introduced through use of either chemical cross-linking agents or electromagnetic radiation, such as ultraviolet light, of both natural and synthetic hydrophilic polymers, including homo and co-polymers. Hydrogels of interest include those prepared through the cross-linking of: polvethers. e g. polyakyleneoxides such as poly(ethylene glycol). poly(ethylene oxide). poly(ethylene oxide)-co- (poly(propyleneoxide)block copolymers; poly(vinyl alcohol); poly(vinyl pyrrolidone); polysaccharides, e.g. hyaluronic acid, dextran. chondroitin sulfate. heparin. heparin sulfate or alginate; proteins, e.g. gelatin, collagen, albumin, ovalbumin or polyamino acids: and the like. Because of their high degree of biocompatibility and resistance to protein adsorption, polyether dervived hydrogels are preferred, with poly(ethylene glycol) derived hydrogels being particularly preferred.
Physical characteristics such as size, shape and surface area can affect the absorption characteristics of the hydrogel composition. Accordingly, the hydrogel composition that is employed may be in a variety of configurations, including particles, beads, rods, sheets, irregular shapes and the like, where those shapes with greater surface area to total mass ratios are preferred, at least in certain embodiments. The porosity of the hydrogel. which is dependent on the amount or degree of crosslinking present in the hydrogel. also affects the absorption characteristics of the hydrogel. For example, where absorption of water and small molecules is desired, a hydrogel with a high degree of crosslinking and a consequent small average porosity will be employed. Conversely, where the selective absorption of small proteins is also desired, a hydrogel with a low degree of crosslinking and a consequent large average porosity will be employed. Of interest for certain embodiments of the subject invention, e.g. where the selective removal of albumin and other components of similar or lower molecular weight is desired, are hydrogels that have a molecular weight "cut-off absorption point (i.e. a molecular weight limitation in excess of which components are not absorbed) of about 80,000 daltons. Other hydrogels of particular interest are for use in certain embodiments are those having molecular weight cutoffs of: 100,000 daltons (where one desires to retain immunoglobulins); 150,000 daltons (where the removal of immunoglobulins is desired); 15.000 daltons (where one desires to retain growth factors in the protein concentrate); etc.
To further tailor the absorptive properties of the hydrogel, the hydrogel can be modified to provide for specific binding of one or more of the components of the blood composition to the surface of the hydrogel. For example, where the selective absorption of albumin in addition to water is desired, the hydrogel can be modified to comprise an albumin specific binding reagent, such as Cibacron blue, as described in Keogh & Eaton, J. Lab. & Clin. Med. (1994) 124:537. Other means of tailoring the hyrdogels to further control the nature of the protein concentrate produced by the subject method include using hydrogels comprising agents that act as water absorbents and/or precipitants, where such agents include ethanol. PEG 400, phosphate buffer and the like.
The hydrogel compositions employed in the subject methods can be prepared by methods known to those skilled in the art. where specific methods for producing a number of different hydrogels which are suitable for use in the subject can be found in the Experimental Section infra. Alternatively, suitable hydrogels or precursors thereof may be purchased from various commercial sources, where such sources include: Shearwater. BASF. Polysciences and the like.
Specific hydrogels suitable for use in the subject methods are described in U.S. Patent No"s.: 5,626.863; 5,573,934; 5.567,435; 5,529,914; 5. 14,380; 5.476.909; 5.041 ,292. the disclosures of which are herein incoφorated by reference. Of particular interest in many embodiments of the subject invention are the hydrogels described in U.S. Patent No. 5,410,016. the disclosure of which is herein incoφorated by interest. Specific hydrogels of interest include: cross-linked poly(ethylene glycol diacrylate); cross-linked poly(ethylene oxide)-(polypropylene oxide)-poly(ethylene oxide) diacrylate; and the like.
The blood composition is contacted with the hydrogel under conditions sufficient for at least some of. and in many cases a substantial amount of. at least the water component of the blood composition to be absorbed from the blood composition into the hydrogel to produce a fibrinogen rich composition and a swollen hydrogel. By substantial amount of water is meant at least about 10 %. usually at least about 50 % and more usually at least about 90 %-95% of the initial water present in the blood composition. Any convenient means of contacting the blood composition with the hydrogel may be employed, such as placing both components into the same container, mixing or agitating the two components to combine them, and the like. The amount of hydrogel that is contacted with the blood composition will be sufficient to absorb the desired amount of water and other components from the blood composition. Where a dry hydrogel composition is employed. 1 gram of dry hydrogel material will be used for fluid volume ranging from about 5 to 200 ml. usually from about 10 to 150 ml. For a partially hydrated hydrogel composition (where partially hydrated means a hydrogel composition comprising from about 20 to 50% polymer), one gram of hydrogel will be used for a volume of fluid ranging from about 1 to 10 ml. usually from about 2 to 3 ml. The two components will be maintained in contact for sufficient time for the desired amount of water (as well as other desired components) to be absorbed by the hydrogel. The amount of time for which contact is maintained between the hydrogel and the composition will vary depending on the particular nature of the hydrogel. i.e. composition, degree of hydration and surface area. For example, a longer period of time may be required when partially hydrated hydrogels are employed, where a shorter period of time may be sufficient when completely dehydrated and/or an excess of hydrogel is employed. Usually contact will be maintained for at least about 30 min and will generally not be maintained for more than 200 hours, where embodiments in which contact does not have to be maintained for more than about 48 hours are preferred. In the mixture (i.e. absoφtion mixture) into which the two components are combined in the contacting step, the pH will be maintained at from 6.5 to 1 1. usually from about 6.5 to 7.5 and more usually from about 7 to 7.5. The temperature of the absoφtion mixture will be maintained at from about 0 to 40. usually from about 0 to 30 and more usually from about 4 to 30 °C. In addition to the blood composition and hydrogel, the absoφtion mixture may further comprise one or more additional agents which serve a variety of puφoses. Such agents include: anticoagulants, such as heparin. buffering agents, e.g. citrate buffer. HEPES, and the like. Absoφtion of water and other components, as described above, by the hydrogel in the absoφtion mixture results in the production of a fibrinogen rich phase and a swollen hydrogel. The resultant volume of protein concentrate, e.g. fibrinogen rich phase, will usually be less than 0.5. usually less than about 0.25 and more usually less than about 0.1 of the initial protein comprising composition. The subject methods of preparing protein concentrate compositions may be used in conjunction with one or more additional protein concentration and/or separation techniques, e.g. chromatography. precipitation with a precipitation agent, and the like, where such methods are known to those of skill in the art. In certain embodiments of the subject invention, the process may further comprise the addition of one or more additional volumes of water to the protein concentrate following the initial absoφtion of water and components from the initial fluid. For example, a first volume of serum could be contacted with a hydrogel and reduced to 0.1 of its initial volume following absoφtion of water by the hydrogel. A volume of water could then be added to the reduced volume and absoφtion be allowed to occur for a second time. whereby further components are removed from the concentrate. This process could be repeated a plurality of times, as desired.
The fibrinogen rich phase is then removed from the swollen hydrogel to yield the subject fibrinogen rich composition. Removal of the fibrinogen rich phase from the swollen hydrogel may be performed using any convenient technique or protocol. For example, the fibrinogen rich may be removed from the swollen hydrogel by aspirating the fibrinogen rich phase from the hydrogel surface. Alternatively, the fibrinogen rich phase may be removed from the swollen hydrogel through centrifugation. which separates the two phases. Other techniques that may be employed include: decanting, filtration, simpie mechanical separation, and the like. The above process may be repeated one or more additional times to obtain a final protein rich composition of desired characteristics. Separation yields a fibrinogen rich composition. The fibrinogen rich composition, because of the method by which it is produced, will be more concentrated in protein than the initial composition, where in certain embodiments the total protein concentration will be about 10 times greater, and in many embodiments the total protein concentration will be about 5 to 9 times greater than the protein concentration of the initial composition. The concentration of fibrinogen in the fibrinogen rich composition will range from about 1 to 100. usually from about 1 to 30 and more usually from about 1 to 2 mg/ml. where in those embodiments in which the fibrinogen rich composition is to be employed in a fibrin glue system, the fibrinogen concentration will range from about 30 to 60 mg/ml.
Depending on the particular nature of the hydrogel system employed, the resultant fibrinogen rich compositions may be substantially free of one or more low molecular weight components naturally present in the intial blood composition, such as albumin, growth factors, and the like. For example, where hydrogels with a molecular weight cutoff, as described above, of 100.000 daltons are employed, the resultant fibrinogen rich compositions will be substantially free of components having a weight that does not exceed 100.000 daltons. Of particular interest in certain embodiments is the preparation of fibrinogen rich compositions with reduced albumin concentration, i.e. compositions that have reduced albumin concentrations over that which would occur during the concentration process.
The subject fibrinogen rich composition may be used as produced above or modified through the addition of one or more different agents that modulate the chemical and/or physical nature of the composition. Additional agents which may added to the composition include: proteins associated with coagulation, e.g. Factor II. fibronectin and the like; viscosity modifiers, such as collagen, sodium hyaluronate. and the like; antioxidants. such as hydroquinone. vitamin E. and the like; buffering agents, such as HEPES and the like; processing aids, antifibrinolytic agents, platelet activating agents, wound healing agents, and the like. Of particular interest in certain embodiments of the invention, particularly where the composition is to be used in a firbin sealant tissue adhesive, is the modification of the fibrinogen rich composition to comprise a visualization agent. Visualization agents (i.e. agents that may help a surgeon see those tissues to which the fibrin glue has been applied) that find use include blood compatible, non-toxic fiourescent compounds and chromogenic dyes, where specific visualization agents of interest are those that provide for color contrast with the background tissue, with blue and green being preferred colors, where specific agents include: indocyanine green, FD& C no. 1. FD & C no. 6. eosin. fluorescein. and the like.
Following preparation of the fibrinogen rich composition, the composition may be used immediately or stored for use at a subsequent time. Any suitable storage means may be employed, where the storage means will typically be sterile where the composition is to ultimately be used in a physiological setting, e.g. where it is to be used in a drug delivery vehicle, as a surgical adhesive and the like, as further described below. One convenient means of storing the composition is to lyophilize the composition and package the lyophilized product in a sterile packaging for subsequent use. such as a syringe. Alternatively, the composition may be stored at a reduced temperature, e.g. from about 4 to -20 °C or lower. The subject method of preparing the fibrinogen rich composition may be performed at both the laboratory scale and scaled up to produce large amounts of fibrinogen rich composition, e.g. from pooled plasma. On the laboratory scale, the process of the invention may be conducted using standard laboratory and medical equipment. For example, whole blood may be withdrawn from a mammalian host into a syringe containing an anticoagulant and emptied into a sterile centrifuge tube containing the absorbable hydrogel. e.g. hydrogel beads. The hydrogel is then allowed to swell in the presence of whole blood for a period of time sufficient for the desired amount of protein concentration to occur, as described above. The resultant concentrated plasma, i.e. fibrinogen rich phase, is then separated from swollen hydrogel and other cellular material, e.g. red blood cells, by any convenient means, e.g. centrifugation and aspiration using a blunt needle. Such laboratory scale processes find particular use in situations where one wishes to prepare an autologous tissue adhesive, e.g. where the adhesive is prepared from a patient's own blood prior to, or even during, a surgical operation. For scale-up preparation from large volumes of initial blood composition, pooled blood plasma, which may be screened for viruses such as Hepatitis B. and AIDS, may be transferred to a batch reactor containing sterile absorbable hydrogel beads, e.g. photo polymerized polyethylene glycol diacrylate (molecular weight range 20.000 daltons). and the like. The beads may then be allowed to absorb water present in the pooled plasma until a desired level of fibrinogen concentration is reached. After reaching the fibrinogen level (typically > 30 mg/ml), the beads are separated from the concentrated solution, i.e. fibrinogen rich composition, either by filtration, centrifugation. or through their settling out due to gravity.
The subject fibrinogen rich compositions find use in a variety of applications, as components of tissue adhesives. in drug delivery vehicles, hemostatic agents, wound healing agents, and the like. In tissue adhesives, the subject compositions will be used in combination with a coagulation component, usually a solution of thrombin and calcium ions, where upon combination of the composition with the coagulation component a fibrin sealant composition is produced which sets into a biocompatible proteinaceous matrix, i.e. a fibrin gel or sealant. A variety of different methodologies and devices for preforming such methodologies have been developed for preparing fibrin sealants from fibrinogen rich compositions, and such methodologies and techniques are suitable to prepare a fibrin sealant from the subject compositions. As with the preparation of fibrin sealants from fibrinogen compositions prepared from prior art methods, fibrin sealants may be prepared from the subject compositions by applying them to a tissue repair site (i.e. a tissue site to which adherence of a second tissue is desired) either simultaneously or sequentially with a thro bin calcium ion setting composition, where suitable thrombin/calcium compositions are readily available from commercial sources and known to those of skill in the art. To apply the fibrin sealant, the two components described above may simply be applied sequentially or simultaneously to the tissue repair site via a needle or syringe or other application system. In certain embodiments, it is preferred to apply the components sequentially so as to "prime" the tissue, which results in improved tissue adhesive results. Where the tissue is primed, a first component of the fibrin glue. e.g. the crosslinker or thrombin. is applied to the tissue repair site. Next, the fibrinogen components, which may include additional crosslinker/thrombin. is applied. In stead of manually applying the fibrin glue to the tissue repair site, one may use specialized devices for applying the two components of the fibrin glue. Representative devices which may be used include those described in US Pat. No. 4.874.368; US Pat. No.4.631.055; US Pat. No.4,735,616; US Pat. No.4.359.049; US Pat. No.4,978.336; US Pat. No.5,1 16,315; US Pat. No.4.902.281 ; US Pat. No.4.932.942; WO 91/09641. and Tange, R.A.. Fibrin Sealant in Operative Medicine: Otolaryngology-Vol. 1 (1986). the disclosures of which are herein incoφorated by reference. Fibrin sealants as prepared above with subject compositions find use as tissue adhesives in a variety of clinical applications, where such applications are reviewed in Schlag & Redl. Fibrin Sealant in Operative Surgery (1986) Vol. 1-7. and include: cardiovascular surgery, orthopaedic surgery, neurosurgery, ophthalmic surgery, general surgery and traumatology, plastic reconstruction and maxillofacial surgery, otorhinolaryngology. and the like. Where convenient, the fibrin sealant may comprise a visualization agent (e.g. where the sealant is used in a laproscopic method). The visualization agent, including those described above, may be present in one or both of. but usually one of. the components of the fibrin sealant, i.e. it may be present in the fibrinogen rich component (where it may have been introduced to the fibrinogen rich composition after its production or introduced into the initial fluid from which the fibrinogen rich compositions is produced (with the amount used selected in view of the absoφtion of agent by the hydrogel)) and/or the thrombin calcium ion component. The subject fibrinogen rich compositions according to the subject invention may also be used in a fibrin gel or sealant or matrix for biologically active agent or drug delivery. Active agents of interest which may be delivered with fibrin sealant compositions prepared as described above include: proteins, carbohydrates, nucleic acids, and inorganic and organic biologically active molecules, where specific biologically active agents include but are not limited to: enzymes, antibiotics, antineoplastic agents, local anesthetics, hormones, antiangiogenic agents, antibodies. neurotransmitters. psychoactive drugs, drugs affecting reproductive organs, and oligoncucleotides such as antisense oligonucleotides. To prepare fibrin sealant drug delivery compositions with the subject fibrinogen rich compositions, one may simply combine a therapeutically effective amount of the active agent with one or both of the components of the fibrin sealant and prepare the sealant as described above. In a preferred embodiment of the subject invention, the active agent or agents are present in a separate phase from the fibrin gei which protects the fibrin gel while it is setting from adverse effects of the active agent and/or modulates the release kinetics of the active agent from the gel. where " separate phase ' could be oil (oil-in-water emulsion), biodegradable vehicle, and the like Biodegradable vehicles in which the active agent may be present include encapsulation vehicles, such as microparticles, microspheres, microbeads, micropeliets and the like, where the ac e agent is encapsulated in a bioerodible or biodegradable polymer such as polyanhydπde, polyglycolic acid, polylactic acid, polyorthocarbonate. polycaprolactone, polytπmethylene carbonate or their copolymers. caging or entrapping molecules, such as cyclodextπns and the like, etc Biodcgradeable vehicle protected active agents are particularly preferred where the active agent is an antibiotic, such as gentamycin. tetracyicine. and the like
In using fibrin sealants prepared from the subject fibrinogen rich compositions as drug delivery vehicles, the fibrin sealant comprising the active agent, where the agent may be originally present in the fibrinogen and/or thrombin component of the sealant gel. which is optionally and preferably present in a biodegradable vehicle, will be administered to a host prior to setting of the fibrin sealant or gel. where upon administration the gel will set and act as a depot for release of the active agent to the host Such methods of drug delivery find use in both systemic and local administration of an active agent. In using the fibrin sealants tor drug delivery as described above, the amount of fibrin sealant and dosage of agent introduced into the host will necessarily depend upon the particular drug and condition being treated Administration may be by any convenient means, such as svnnge. cannula. trochar. and the like
While various illustrative uses of the fibrinogen rich compositions prepared b> the subject methods have been reviewed above, as explained supra the subject methods are not limited to the preparation of fibrinogen rich compositions, but can be used to produce other protein concentrates I or example, by selecting the appropriate hydrogel with the appropriate absoφtive properties, one can prepare albumin rich compositions
Albumin rich compositions can be used in the preparation of albumin tissue adhesives or glues analogous to fibrin glues I or albumin glues, the albumin present in the albumin rich phase can be crosslinked using a number of different cross-linking means, including the use of chemical crosslinking agents, such as polyaldehydes. thermal crosslinking agents, such indocyanine green in combination with light (e g laser emitting at 780 nm). and the like. See U.S. Patent No. 5.583,1 14, the disclosure of which is herein incoφorated by reference.
In certain embodiments of the invention, a water soluble, biodegradeable synthetic cross-linkers will find use in the preparation of protein gels, such as fibrin glues and albumin glues mentioned above, or other compositions in which polymeric compounds are cross-linked, as described herein and in other applications known to those of skill in the art. The water soluble, biodegradeable synthetic cross-linkers will be multi-functional, where by multifunctional is meant that the cross-linkers will be other than monofunctional, where illustrative multifunctional cross-linkers are difunctional, trifunctional, tetrafunctional. pentafunctional. hexafunctional.... up to "'n"'-functional. where n is an integer representing the number of different end cap functional regions present on the polymeric crosslinkers.
The water soluble, biodegradable polymeric crosslinkers will have a core that is a biologically inert polymeric unit; (b) an extension at each end of the core that is a biodegradable polymeric unit: and (c) an end cap on each extension that is a reactive moiety.
The biologically inert polymeric unit or region that makes up the core of the compound will be an inert polymeric block that is biocompatible. where the block may or may not be biodegradable. Preferably, the core is a polymeric region that is water soluble, where preferred polymers from which the core may be derived include: polvethers. e.g. polyakyleneoxides such as poly(ethylene glycol). poly( ethylene oxide). poly(ethylene oxide )-co-(poly(propyleneoxide)block copolymers: poly(vinyl alcohol); poly(vinyl pyrrolidone). poly(amino acids), poly(ethyloxzoline). dextran and the like, where polyethers. and more particularly polyoxyalkylenes. are preferred. The biodegradable extension component of the subject crosslinkers will be a component that degrades under physiological conditions into non-toxic products, where the biodegradable extension component will generally be hydrolyzable. Hydrolyzable components of interest include: polymers, copolymers and oligomers of: glycolide. dl-lacide. d-lactide. 1-lactide. caprolactone. dioxanone and trimethylene carbonate or their copolymers and the like. In many instances it is desirable to have polylinkers with enzymatically hydrolyzable biodegradable extension components, e.g. regions cleavable by metalloproteinases and collagenases. where such components will generally be peptidic Illustrative biodegradable regions include polyhydroxyacids. polyorthocarbonates, polyanhydπdes. polylactones. polyaminoacids and polyphosphates and the like The size or length of biodegradable region may be varied to in a number of ways For example, by using a crosslinker based on glycohde in the crosslinking of proteins, the resultant crosslinked protein degrades much faster than crosslinked proteins based on polycaprolactone cross- linkers Thus, by choosing appropriate biodegradable polymer regions in the crosslinker. a suitable degradation profile of the resultant crosslinked protein can be obtained With difunctional cross-linkers, peptidic biodegradable regions are preferred The reactive moiety that is the end-cap on each of the extensions is an activated functional group which provides for covalent bonding to proteins under in vivo conditions without free radical initiation Such groups include carbodiimidazole. sulfonyl chloride, chlorocarbonates. n-hydroxysuccιnιmd\ l ester, succinimidyl ester, epoxides. aryl hahdes. sulfasuccinimidyl esters, maleimides. and the like FIG 5 provides further description of such polymeric cross-linkers The biodegradable region of the crosslinker is represented by («* «■*). the activated reactive end-group is represented by (■» ), and the inert, biocompatible block is represented by (- ) Structure A shows a linear water soluble biodegradable polymer end-capped with two reactive functionalities like carbodiimidazole (CDI) The linear water soluble region is a polyalkylene oxide, preferably polyethv lene gl> col which is extended with the biodegradable region which is a copolymer or homopolymer of tnmethylene carbonate This polymer is then terminated with CDI Structure B is a branched or star shaped tπfunctional biodegradable polymer which has inert polymer at the center The inert polymer is extended with oligomeric biodegradable extensions which are then terminated by reactive functional end group Structures C and D show a multibranched tetrafunctional biodegradable polymer This polymer again has a water soluble core at the center which is extended by small oligomeric extensions of biodegradable polymer and then terminated with reactive functional groups like carbodiimidazole groups Structure E shows a multifunctional star or graft type biodegradable polymer This polymer has a water soluble polymer, like polyalkylene oxide, at the core which is then extended with biodegradable polymer The biodegradable polymer is terminated with reactive end groups The structures shown in Figure 5 have three common structural features. A water soluble core which is made by a polymer like polyethylene glycol. The core is extended with an oligomeric extension of biodegradable polymer such as polylactic acid. The degree of polymerization of biodegradable core is kept small (preferable less than 10) to maintain water solubility. The molecular weight of PEG is kept less than 35, 000 daltons. as higher molecular weights may be difficult to eliminate by the body. The biodegradable polymer is then end-capped with reactive functional groups which are capable of reacting with proteins or its functional groups like amine. thiols and carboxylic acids preferably in water and preferably at pH 4 to 9. more preferably at pH 7.0 to 7.4.
The polymeric crosslinkers may be prepared using variety of different synthetic methods. In a prefeired embodiment, the polymer described in structure A can be obtained by a ring opening polymerization of trimethylene carbonate initiated by a dihydroxy compound such as polyethylene glycol molecular weight 2000 d in presence of a suitable catalyst such as stannous octoate. The hydroxy groups of the copolymer thus obtained are then activated with carbodiimidazole (CDI). The CDI activated polymer can then be reacted with a protein concentrate as prepared by this invention to form a crosslinked gel. The reaction conditions of the crosslinking reaction will depend on the nature of activating group employed. Preferred reactions are at pH 5 to 8. most preferred at pH 7.4. The resultant gel degrades due to hydrolysis of the biodegradable polymer such as polytrimethylene carbonate polymer inside the crosslinker. The crosslinking density of the resultant network can be controlled by the overall molecular weight of the structure. A lower molecular weight such as 600 will give much higher crosslinking density as compared to a higher molecular weight crosslinker such a with molecular weight 10000 daltons. The high molecular weight linear crosslinker is preferred in obtaining elastic gels. The reaction between proteins and crosslinkers can be carried out directly on tissues and used as tissue glue. A trifunctional biodegradable crosslinker can be obtained by initiating a polymerization of lactide with trihydroxy polyethylene glycol (ethoxylated trimethylol propane triol) in presence of stannous octoate . The degree of polymerization of lactide is kept less than 5. This is achieved by choosing a molar ratio of PEG with lactide (molar ratio of lactide to PEG is 6, 2 per branch). The PEG-lactate trifunctional polymer is isolated. The hydroxy end groups of copolymers are then activated with CDI. Since this a trifunctional crosslinker, it will give higher crosslinking density as compared to similar molecular weight difuctional crosslinker. This gives additional flexibility in controlling crosslinking density of a crosslinked structure and hence their mechanical and biodegradation properties. The tertafuctional structures are obtained by reacting the polyalkylene oxide copolymer such as Tetronic 908 (obtained from BASF coφoration) with caprolactone in presence of stannous octoate. The reaction is carried out in melt at 180 °C for 6 hours under nitrogen atmosphere. The molar ratio of caprolactone to Tetronic 908 is kept to 12. which maintains water solubility of Tetronic 908-caprolactone copolymer in water. The polymer is activated with CDI and used in crosslinking reaction with proteins.
The protein concentrates prepared by the subject methods also find use in the preparation of novel hydrogel wound dressings. To prepare hydrogel wound dressings from the subject protein concentrates, such as the fibrinogen rich compositions discussed above, where the protein concentrate serves as the "aqueous composition component." the protein concentrates are combined with a macromonomer component and a cross-linking agent under conditions sufficient to produce a hydrogel comprising the protein concentrate composition interspersed throughout the cross-linked gel network. Macromonomers that find use in the preparation of hydrogel wound dressings are non-toxic, water soluble, non-ionic macromonomers that comprise a polymerizable group, preferably an addition polymerizable group, where macromonomers which can be crosslinked via free radical polymerization are preferred. For example, water soluble polymers having unsaturated polymerizable groups such as acrylate. methacrylate, itaconate, and the like find use. Water soluble polymers of interest which can be modified with unsaturated polymerizable groups include: polvethers. such as polyalkyleneoxide polymers and copolymers. polyethyleneoxide. polyethylene glycol. poiyethyleneoxide-polypropyleneoxide block, random or graft copolymers; polyvinyl alcohol; polyvinyl pyrrolidinone: and the like, where polyether polymers or derivatives thereof are particularly preferred as macromonomers. If desired, two or more macromonomers can be copolymerized to obtain suitable hydrogel properties. While preparation of the hydrogel wound dressings of the subject invention has been described in terms of using the protein concentrates of the present invention as the aqueous composition component that is combined with the macromonomer and the cross-linking agent, other aqueous composition components may also be employed to prepare hydrogel wound dressings according to the subject invention. Other aqueous compositions that may be employed as the aqueous composition component to prepare the subject hydrogel wound dressings include: whole blood, serum, platelet rich plasma (where the platelets may or may not be activated), tissue culture medium, and the like. The hydrogel wound dressings of the subject invention can be prepared just prior to use or prepared and then stored for subsequent use.
The cross-linking agent is generally a non-toxic polymerization initiator, such as a free radical initiator. Several free radical initiating systems can be used to polymerize the macromonomers containing polymerizable groups such as acrylate or methacrylate. Some of the preferred examples are: Darocur 2959 (initiated around 360 nm). Irgacure 651 (initiated around 360 nm). eosin-triethanol amine (initiated around 510 nm) . methylene blue-triethanol amine (initiated around 632 nm). sodium persulfate (initiated around 50 °C). ammonium persulfate (initiated at 50 °C). Glucose oxidase-glucose-ferrous sulfate (initiated around 37 °C in presence of dissolved oxygen in the formulation) and the like.
Optionally, other pharmaceutically acceptable catalysts and cocatylysts can be added to the macromonomer solution to accelerate the polymerization speed and/or to improve its shelf life. For example, small amounts of vinyl pyrrolidinone (concentration around 1 -10 micrometers per ml) can be added while using eosin- triethanol photoinitiating system. Inhibitors such as hydroquinone may be added to prevent premature polymerization of macromonomer during its storage. Optionally, the wound dressings described may be prepared with bioactive compounds such as antibiotics to reduce the bacterial infection.
Other agents that may be included in the composition to modulate the ultimate properties of the hydrogel wound dressing prepared therefrom include proteinaceous polymers, such as collagen, other polymeric compounds, such as hyaluronic acid (including derivatives thereof), dextran and the like. Of particular interest are hydrogel wound dressing composites which include, in addition to the hydrogel, at least one of collagen and hyaluronic acid, where in such composites. PEG is the preferred hydrogel.
Hydrogel wound dressings produced in accordance with the present invention can be prepared with desired physical and chemical properties by choosing specific structural features, such as nature of polymerizable group of the macromonomer. the number of polymerizable groups present per macromonomer chain, the chain length and chemical structure of the macromonomer. where the modulation of such parameters to obtain a hydrogel wound dressing of desired characteristics is within the skill of those in the art.
The hydrogel wound dressing composite produced according to the present invention can be produced in various shapes and sizes, such as: films, ropes, rods, plugs, thin or thick sheets, moldings and laminates. Hydrogel wound dressings prepared according to the present invention can be modified further, if necessary or desired, by the addition of pharmaceutically acceptable antioxidants. plasticizers. coloring agents, fillers, fibers, fiber meshes, adhesive backing sheets and the like.
Optionally, the hydrogel wound dressing composite produced according to the present invention can be reinforced with flexible or rigid fibers, fiber mesh, fiber cloth and the like to produce a fiber reinforced hydrogel wound dressing. The insertion of fibers or fibrous structures improves flexibility and tear resistance of the hydrogel wound dressings. Such structures can be produced using any convenient protocol. In a preferred method, the aqueous macromonomer formulation is added to the fiber cloth or net such as cotton gauze. The liquid then flows into the interstices of cloth and is then polymerized to produce a hydrogel. where care is taken to ensure that the fibers or fiber mesh are buried completely inside the hydrogel material. The fibers used in reinforcing the hydrogel network are preferably hydrophilic in nature to ensure better compatibility with the hydrogel network. Also a transparent, flexible plastic film such a polyethylene plastic film which is permeable to oxygen, may be applied on top of the hydrogel wound dressings (opposite side of wound contact) and which may supplied with a plastic mold described in this invention. This plastic film prevents the moisture loss from the hydrogel. The hydrogel wound dressings produced according to the present invention can be used on variety of wounds The wounds may be surgical wounds, first, second or third degree burns, skin lesions, decubitus ulcers, venous ulcers, bed sore and the like Also provided by the subject invention are kits for preparing the subject fibrinogen rich compositions, kits for preparing the wound dressings and kits for preparing fibrin sealants from the subject fibrinogen rich compositions Kits for preparing the fibrinogen rich compositions will comprise at least a a hydrogel composition and instructions for preparing the fibrinogen rich composition according to the methods of the subject invention, where the instructions may be present in the kit as an insert, incoφorated into the containers and/or packaging of the kit, and the like Kits for preparing the fibrinogen rich compositions may further comprise anticoagulants, e g heparin. and one or more containers e g syringes, vials and the like, for use in preparing the fibrinogen rich composition, and the like, where the kit components will generallv be sterile, particularly where the ultimate use of the composition involves introduction into a host, e g a patient undergoing surgery Kits for preparing a fibrin sealant according to the subject invention will comprise at least a fibrinogen rich composition prepared according to the subject invention and a thrombin calcium setting component present in separate containers The kit mav further comprise a device for delivery the fibrin sealant to a tissue repair site as described above Optionally, the kits may also comprise an visualization agent and/or an active agent, preferably present in a biodegradable vehicle As with the kits for preparing the subject fibrinogen rich compositions, kits for preparing fibrin sealants will also typically comprise instructions for preparing the fibrin sealant, which instructions mav be present as a package insert and/or associated with the containers present in the kit and/or packaging of the kit
Kits for preparing the subject wound dressings will comprise at least the macromonomer component described above and instructions for carrying out the subject methods of preparing the wound dressings Generally, the kits will also comprise a crosslinking agent The kits may further comprises a fluid of interest which is to be entrapped in the hydrogel. such as the concentrated protein composition or other fluids as described above Other components that may present in the kits include a container/mold for preparing the wound dressings, and the like Also provided are devices for automatically performing one or more of the steps of the subject method to prepare a protein concentrate from an initial protein comprising aqueous composition. The devices of subject invention will generally comprise a container means having a quantity of hydrogel and of sufficient volume for the hydrogel to come into contact with the initial protein composition. The container will further comprise at least one opening for introducing the initial composition into the container and removing the resultant protein concentrate from the container. Generally, the devices will comprise a contacting means for ensuring fluid transfer between the fluid introduced into the container and the hydrogel present therein. In some embodiments, the contacting means is a simple as a polymeric interface layer that is resistant to protein absoφtion adsoφtion. In other embodiments, the contacting means may comprise a filtering means, such as a glass filter or membrane, which is impermeable to the hydrogel. Representative devices according to the subject invention are further described in terms of the figures. In Figure 6 is represented a first device embodiment according to the subject invention. Device 10 is a plastic/glass bottle 12 (could also be a bag) containing hydrogel beads 14 separated from the remainder of the bottle interior by filter mesh 18 which is permeable to liquids but not to the hydrogel beads (i.e. the contacting means). The bottle has a narrow neck 20 that has a volume measurement printed thereon 22. In using this device embodiment, plasma is added to the hydrogel beads and the swelling process is started. The bottle is inverted every 10 minutes to separate the concentrated (the filter prevents the hydrogel from entering the narrow neck region. In the narro neck region (upon inversion), the volume of plasma is measured. When the desired volume is reached, the concentrated plasma is removed using a suitable technique. Fig. 7 depicts a second embodiment of a device according to the subject invention. Cylindrical device 30 has entry port 32 for the introduction of fluids into the device. The entry port has a seal (e.g. silicon such as those employed on blood banking blood bags) through which fluids can be sterily introduced into the device or a removable lid (not shown) which can be used in an asceptic technique. Areas 34 contain hydrogel which can absorb fluid and smaller molecules. For use in preparing cryoprecipitate. the hydrogel might have a molecular weight cutoff of about 80.000 so albumin would be absorbed into the hydrogel. In the embodiment shown, the hydrogel compartment would not change physical volume, but has room for the hydrogel to swell and expand. However, other embodiments in which the physical volume of the hydrogel compartment is expandable are envisioned. Collection compartment 36 is a clyindrical shaped cup (though other shaped compartments are also contemplated. such as rectangular etc.) that has walls made of material that does not absorb protein and is also impermeable to fluid. Exit port 38 allows for the sterile removal of fluid, i.e. a septum that can be punctured with a needle.39 is the top layer of the hydrogel compartment which is a material resistant to protein adsoφtion/absoφtion (i.e. the contacting means). The top layer could be fabricated from an appropriate polymeric material, e.g. PEG. or be a membrane which allows for passage of water and other small molecules, such as those used in the purification industry. Use of a membrane provides greater flexibility in the nature of the hydrogel that may be employed, as direct contact of the protein components of the fluid and the hydrogel can be prevented by the membrane. Accordingly, the hydrogel in such devices need not necessarily be a hydrogel that is non-denaturing, as that term is employed above. Fig. 7(e) provides a top view of the device. Figs. 7 (b) to 7 (d) show the device in use. In using the device to prepare a protein concentrate from an aqueous composition, fluid 40 is introduced through port 32. Water and other small molecules are absorbed into hydrogel 34. As the fluid is absorbed, the level of the fluid falls until it reaches the top of the collection compartment 36. Concentrate is then removed from the collection compartment through exit 38. The device is configured with dimensions chosen based on the intended nature of use. For example, in the preparation of a fibrinogen rich composition from plasma, in many situations the device will have dimensions sufficient to hold up to 1 unit of blood. Where desired, the entire device can be rotated at low φm (usually less than 500) to facilitate mixing of the fluid and reduction of fouling by proteins of the top or interface layer of the hydrogel or the membrane 39 covering the hydrogel component. Furthermore, coatings or active coatings or gel microspheres with active coatings could be placed on the periphery of the device (on the inside of the outside wall of the device above the hydrogel compartment) which would specifically absorb albumin, other proteins or specific cell types for cell purification. If the membrane 39 over the fluid absorbing component 34 is properly selected, a wider range of swelling polymers may be employed as the hydrogel component as mentioned above. Accordingly, polymer compositions such as those found in disposable diapers to absorb urine may be employed. Alternatively, the device could operate with hydrogel beads that are excluded from the collection cup 36 by having a means over the collection cup.
The following examples are offered by way of illustration and not by way of limitation.
EXPERIMENTAL A. Materials and Methods
Polyethylene glycols ( Merck, mol. wt 20000 and BDH mol. wt 6000) were used as received. Pluronic® and Tetronic® polyols were purchased from BASF coφoration. Photoinitiator Darocure -W 2959 was purchased from Ciba Geigy. The biodegradable polymers like polylactic acid, polyglycolic acid are purchased from Polysciences. All other reagents, solvents are of reagent grade and are purchased from commercial sources such as Fluka. Aldrich and Sigma. Small laboratory equipment was purchased from Fisher or Cole-Parmer.
B. Synthesis of Hydrogels 1. Synthesis of Polyethylene Glycol Diacrylate
In a 1.000 ml 3 neck reaction flask, attached with condenser, nitrogen inlet and thermometer. 100 grams of PEG (molecular weight 20.000 daltons) and 600 ml dry toluene are added. After distilling 100 ml of toluene from the flask, the PEG solution is cooled to 50-60 °C. To this mixture. 2.1 ml of triethylamine and 1.2 ml of acryloyl chloride is added under nitrogen atmosphere. The reaction is stirred for 10-12 hours at ambient temperature under nitrogen atmosphere. The triethylamine hydrochloride (a reaction byproduct) is removed by filtration and the product is recovered by pouring the filtrate in to large excess hexane. It is further purified by several dissolution- precipitation steps using tetrahydrofuran(THF)-hexane as solvent - nonsolvent respectively. The product is further dried at 40 °C under vacuum until constant weight is observed. Yield 60 gram. Other macromonomer containing PEG with different molecular weights, and different number of unsaturated groups are easily synthesized using the procedure given above. For example. 30 grams PEG molecular weight 6000 daltons is reacted with 2.67g acryloyl chloride and 3.03 g triethyl amine using a similar procedure mentioned above to give PEG 6.000 diacrylate. Similarly PEG 10000 diacrylate is prepared by reacting 30 grams of PEG molecular weight 10.000 Da with 1.82 g of triethyl amine and 1.61 g of acryloyl chloride. Polyethyleneoxide (PEO) molecular weight 35.000 Da (30 g) is reacted with 0.52 g of triethyl amine and 0.46 g of acryloyl chloride to get a corresponding diacrylate derivative. Some polyethylene glycol based macromonomers may also be purchased using commercial sources such as Sartomer. Polysciences.
2. Synthesis of Polyethyleneoxide-Polypropyieneoxide-Polyethyleneoxide Diacrylate 50 grams of Pluronic® F 127 (a PEO-PPO-PEO block copolymer with 70%
PEO content, molecular weight 12500 daltons. purchased from BASF coφoration) is dried under vacuum at 80-100 °C. The polymeric diol is then transferred to a 3 neck reaction flask equipped with nitrogen inlet and thermometer. 500 ml toluene. 3.3 ml of triethylamine amine and 1.9 ml of acryloyl chloride are added to the reaction mixture under dry nitrogen atmosphere. The reaction mixture is stirred overnight and filtered to remove triethylamine hydrochloride. The filtrate is added to 3000 ml hexane to precipitate the diacrylate derivative of Pluronic F127. The macromonomer is purified by several dissolution-precipitation steps from THF-hexane solvent-nonsolvent system. Finally the diacrylate is dried under vacuum at 40 °C to a constant weight. Other derivatives of Pluronics with different arrangement of PEO-PPO blocks and with different HLB values can also be acrylated in a similar manner.
3. Preparation of sterile gels using photopolymerization method
10 grams of PEG diacrylate. synthesized by a procedure described above, are dissolved in 20 grams of phosphate buffer solution ( pH 7.4. 0.2 g/L KCl. 0.2 g/L of H,PO4. 8.0 g/L of NaCl and 1.15 g/L Na2HPO4). To this solution 600 μl of photoinitiator solution (300 mg of DarocurS- 2959, Ciba Geigy. dissolved in 700 mg ethanol) is added. The following procedure is carried out in a sterile hood. The aqueous macromonomer solution is sterile filtered using 50 ml syringe and 0.2 mm syringe filter. 200 μl of sterile solution is transferred into transparent plastic mold (a single well of a sterile 96 well tissue culture plate). The solution is then exposed to long wavelength UV light (Blak Ray light source, model 3-100A. Flood 365 nm. intensity 10 mW/cm:) for 120 seconds. The polymerized gel is removed from the mold. The resultant gel may be freeze dried or freeze dried coupled with limited hydration following preparation. Several of such hydrogel beads are synthesized and stored in a sterile container for further use.
4. Preparation of Sterile Gel Microspheres
5 grams of PEG diacrylate. molecular weight 10.000 daltons are dissolved in 15 grams of trietholamine/triethanolamine hydrochloride solution (90 mM. pH =7.4) in a 50 ml amber colored glass bottle. To this solution 30 μl of Eosin Y solution (1 mg/ml in PBS) and 30 μl vinyl pyrrolidinone are added. The mixture is protected from visible light using aluminum foil. The following operations are carried out in a sterile hood: The macromonomer solution is filtered using 50 ml syringe and 0.2 μm syringe filter. The filtered sterile solution is filled into 50 ml plastic syringe (wrapped in aluminum foil for protection against light) with 22 gauge needle. A 100 ml of sterile mineral oil which is constantly being stirred using a magnetic stir bar is exposed to green light source (American Argon ion laser. Model 905 emitting at 532 nm. 100 mW/cπr). The macromonomer solution is flushed out of needle and the droplets are collected into sterile mineral oil which is under green light irradiation. After 5 minutes of irradiation of monomer solution to green light, the photopolymerized hydrogel beads are separated from mineral oil by filtration. The hydrogel beads are further washed with sterile hexane to remove traces of mineral oil. The hydrogels beads are then stored in sterile saline solution for 5 hours to remove initiator fragments, unreacted or uncrosslinked macromonomer etc. The washed beads are then isolated by filtration and lyophilized under sterile conditions.
5. Synthesis of multifunctional thermosensitive macromonomer 30 g of Tetronic 908 polyol is dissolved in 400 ml dry benzene. 100 ml of benzene is distilled to remove traces of water from the polyol. The solution is cooled to 30 "C and 1.45 g triethylamine and 1 .90 g acryloyl chloride are added. The reaction mixture is refluxed for 1 h under argon atmosphere. It is then cooled and then filtered to removed triethylamine hvdrochloride. The filtrate is then added to 2000 mi hexane to precipitate the polymer. The polymer is purified by several precipitations from THF- hexane solvent-nonsolvent system. Further solvent removal/drying is achieved by vacuum drying overnight at 60 °C.
6. Preparation of Hydrogels by Radical Thermal Polymerization Method
10 grams of PEG diacrylate molecular weight 20.000 daltons are dissolved in 20 gram of PBS buffer ( pH = 7.4). To this solution 300 mg ammonium persulfate are added as a thermal free radical polymerization catalyst and the mixture is sterile filtered. This sterile solution is then transfeired into several sterile 5- 10 mm diameter glass test tubes. The test tubes are then capped and transferred into lab oven maintained at 70 °C. After 15 minutes, the test tubes are removed, cooled and the polymerized gels are removed by breaking the glass container. The gel rods are cut into small 1 -2 cm pieces and stored. Sterility of gel rods is maintained throughout the entire operation. The resultant hydrogels. when contacted with water, absorb water over time and increase in mass, as shown in Fig. 3.
C. Synthesis of Drug Compositions
1 . Preparation of Rifampin Sulfate Loaded Polylactic Acid Particles
In a 50 ml glass beaker. 2 g of polylactic acid is dissolved in 20 ml methylene chloride. To this mixture is added 1 g of gentamycin sulfate. The resultant slurry is thoroughly mixed and poured on a glass plate. After initial methylene chloride removal from the slurry, the partially dried film is dried in vacuum oven at 40 °C for 48 hours. The resultant gentamycin-PLA composite is pulverized into small particles by cryogenic grinding at liquid nitrogen temperature. The particles may be sieved to obtain a particular size distribution. 2 Preparation of Gentamycin Loaded Microspheres of Polylactate- o-polyglycolate (50 50) Polymer
0.1 g of Rifampin is dissolved in 5 ml PBS (pH 7 2) containing 10 mg of bovine serum albumin The resultant aqueous solution is added to 30 ml methylene chloride containing 5 grams of polymer and then emulsified by a brief sonification for 30 seconds The water-in-oil (W/O) emulsion is reemulsified in 2,000 ml 0 1% (w/v) polyvinyl alcohol (PVA) with stirring for 3 hours at room temperature The hardened microspheres are washed three times with PBS buffer to remove unencapsulated rifampin The microspheres are recovered by centrifugation and freeze-drying The residual solvent is removed by drying the microspheres in vacuum oven at 37 °C for 48 hours Alternatively 0 1 g of Rifampin is dissolved in 5 ml acetone The acetone solution is then added to 30 ml solution of polylactate-co-polylactate(50 50) in methylene chloride (4% vvt/v) I he mixture is then spray dried using standard laboratory spray dried
D Preparation of Fibrinogen Composition Using Hydrogels 1
In a sterile glass bottle, having a rubber septum cap and containing heparin anticoagulant. 20 ml of patient s blood is transferred using a sterile needle transfer technique or standard Schlenk line techniques I he tube is then centπfuged at 3.200 rpm for 10 minutes The resultant blood plasma is transferred using a sterile needle transfer technique to another sterile bottle containing 4 g of sterile PEG 20.000 diacrylate gel rods prepared by the method described in example 6 above The hydrogel selectively absorbs water and other low molecular weight proteins such as albumin, plasminogen and compounds like heparin leaving behind a concentrated solution of fibrinogen and Factor XIII The hydrogel absoφtion time (usually minutes to several hours) is controlled so as to obtain a desired volume/concentration of a final solutιon( typically 90-95 % water is removed from the plasma) This concentrated protein solution gels with commercially available thrombin. Ca : ion solution
2 Preparation with Hydrogel Beads Blood plasma is concentrated with hydrogel beads using a method analogous to that described in example DI . above. The process is shown schematically in Fig. 1. In the schematic shown in Fig. 1. dry or partially dry hydrogel beads are placed in a sterile tube as shown in Fig. 1 A. Next, a blood plasma solution is transferred to the tube as shown in Fig. B. The mixture is allowed to remain in contact for a sufficient period of time for the hydrogel beads to selectively absorb water from the plasma, whereby a fibrinogen rich composition is produced, as shown in Fig. IC. A small amount of plasma is taken out periodically and is analyzed for total protein concentration using a biurette method, the results of which are shown in Fig. 4.
E. Preparation of colored fibrin adhesive solution
1. Indocyanine Green
5 ml of concentrated solution of fibrinogen solution as prepared above in Example D is added to a sterile bottle containing 0.1 ml indocyanine green solution in PBS ( cone. 0.1 to 10 mg/ml). The green precursor solution is easy to visualize and apply under normal and laparoscopic surgical environment. This green solution is used with thrombin solution as a colored fibrin adhesive system.
2. Preparation of fluorescent adhesive formulation 5 ml of concentrated solution of fibrinogen solution is added to a sterile bottle containing 0.1 ml sodium fluorescine in solution in PBS (cone. 0.1 to 10 mg/ml). The light green fluorescent precursor solution is easy to visualize and apply under normal and laparoscopic surgical environment. This green solution is used with thrombin solution as a colored fibrin adhesive system.
F. Preparation of Fibrin-biodegradable Microparticle Composite
5 ml concentrated solution of fibrinogen or the commercial fibrinogen containing precursor of fibrin adhesive is mixed with 0.5 grams polylactic acid or polyhydroxy acid microspheres loaded with bioactive compound such as gentamycin. The resultant siurry is transported to a localized disease site inside the human or animal body using minimally invasive surgical device such as laparoscope and crosslinked in situ using thrombin solution. G. Preparation of Protein Composites Crosslinked with Biodegradable Polymeric Crosslinking Agents
1. Synthesis of water soluble difuctional. biodegradable crosslinker based on polyalkylene oxide Synthesis of polyethylene glycol-trimethylene carbonate polyol-
30 g of polyethylene glycol having a molecular weight 2000 is dried at 90- 100 °C in a glass sealing tube. The tube is then cooled and transferred inside an air bag where 12.24 g of trimethylene carbonate and 50 mg of stannous octoate are added to the tube. The glass tube is then sealed under nitrogen atmosphere and heated with stirring at 155 °C and maintained at this temperature for 6 hours. The polyethylene glycol-polytrimethylene carbonate polymer is cooled and recovered by breaking the giass sealing tube. It is further purified by several precipitations from toluene-hexane solvent-nonsolvent system. The product is dried in vacuum at 40 °C and used immediately in the activation reaction.
2. Synthesis of water soluble, tetrafunctional crosslinker based on polyalkylene oxide
Synthesis of Tetronic 908-caprolactone polyol( PCLP)-30 g of Tetronic 908 is charged in a dry 3 neck flask equipped with magnetic stirrer and vacuum inlet. The flask is then heated in a silicone oil bath at 100 °C for 12 hours to dry the Tetronic 908. The flask is cooled to room temperature and 1 .642 g of caprolactone and 0.02 g of stannous 2-ethylhexanoate are added to the flask. The tlask is heated to 180 UC for 6 hours under nitrogen atmosphere. The reaction is product is then dissolved in 200 ml dry toluene (warming of toluene accelerates dissolution). The toluene solution is added to 2,000 ml dry heptane with constant stirring. The product is isolated by filtration. Further purification is accomplished by precipitation of toluene solution of PCLP in heptane. The product is dried in vacuum at 40 UC and used immediately in the activation reaction.
3. a) Activation polyalkyleneoxide lactate copolymer with carbodiimidazole:
30 g of Tetronic 908-caprolactone copolymer is dissolved in 400 ml dry benzene. About 100 ml of benzene is distilled off and the solution is cooled to 50 °C. 2.24 g of carbodiimidazole is added to reaction mixture . The mixture is refluxed for 2 h under nitrogen atmosphere . At the end of 2 hour period, the solution cooled added to 4,000 ml hexane to precipitate the polymer. It is further purified by repeated (3 times) precipitation from toluene-hexane system. The polymer is dried under vacuum at 40 °C.
b) Activation polyalkyleneoxide lactate copolymer with N-hydroxysuccinimidyl ester: 30 g of Tetronic 908-caprolactone copolymer is dissolved in 400 ml dry benzene. About 100 ml of benzene is distilled off and the solution is cooled to 50°C. 2.50 g of succinic anhydride is added to reaction mixture . The mixture is refluxed for 5 hours under nitrogen atmosphere . At the end of 5 hour period, the solution cooled and then added to 4.000 ml hexane to precipitate the polymer. It is further purified by repeated (3 times) precipitation from toluene-hexane system. The polymer is dried under vacuum at 40 °C. c) Activation of acid terminated polymer with dicyclohexylcarbodiimide (DCC)
10 g of Tetronic 908-caprolactone succinate prepared by method described above is dissolved in 100 ml dry methylene chloride. The mixture is cooled to 0 °C in ice bath and 0.5 g of 4-dimthylaminopyridine and 1 g of dicyclohexylcarbodiimide (DCC) are added. The mixture is stirred at 0 °C for 6 hour and filtered. The filtrate is then added to 2.000 ml dry hexane to precipitate the activated succinimydyl ester. The product is isolated by filtration, dried under vacuum and stored under argon at 4 "C.
4. Preparation of Crosslinked Protein Composites
The CDI/succinimydil activated crosslinker polymers as prepared above are dissolved in aqueous buffer solution and reacted with albumin rich solutions to form a gel.
H. Preparation of Hydrogel Wound Dressings
1. Preparation of sterile hydrogel wound dressings using photopolymerization method
- j- 5 grams of PEG 10.000 diacrylate. synthesized by a procedure described in example Bl . is dissolved in 20 grams of phosphate buffer solution ( pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH:PO , 8.0 g/L of NaCl and 1.15 g/L Na,HPO,). To this solution 100 microliter of a photoinitiator solution (300 mg of Darocur® 2959, Ciba Geigy, dissolved in 700 mg ethanol) is added. The following procedure is carried out in a sterile hood:
The aqueous macromonomer solution containing photoinitiator is sterile filtered using 50 ml syringe and 0.2 mm syringe filter. About 10-12 ml of sterile solution is transferred into transparent plastic mold (10 cm x 5 cm x 2 mm). The solution is then exposed to long wavelength UV light (Blak Ray light source, model 3-100A. Flood 365 nm. intensity 30 mW/cm2) for 5 minutes. The polymerized, flexible gel is removed from the mold. This sterile gel can be directly applied over the wound.
2. Gamma radiation crosslinking of macromonomers 3 g of PEG 20.000 diacrylate is dissolved in 12 ml PBS buffer. The aqueous monomer solutions is sterile filtered into glass mold (cavity size 10 cm x 5 cm x 2 mm). The solution is then irradiated at ambient temperature with a 6 Co source with doses up to 200 kGy at a dose rate of 1 kGy/h. After irradiation, the sterile crosslinked gel is removed from the mold and used as wound dressing.
3. Preparation of sterile lyophilized macromonomer formulation
3 grams of PEG 10.000 diacrylate. synthesized by a procedure described in example . is dissolved in 12 grams of phosphate buffer solution ( pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH.PO,, 8.0 g/L of NaCl and 1.15 g/L NaTlPO,). To this solution 60 microliter of photoinitiator solution (300 mg of Darocur® 2959. Ciba Geigy, dissolved in 700 mg ethanol) is added. The aqueous macromonomer solution containing photoinitiator is sterile filtered using 50 ml syringe and 0.2 mm syringe into a sterile amber colored 25 ml glass bottle. The solution is lyophilized while maintaining sterility to remove water. At the end of the lyophilization cycle, the vial is capped with a sterile rubber septum. The lyophilized macromonomer powder containing photoinitiator is used in making 'in situ' formable wound dressings. 4 Autologus or single donor single donor blood or blood components encapsulated wound dressing
15-20 ml of fresh human or animal blood is withdrawn in a standard syringe or similar medical device containing anticoagulant such as heparin or acid citrate buffer (2 % glucose. 0 15M citrate. pH 4 2. 1 ml buffer per 10 ml blood) The blood is then centπfuged in order to separate out the blood plasma The plasma is then removed from the centrifuge tube and transferred into a sterile macromonomer lyophilized powder containing initiator (such as made in example 3 to make a 5 -40 % solution, preferably 10% solution of the macromonomer in plasma (some proteins such as fibrinogen may precipitate depending on the macromonomer and its concentration used) The macromonomer-plasma solution/dispersion is then transferred into sterile plastic dish or mold ot size 10 cm \ 5 cm x 2 mm using a sterile syringe and needle The contents ot the mold are then exposed to long UV irradiation for 3 minutes to crosslink the macromonomer The crosslinked sterile hydrogel can be directly applied on the wound
In another variation of this procedure. 15-20 ml of fresh human or animal blood is withdrawn in a closed sterile plastic or glass vial without anticoagulant The blood is allowed to coagulate for 30 to 60 minutes The top clear solution (serum) is withdrawn from the vial using a syringe This serum is then used in preparation of hydrogel dressing using a procedure mentioned previously
5 Preparation of hydrogel wound dressing containing platelets
15 to 20 ml of fresh human or animal blood is isolated using a standard procedure The blood is then centπfuged at low rpm (around 3.000 rpm) in order to separate the platelet rich plasma The platelet rich plasma is then added to a sterile lyophilized macromonomer initiator formulation such as described in example 3 The entire mixture is then poured into sterile plastic or glass mold of suitable size for example 10 cm x 5 cm \ 2 mm cavity size and exposed to long UV or visible light for 3 minutes to crosslink macromonomer The gel containing encapsulated platelets is removed and may be directly applied on the wound
-33- Platelets may be activated using thrombin or suitable activating agent before or after the encapsulation process. The activated platelets release platelets derived growth factors which are then released in controlled manner by the hydrogel matrix.
6. Preparation of hydrogel wound dressing containing tissue culture medium (without serum)
A sterile amber colored glass vial containing 3 grams macromonomer and photoinitiator ( similar to described in example 6) is dissolved in 12 ml tissue culture medium (for example Dulbecco's modification of Eagle's medium, a medium suitable for culturing human foreskin fibroblasts). The macromonomer solution in tissue culture medium is then sterile filtered into a plastic mold of desired shape for example 10 cm x 5 cm x 2 mm size and exposed to long UV light as mentioned in previous examples. The polymerized hydrogel is then removed from the mold and used as wound dressing.
7. Preparation of hydrogel wound dressing containing tissue culture medium (with autologus or single donor blood serum)
15-20 ml of fresh human or animal blood is withdrawn in a standard syringe and allowed to stand to coagulate for 0.25 to 2 hours. The blood is then centrifuged in order to separate out the blood serum. 2 ml of this blood serum is then mixed with 18 ml of tissue culture medium such as described in previous example. 12 ml of this mixture is then added to a lyophilized macromonomer formulation prepared as described in example 6. This macromonomer is then sterile filtered into a plastic sterile mold 10 cm x 5 cm x 2 mm size and exposed to long UV light for 3 minutes. The crosslinked macromonomer or hydrogel contains tissue culture medium and serum.
8. Preparation of polyalkylene oxide-hyaluronic acid hydrogel wound dressing
3 grams of PEG diacrylate. synthesized by a procedure described in example BI . is dissolved in 12 grams of phosphate buffer solution ( pH 7.2. 0.2 g/L KCl. 0.2 g/L of KH.POj, 8.0 g/L of NaCl and 1.15 g/L Na.HPO, and 0.5% sodium hyaluronate) . To this solution 60 microliter of photoinitiator solution (300 mg of Darocur® 2959. Ciba Geigy, dissolved in 700 mg ethanol) is added. The following procedure is carried out in a sterile hood.
The aqueous macromonomer solution containing photoinitiator and hyaluronic acid is sterile filtered using 50 ml syringe and 0.2 μm syringe filter. About 10-12 mL of sterile solution is transferred into transparent plastic mold (cavity size 10 cm x 5 cm x 2 mm). The solution is then exposed to long wavelength UV light (Blak Ray light source, model 3-100A, Flood 365 nm. intensity 30 mW/cnr) for 5 minutes. The polymerized, flexible gel is removed from the mold. This sterile gel can be directly applied over the wound.
9. Preparation of polyalkylene oxide-collagen composite hydrogel wound dressing 3 grams of PEG diacrylate. synthesized by a procedure described in example Bl . is dissolved in 12 grams of phosphate buffer solution ( pH 7.4. 0.2 g/L KCl. 0.2 g/L of KH2P04, 8.0 g/L of NaCl and 1.15 g/L Na.HPO, ) . 60 microliter of photoinitiator solution (300 mg of Darocur® 2959. Ciba Geigy, dissolved in 700 mg ethanol) is added to the macromonomer solution and the mixture is then sterile filtered into 25 ml sterile glass vial. 100 mg of sterile collagen lyophilized powder is then added to the macromonomer solution. The dispersion is well mixed and then transferred into transparent plastic mold (cavity size 10 cm x 5 cm x 2 mm). The dispersion is then exposed to long wavelength UV light (Blak Ray light source, model 3- 100A. Flood 365 nm. intensity 30 mW/cnr) for 5 minutes. The polymerized, flexible gel is then removed from the mold and used as poiyalkyleneoxide-collagen hydrogei wound dressing.
10. In situ formation of tissue conformal wound dressing using aqueous thermosensitive macromonomer solutions
10 grams of thermosensitive macromonomer (synthesized as described in example
B5) is dissolved in 20 ml cold (0-15 °C) PBS solution. To this solution, 90 microliter of Darocur 2959 solution ( 300 mg in 0.7 ml ethanol) is added and mixed. The cold solution is then transferred into 50 ml cold syringe and filtered using 0.2 micron syringe filter into another vial, (the solution should be cold (0-5 °C) in order to filter).
The filtered cold solution is directly applied over wound surface or injected into a wound cavity The cold solution conforms to the wound geometry or cavity and the body temperature causes physical gelation of the macromonomer The physically gelled solution is then irradiated with long UV light to crosslink the macromonomer solution The light induces chemical crosslinking and forms a chemically crosslinked hydrogel which has good absorptive and mechanical properties The polymerized gel is also non adherent to the tissue and can be easily removed from the wound site
It is evident from the above results and discussion that an improved method of preparing protein concentrates, and particularly fibrinogen rich compositions, from blood compositions such as whole blood and plasma are provided The subject methods are simple and easv to practice and require less time than prior methods of preparing fibrinogen rich compositions Furthermore, the above methods allow greater control over the composition ot the fibrinogen rich compositions than do prior preparation methods Importantly, the subject methods provide for protein concentrates in which the proteins are not denatured
All publications and patent applications cited in this specification are herein incoφorated by reference as if each individual publication or patent application were specifically and individually indicated to be incoφorated b\ reference 1 he citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention
Although the foregoing invention has been described in some detail bv way o^ illustration and example for puφoses of clarity of understanding, it is readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims
-m

Claims

WHAT IS CLAIMED IS
1 A method of preparing a protein concentrate, said method comprising contacting an initial protein comprising composition with a non-protein denaturant hydrogel, maintaining said contact for a period of time sufficient for said hydrogel to absorb a substantial amount of at least the water from said composition to produce a swollen hydrogel and a protein rich phase, and separating said protein rich phase from said swollen hydrogel, whereby said protein concentrate is produced
2 The method according to Claim 1. wherein said hydrogel is biocompatible
3 I he method according to Claim 1. wherein said h\ drogel is a polyether
4 The method according to Claim 1. wherein said protein comprising composition is a physiological fluid composition
5 The method according to Claim 1. wherein said physiological fluid composition is a blood composition
6 A method of preparing a fibrinogen rich composition, said method comprising contacting plasma with a biocompatible non-protein denaturant
Figure imgf000041_0001
drogel capable of increasing in mass upon contact with said plasma to produce a mixture, maintaining said contact for a period of time sufficient for said hydrogel to absorb a substantial amount of at least the water from said plasma to produce a swollen hydrogel and a fibrinogen rich phase, and separating said fibrinogen rich phase from said swollen hydrogel, whereby said fibrinogen rich composition is produced
7 The method according to Claim 6. wherein said plasma comprises an anticoagulant
8 The method according to Claim 6, wherein said method further comprises obtaining blood from a mammalian host and preparing said plasma from said blood
9 The method according to Claim 6. wherein said hydrogel is a polyether
10 The method according to Claim 6. wherein said separating comprises centrifugation
1 1 The protein concentrate composition prepared according to Claim 1
12 The fibrinogen rich composition prepared according to Claim 6
13 A fibrinogen rich composition comprising fibrinogen and substantially no albumin
14 The fibrinogen rich composition according to Claim 1 . wherein said composition further comprises Factor XIII
15 1 he fibrinogen rich composition according to Claim 13. wherein said composition does not comprise components having a moleculai weight of less than about 100 kd
16 The fibrinogen rich composition according to Claim 13 wherein said composition further comprises growth factors
17 I he fibrinogen rich composition according to Claim 13 wherein said composition comprises an active agent
18 1 he fibrinogen rich composition according to Claim 17. wherein said active agent is present in a biodegradable vehicle
19. The fibrinogen rich composition according to Claim 13. wherein said composition further comprises an visualization agent.
20. The fibrinogen rich composition according to Claim 19, wherein said visualization agent is a biocompatible dye.
21. A kit for use in preparing a fibrin gel, said kit comprising: the fibrinogen composition according to Claim 1 1 ; and a coagulation agent comprising thrombin and calcium ion.
22. The kit according to Claim 21 , wherein said kit further comprises an active agent.
23. The kit according to Claim 21. wherein said active agent is present in a biodegradable vehicle.
24. The kit according to Claim 21. wherein said kit further comprises an visualization agent.
25. A kit for use in the preparation of a protein concentrate, said kit comprising: a biocompatible hydrogel; and instructions for preparation of said protein concentrate composition, wherein said instructions comprise directions for performing a method according to Claim 1.
26. The kit according to Claim 25. wherein said kit is sterile.
27. The kit according to Claim 25. wherein said kit further comprises an anticoagulant.
28. The kit according to Claim 25. wherein said kit further comprises containers designed for use in said method of preparation.
29 A kit for use in the preparation of a fibrinogen rich composition, said kit comprising a biocompatible hydrogel, an anticoagulant, and one or more containers for combining said hydrogel and anticoagulant with whole blood or a derivative thereof, wherein said components are sterile
30 A method for adhering a first article to a second article, said method comprising applying at a site of said first article the fibrinogen rich composition of Claim 1 1 and a coagulating agent comprising thrombin and calcium ion, contacting said second article to said first article at said site, whereby said first article is adhered to said second article
31 I he method according to Claim 30. wherein said first and second articles are physiological tissue and said site is a tissue repair site
32 A fibrin sealant comprising an active agent in an encapsulating vehicle
33 The fibrin sealant according to Claim 32 wherein said encapsulating \ chicle is a biodegradeable microencapsulation matrix
34 1 he fibrin sealant according to Claim 32. wherein said encapsulating \ehicle is a cyclodextπn
35 The fibrin sealant according to Claim 32. wherein said fibrin sealant is prepared from a fibrinogen rich composition according to Claim 1 1
36 A fibrin sealant comprising a visualization agent
37. The fibrin sealant according to Claim 36. wherein said visualization agent is a fluorescent agent.
38. The fibrin sealant according to Claim 36. wherein said visualization agent is indocyanine green.
39. The fibrin sealant according to Claim 36. wherein said fibrin sealant is prepared from a fibrinogen rich composition according to Claim 1 1.
40. A polymeric crosslinking agent comprising: an inert water soluble polymeric component: a biodegradable component: and a protein reactive functional component: with the proviso that when said crosslinking agent is difunctional. said biodegradable component is peptidic.
41. The crosslinking agent according to Claim 40. wherein said crosslinking agent is linear.
42. The crosslinking agent according to Claim 40. wherein said crosslinking agent comprises a plurality of branches, wherein said plurality is greater than two.
43. The crosslinking agent of Claim 42. wherein said inert polymeric component is flanked at each end with said biodegradable component which is flanked at each end with said reactive functional group.
44. A method of preparing a wound dressing, said method comprising: combining a an aqueous fluid comprising one or more physiologically active agents with a monomeric composition capable of polymerizing into a biocompatible hydrogel. and polymerizing said monomeric composition; whereby said wound dressing is produced.
45 The method according to Claim 44. wherein said physiological fluid is natural or synthetic.
46 The method according to Claim 44 wherein said monomeric composition further comprising a crosslinking agent
47 The wound dressing produced according to Ciaim 44
48 A wound dressing comprising an aqueous fluid comprising a physiologically active agent entrapped in a biocompatible hydrogel polymeric matrix.
49 The wound dressing according to Claim 48. wherein said dressing comprises at least one of hyaluronic acid and collagen
50 The wound dressing according to Claim 49. wherein said hydrogel is a polyether
51 A kit for preparing a wound dressing according to the method of Claim 44. said kit comprising said monomeric composition and instructions for preparing said wound dressing
52 A device for preparing a protein concentrate from a physiological fluid, said device comprising a hydrogel: a containment means for housing said physiological fluid. an entry means for introducing said fluid into said containment means, and a contacting means for ensuring fluid transfer contact of said hydrogel and said fluid in said containment means
53 The device according to Claim 52. wherein said contacting means is a selective filtering means which is impermeable to hydrogel
54. The device according to Claim 52. wherein said device further comprises a concentrate collection compartment.
55. The device according to Claim 54, wherein said device further comprises an exit port in fluid communication with said collection compartment.
56. The device according to Claim 55. wherein the walls of said collection compartment are impermeable to fluid.
PCT/US1997/016897 1996-09-23 1997-09-22 Methods and devices for preparing protein concentrates WO1998012274A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
AU46486/97A AU4648697A (en) 1996-09-23 1997-09-22 Methods and devices for preparing protein concentrates
US10/010,715 US7009034B2 (en) 1996-09-23 2001-11-09 Biocompatible crosslinked polymers
US10/068,807 US6887974B2 (en) 1996-09-23 2002-02-05 Crosslinking agents and methods of use
US10/293,453 US7211651B2 (en) 1996-09-23 2002-11-13 Proteinaceous gels having visualization agents and methods of use thereof
US10/364,592 US7057019B2 (en) 1996-09-23 2003-02-11 Crosslinked albumin hydrogels
US11/293,892 US7332566B2 (en) 1996-09-23 2005-12-02 Biocompatible crosslinked polymers with visualization agents
US11/725,751 US7605232B2 (en) 1996-09-23 2007-03-20 Hydrogels for protein concentration
US12/069,821 US7592418B2 (en) 1996-09-23 2008-02-13 Biocompatible crosslinked polymers with visualization agents
US12/156,085 US8003705B2 (en) 1996-09-23 2008-05-29 Biocompatible hydrogels made with small molecule precursors
US12/496,060 US20090324721A1 (en) 1996-09-23 2009-07-01 Hydrogels Suitable For Use In Polyp Removal
US12/555,970 US8557535B2 (en) 1996-09-23 2009-09-09 Methods for preparation of platelet rich plasma

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US2652696P 1996-09-23 1996-09-23
US60/026,526 1996-09-23
US3990497P 1997-03-04 1997-03-04
US60/039,904 1997-03-04
US4041797P 1997-03-13 1997-03-13
US60/040,417 1997-03-13

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US09147897 A-371-Of-International 1999-08-30
US09/454,900 Continuation-In-Part US6566406B1 (en) 1996-09-23 1999-12-03 Biocompatible crosslinked polymers
US10/010,715 Continuation-In-Part US7009034B2 (en) 1996-09-23 2001-11-09 Biocompatible crosslinked polymers
US10/068,807 Division US6887974B2 (en) 1996-09-23 2002-02-05 Crosslinking agents and methods of use

Publications (1)

Publication Number Publication Date
WO1998012274A1 true WO1998012274A1 (en) 1998-03-26

Family

ID=27362790

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/016897 WO1998012274A1 (en) 1996-09-23 1997-09-22 Methods and devices for preparing protein concentrates

Country Status (3)

Country Link
US (5) US6887974B2 (en)
AU (1) AU4648697A (en)
WO (1) WO1998012274A1 (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1032367A2 (en) * 1997-11-17 2000-09-06 Haemacure Corporation Fibrin sealants or adhesives comprising a hyaluronic acid derivative material
JP2001278917A (en) * 2000-03-29 2001-10-10 Toagosei Co Ltd Composition
US6514534B1 (en) 1998-08-14 2003-02-04 Incept Llc Methods for forming regional tissue adherent barriers and drug delivery systems
WO2004009207A1 (en) * 2002-07-18 2004-01-29 Hanuman Llc Plasma concentrating apparatus and method
US6899889B1 (en) * 1998-11-06 2005-05-31 Neomend, Inc. Biocompatible material composition adaptable to diverse therapeutic indications
US6905612B2 (en) 2003-03-21 2005-06-14 Hanuman Llc Plasma concentrate apparatus and method
US6965014B1 (en) * 1996-01-16 2005-11-15 Baxter International Inc. Fibrin material and method for producing and using the same
EP1695724A1 (en) * 2001-05-09 2006-08-30 Baxter International Inc. Fibrin material and method for producing and using the same
US7282584B2 (en) 2004-06-16 2007-10-16 Straumann Holding Ag Methylene blue
WO2009014776A1 (en) * 2007-07-23 2009-01-29 Carnegie Mellon University Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
WO2008134774A3 (en) * 2007-04-27 2009-11-26 Biomet Manufacturing Corp. Blood products with binding agent
US7741427B2 (en) 2004-06-16 2010-06-22 Straumann Holding Ag Barrier membrane
US7862538B2 (en) 2008-02-04 2011-01-04 Incept Llc Surgical delivery system for medical sealant
US7872068B2 (en) 2006-05-30 2011-01-18 Incept Llc Materials formable in situ within a medical device
US7919112B2 (en) 2004-08-26 2011-04-05 Pathak Holdings, Llc Implantable tissue compositions and method
US8383161B2 (en) 2009-12-15 2013-02-26 Incept, Llc Radioopaque covalently crosslinked hydrogel particle implants
US8383144B2 (en) 1998-11-06 2013-02-26 Neomend, Inc. Tissue adhering compositions
US8409606B2 (en) 2009-02-12 2013-04-02 Incept, Llc Drug delivery through hydrogel plugs
US8409249B2 (en) 1998-11-06 2013-04-02 Neomend, Inc. Systems, methods, and compositions for achieving closure of suture sites
US8529959B2 (en) 2006-10-17 2013-09-10 Carmell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US8529960B2 (en) 2002-03-18 2013-09-10 Carnell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US8795709B2 (en) 2006-03-29 2014-08-05 Incept Llc Superabsorbent, freeze dried hydrogels for medical applications
US8961501B2 (en) 2010-09-17 2015-02-24 Incept, Llc Method for applying flowable hydrogels to a cornea
US8986730B2 (en) 2004-11-05 2015-03-24 Incept, Llc Methods for sealing a vascular puncture
US9011687B2 (en) 2005-04-27 2015-04-21 Biomet Biologics, Llc Method and apparatus for producing autologous clotting components
US9125807B2 (en) 2007-07-09 2015-09-08 Incept Llc Adhesive hydrogels for ophthalmic drug delivery
US9180142B2 (en) 2009-01-27 2015-11-10 Reddress Ltd. Wound dressings, methods and apparatus for making same and storage and use thereof
WO2016084097A1 (en) * 2014-11-13 2016-06-02 Hemarus Therapeutics Ltd. Process for producing fibrinogen and thrombin
US9463004B2 (en) 2009-05-04 2016-10-11 Incept, Llc. Biomaterials for track and puncture closure
US9550028B2 (en) 2014-05-06 2017-01-24 Biomet Biologics, LLC. Single step desiccating bead-in-syringe concentrating device
EP3113722A4 (en) * 2014-03-07 2017-12-06 Endologix, Inc. Forming hydrogels and materials therefor
CN114096286A (en) * 2019-07-12 2022-02-25 加特技术公司 Biocompatible flexible hemostatic tablet
US11298444B2 (en) 2005-04-01 2022-04-12 Trivascular, Inc. Non-degradable, low swelling, water soluble radiopaque hydrogel polymer

Families Citing this family (187)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8603511B2 (en) 1996-08-27 2013-12-10 Baxter International, Inc. Fragmented polymeric compositions and methods for their use
US6066325A (en) * 1996-08-27 2000-05-23 Fusion Medical Technologies, Inc. Fragmented polymeric compositions and methods for their use
US6478808B2 (en) 1997-12-17 2002-11-12 Closys Corporation Clotting cascade initiating apparatus and methods of use and methods of closing wounds
US6159232A (en) 1997-12-16 2000-12-12 Closys Corporation Clotting cascade initiating apparatus and methods of use and methods of closing wounds
US6632457B1 (en) 1998-08-14 2003-10-14 Incept Llc Composite hydrogel drug delivery systems
AU778318B2 (en) * 2000-02-03 2004-11-25 Tissuemed Limited Device for the closure of a surgical puncture
US6410059B1 (en) * 2000-10-20 2002-06-25 Council Of Scientific And Industrial Research Pharmaceutical composition containing cow urine distillate and an antibiotic
US9080146B2 (en) 2001-01-11 2015-07-14 Celonova Biosciences, Inc. Substrates containing polyphosphazene as matrices and substrates containing polyphosphazene with a micro-structured surface
US7172739B2 (en) * 2001-05-22 2007-02-06 University Of Vermont And State Agricultural College Protein fractionation
WO2003017987A1 (en) * 2001-08-31 2003-03-06 Mcgill University Biodegradable polymeric nanocapsules and uses thereof
US7005143B2 (en) * 2002-04-12 2006-02-28 3M Innovative Properties Company Gel materials, medical articles, and methods
US7329414B2 (en) 2002-05-03 2008-02-12 Biopsy Sciences, Llc Biodegradable polymer for marking tissue and sealing tracts
US20030205538A1 (en) 2002-05-03 2003-11-06 Randel Dorian Methods and apparatus for isolating platelets from blood
US7179391B2 (en) 2002-05-24 2007-02-20 Biomet Manufacturing Corp. Apparatus and method for separating and concentrating fluids containing multiple components
US20040182795A1 (en) * 2003-03-21 2004-09-23 Randel Dorian Apparatus and method for concentration of plasma from whole blood
US7832566B2 (en) 2002-05-24 2010-11-16 Biomet Biologics, Llc Method and apparatus for separating and concentrating a component from a multi-component material including macroparticles
US7992725B2 (en) 2002-05-03 2011-08-09 Biomet Biologics, Llc Buoy suspension fractionation system
US20060278588A1 (en) 2002-05-24 2006-12-14 Woodell-May Jennifer E Apparatus and method for separating and concentrating fluids containing multiple components
US7845499B2 (en) 2002-05-24 2010-12-07 Biomet Biologics, Llc Apparatus and method for separating and concentrating fluids containing multiple components
EP1555957A4 (en) * 2002-10-04 2010-11-24 Nanomatrix Inc Sealants for skin and other tissues
NL1021843C2 (en) * 2002-11-05 2004-05-07 Univ Twente Biomedical structures of poly (1,3-trimethylene carbonate) (poly (TMC)).
US7883500B2 (en) * 2003-03-26 2011-02-08 G&L Consulting, Llc Method and system to treat and prevent myocardial infarct expansion
DE602004025217D1 (en) * 2003-04-04 2010-03-11 Tissuemed Ltd
US7331979B2 (en) * 2003-06-04 2008-02-19 Access Closure, Inc. Apparatus and methods for sealing a vascular puncture
US9289195B2 (en) 2003-06-04 2016-03-22 Access Closure, Inc. Auto-retraction apparatus and methods for sealing a vascular puncture
US20060235114A1 (en) * 2003-07-28 2006-10-19 Teijin Limited Temperature-responsive hydrogel
US20060019868A1 (en) * 2004-01-30 2006-01-26 Pendharkar Sanyog M Hemostatic compositions and devices
US8440225B2 (en) * 2003-08-07 2013-05-14 Ethicon, Inc. Process of making flowable hemostatic compositions and devices containing such compositions
US7927626B2 (en) * 2003-08-07 2011-04-19 Ethicon, Inc. Process of making flowable hemostatic compositions and devices containing such compositions
US7786213B2 (en) * 2003-10-15 2010-08-31 The Regents Of The University Of California Biomacromolecule polymer conjugates
US20050239045A1 (en) * 2003-12-08 2005-10-27 Arkray Inc. Microorganism or cell collecting method, and microorganism or cell collecting implement used for the method
US20050245876A1 (en) * 2003-12-24 2005-11-03 Accessclosure, Inc. Apparatus and methods for facilitating access through a puncture including sealing compound therein
US20050191286A1 (en) * 2004-02-09 2005-09-01 Gandy James B. Lyophilized platelet rich plasma for the use in wound healing (chronic or acute) and bone or tissue grafts or repair
GB0403406D0 (en) * 2004-02-17 2004-03-24 Biotransys Ltd Preparation of carriers for drug delivery and other therapeutic applications
GB0416285D0 (en) * 2004-07-21 2004-08-25 Ciba Spec Chem Water Treat Ltd Method of treating polymers
MX2007001554A (en) * 2004-08-03 2007-04-10 Tissuemed Ltd Tissue-adhesive materials.
US8348971B2 (en) 2004-08-27 2013-01-08 Accessclosure, Inc. Apparatus and methods for facilitating hemostasis within a vascular puncture
US9801982B2 (en) * 2004-09-28 2017-10-31 Atrium Medical Corporation Implantable barrier device
US8263102B2 (en) 2004-09-28 2012-09-11 Atrium Medical Corporation Drug delivery coating for use with a stent
US8312836B2 (en) * 2004-09-28 2012-11-20 Atrium Medical Corporation Method and apparatus for application of a fresh coating on a medical device
US9012506B2 (en) 2004-09-28 2015-04-21 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US8367099B2 (en) 2004-09-28 2013-02-05 Atrium Medical Corporation Perforated fatty acid films
US20090011116A1 (en) * 2004-09-28 2009-01-08 Atrium Medical Corporation Reducing template with coating receptacle containing a medical device to be coated
WO2006036984A2 (en) * 2004-09-28 2006-04-06 Atrium Medical Corporation Stand-alone film and methods for making the same
US9000040B2 (en) 2004-09-28 2015-04-07 Atrium Medical Corporation Cross-linked fatty acid-based biomaterials
US20210299056A9 (en) 2004-10-25 2021-09-30 Varian Medical Systems, Inc. Color-Coded Polymeric Particles of Predetermined Size for Therapeutic and/or Diagnostic Applications and Related Methods
KR101153785B1 (en) 2004-10-25 2012-07-09 셀로노바 바이오사이언시스 저머니 게엠베하 Loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US9107850B2 (en) 2004-10-25 2015-08-18 Celonova Biosciences, Inc. Color-coded and sized loadable polymeric particles for therapeutic and/or diagnostic applications and methods of preparing and using the same
US9114162B2 (en) 2004-10-25 2015-08-25 Celonova Biosciences, Inc. Loadable polymeric particles for enhanced imaging in clinical applications and methods of preparing and using the same
US8262693B2 (en) * 2004-11-05 2012-09-11 Accessclosure, Inc. Apparatus and methods for sealing a vascular puncture
US7201918B2 (en) * 2004-11-16 2007-04-10 Microvention, Inc. Compositions, systems and methods for treatment of defects in blood vessels
JP4510898B2 (en) * 2005-02-07 2010-07-28 ハヌマン リミテッド ライアビリティ カンパニー Plasma concentrator
US7611494B2 (en) 2005-02-08 2009-11-03 Confluent Surgical, Inc. Spray for fluent materials
US7806856B2 (en) * 2005-04-22 2010-10-05 Accessclosure, Inc. Apparatus and method for temporary hemostasis
JP5038294B2 (en) 2005-04-22 2012-10-03 アクセスクロージャー,インク. Device and method for blocking tissue puncture
US8002742B2 (en) * 2005-04-22 2011-08-23 Accessclosure, Inc. Apparatus and methods for sealing a puncture in tissue
US20070100199A1 (en) * 2005-11-03 2007-05-03 Lilip Lau Apparatus and method of delivering biomaterial to the heart
BRPI0520502A2 (en) * 2005-08-30 2009-06-13 Mackie J Walker Jr dehydrated composition usable for mammal therapy, wound healing composition, wound healing method, wound healing, and wound healing method
ES2377479T3 (en) * 2005-09-27 2012-03-28 Universite Joseph Fourier - Grenoble 1 HYDROGEL FUNCTIONED WITH A POLYMERIZABLE PORTION AND ITS USES AS BIOSENSORS OR BIORREACTORS.
US9427423B2 (en) 2009-03-10 2016-08-30 Atrium Medical Corporation Fatty-acid based particles
US9278161B2 (en) 2005-09-28 2016-03-08 Atrium Medical Corporation Tissue-separating fatty acid adhesion barrier
US20070087061A1 (en) * 2005-10-14 2007-04-19 Medafor, Incorporated Method and composition for creating and/or activating a platelet-rich gel by contact with a porous particulate material, for use in wound care, tissue adhesion, or as a matrix for delivery of therapeutic components
CA2626030A1 (en) 2005-10-15 2007-04-26 Atrium Medical Corporation Hydrophobic cross-linked gels for bioabsorbable drug carrier coatings
EP1951260A4 (en) * 2005-10-21 2009-11-11 Bezwada Biomedical Llc Functionalized phenolic compounds and absorbable therefrom
DE602007007725D1 (en) * 2006-01-17 2010-08-26 Baxter Healthcare Sa MIXING DEVICE, SYSTEM AND METHOD
US20090038701A1 (en) 2006-01-17 2009-02-12 Baxter International Inc. Device, system and method for mixing
CA2640629A1 (en) 2006-02-03 2007-08-09 Tissuemed Limited Tissue-adhesive materials
US20090018575A1 (en) * 2006-03-01 2009-01-15 Tissuemed Limited Tissue-adhesive formulations
US7597882B2 (en) * 2006-04-24 2009-10-06 Incept Llc Protein crosslinkers, crosslinking methods and applications thereof
GB0609778D0 (en) * 2006-05-17 2006-06-28 Agt Sciences Ltd Delivery means
US8567609B2 (en) 2006-05-25 2013-10-29 Biomet Biologics, Llc Apparatus and method for separating and concentrating fluids containing multiple components
CA2655026C (en) 2006-06-15 2016-08-02 Microvention, Inc. Embolization device constructed from expansible polymer
CA2662169C (en) * 2006-09-08 2018-03-20 Symphony Medical, Inc. Intramyocardial patterning for global cardiac resizing and reshaping
EP2077765A2 (en) 2006-09-13 2009-07-15 Accessclosure, Inc. Apparatus for sealing a vascular puncture
JP2008096503A (en) * 2006-10-06 2008-04-24 Toshiba Corp Method for manufacturing powdery photosensitive composition, photosensitive composition and optical recording medium formed from this photosensitive composition
US9492596B2 (en) * 2006-11-06 2016-11-15 Atrium Medical Corporation Barrier layer with underlying medical device and one or more reinforcing support structures
WO2008057344A2 (en) * 2006-11-06 2008-05-15 Atrium Medical Corporation Coated surgical mesh
AU2008214319A1 (en) * 2007-02-06 2008-08-14 Incept, Llc Polymerization with precipitation of proteins for elution in physiological solution
EP2142208A4 (en) * 2007-04-03 2013-01-16 Aeris Therapeutics Llc Lung volume reduction therapy using crosslinked biopolymers
EP2146667A2 (en) * 2007-04-11 2010-01-27 Henry Ford Health System Cardiac repair, resizing and reshaping using the venous system of the heart
US8328024B2 (en) 2007-04-12 2012-12-11 Hanuman, Llc Buoy suspension fractionation system
JP5479319B2 (en) 2007-04-12 2014-04-23 バイオメット・バイオロジックス・リミテッド・ライアビリティ・カンパニー Buoy suspension fractionation system
CN101711168B (en) 2007-04-13 2013-05-01 库罗斯生物外科股份公司 Polymeric tissue sealant
US20080287633A1 (en) * 2007-05-18 2008-11-20 Drumheller Paul D Hydrogel Materials
WO2008154033A2 (en) * 2007-06-11 2008-12-18 Symphony Medical, Inc. Cardiac patterning for improving diastolic function
EP2011524A1 (en) 2007-07-02 2009-01-07 Omrix Biopharmaceuticals Ltd. Fibrin glue with a visualization agent
GB0715514D0 (en) * 2007-08-10 2007-09-19 Tissuemed Ltd Coated medical devices
US8217134B2 (en) * 2007-08-30 2012-07-10 Bezwada Biomedical, Llc Controlled release of biologically active compounds
US8026285B2 (en) 2007-09-04 2011-09-27 Bezwada Biomedical, Llc Control release of biologically active compounds from multi-armed oligomers
US8048980B2 (en) * 2007-09-17 2011-11-01 Bezwada Biomedical, Llc Hydrolysable linkers and cross-linkers for absorbable polymers
US8053591B2 (en) * 2007-09-26 2011-11-08 Bezwada Biomedical, Llc Functionalized biodegradable triclosan monomers and oligomers for controlled release
US7993367B2 (en) 2007-09-28 2011-08-09 Accessclosure, Inc. Apparatus and methods for sealing a vascular puncture
EP2209426A4 (en) * 2007-11-02 2015-04-22 Incept Llc Apparatus and methods for sealing a vascular puncture
CA2709379C (en) 2007-12-21 2016-08-16 Microvention, Inc. Hydrogel filaments for biomedical uses
EP2567692B1 (en) 2008-02-27 2016-04-06 Biomet Biologics, LLC Use of a device for obtaining interleukin-1 receptor antagonist rich solutions
WO2009111338A1 (en) * 2008-02-29 2009-09-11 Biomet Manufacturing Corp. A system and process for separating a material
US8642831B2 (en) 2008-02-29 2014-02-04 Ferrosan Medical Devices A/S Device for promotion of hemostasis and/or wound healing
US9386990B2 (en) * 2008-03-19 2016-07-12 University Of Florida Research Foundation, Inc. Nerve repair with a hydrogel and adhesive
US8029533B2 (en) 2008-04-04 2011-10-04 Accessclosure, Inc. Apparatus and methods for sealing a vascular puncture
US9364206B2 (en) 2008-04-04 2016-06-14 Access Closure, Inc. Apparatus and methods for sealing a vascular puncture
US20090259210A1 (en) * 2008-04-10 2009-10-15 Sabbah Hani N Method, apparatus and kits for forming structural members within the cardiac venous system
US8263704B2 (en) * 2008-04-23 2012-09-11 Tyco Healthcare Group Lp Bioabsorbable surgical composition
CN101998868B (en) * 2008-05-01 2014-06-04 泰尔茂株式会社 Visible medical treatment material
US8367747B2 (en) * 2008-05-23 2013-02-05 Bezwada Biomedical, Llc Bioabsorbable polymers from bioabsorbable polyisocyanates and uses thereof
US8207264B2 (en) * 2008-07-11 2012-06-26 Tyco Healthcare Group Lp Functionalized inclusion complexes as crosslinkers
US20100092533A1 (en) 2008-10-15 2010-04-15 Joshua Stopek Bioabsorbable Surgical Composition
EP2364112B1 (en) 2008-11-12 2016-03-09 Accessclosure, Inc. Apparatus and methods for sealing a vascular puncture
US8187475B2 (en) 2009-03-06 2012-05-29 Biomet Biologics, Llc Method and apparatus for producing autologous thrombin
US20100246316A1 (en) * 2009-03-31 2010-09-30 Baxter International Inc. Dispenser, kit and mixing adapter
US8313954B2 (en) 2009-04-03 2012-11-20 Biomet Biologics, Llc All-in-one means of separating blood components
US9463260B2 (en) * 2009-06-29 2016-10-11 Covidien Lp Self-sealing compositions
US9011800B2 (en) 2009-07-16 2015-04-21 Biomet Biologics, Llc Method and apparatus for separating biological materials
US20110038910A1 (en) 2009-08-11 2011-02-17 Atrium Medical Corporation Anti-infective antimicrobial-containing biomaterials
US9271925B2 (en) 2013-03-11 2016-03-01 Bioinspire Technologies, Inc. Multi-layer biodegradable device having adjustable drug release profile
EP2477617B1 (en) 2009-09-18 2018-01-31 Bioinspire Technologies Inc. Free-standing biodegradable patch
CN102665606A (en) 2009-09-24 2012-09-12 微排放器公司 Injectable hydrogel filaments for biomedical uses
KR101745748B1 (en) 2009-10-26 2017-06-12 마이크로벤션, 인코포레이티드 Embolization device constructed from expansile polymer
EP2498764B1 (en) 2009-11-09 2017-09-06 Spotlight Technology Partners LLC Fragmented hydrogels
CA2780294C (en) 2009-11-09 2018-01-16 Spotlight Technology Partners Llc Polysaccharide based hydrogels
US8641661B2 (en) 2010-01-05 2014-02-04 Baxter International Inc. Mixing system, kit and mixer adapter
CA2726566A1 (en) * 2010-01-11 2011-07-11 Baxter International Inc. Pipette system, pipette tip assembly and kit
US8591391B2 (en) 2010-04-12 2013-11-26 Biomet Biologics, Llc Method and apparatus for separating a material
WO2011140517A2 (en) 2010-05-07 2011-11-10 Medicus Biosciences, Llc Methods for treating diseases of the lung
US20110282464A1 (en) 2010-05-12 2011-11-17 Timothy Sargeant Reactive Surgical Implants
US10231721B2 (en) 2010-06-24 2019-03-19 St. Croix Surgical Systems, Llc Failsafe percutaneous wound barrier
US10322213B2 (en) 2010-07-16 2019-06-18 Atrium Medical Corporation Compositions and methods for altering the rate of hydrolysis of cured oil-based materials
US20120065600A1 (en) * 2010-09-10 2012-03-15 E. I. Du Pont De Nemours And Company. Device for dispensing microliter quantities of a material into a longitudinally extending wound site
US8518440B2 (en) 2010-12-21 2013-08-27 Confluent Surgical, Inc. Biodegradable osmotic pump implant for drug delivery
KR101272484B1 (en) * 2011-01-19 2013-06-10 세원셀론텍(주) A radiation crosslinked collagen gel, its manufacture and its usage method
ES2870965T3 (en) 2011-01-19 2021-10-28 Access Closure Inc Procedures to seal a vascular puncture
US9820728B2 (en) 2011-01-19 2017-11-21 Access Closure, Inc. Apparatus and methods for sealing a vascular puncture
WO2012145431A2 (en) 2011-04-18 2012-10-26 Microvention, Inc. Embolic devices
US9386968B2 (en) 2011-05-11 2016-07-12 Access Closure, Inc. Apparatus and methods for sealing a vascular puncture
US8968926B2 (en) 2011-07-15 2015-03-03 Covidien Lp Degradable implantable galvanic power source
US8968927B2 (en) 2011-07-15 2015-03-03 Covidien Lp Degradable implantable battery
US11083821B2 (en) 2011-08-10 2021-08-10 C.P. Medical Corporation Biocompatible hydrogel polymer formulations for the controlled delivery of biomolecules
US10111985B2 (en) 2011-08-10 2018-10-30 Medicus Biosciences, Llc Biocompatible hydrogel polymer formulations for the controlled delivery of biomolecules
US10226417B2 (en) 2011-09-16 2019-03-12 Peter Jarrett Drug delivery systems and applications
KR20190090048A (en) 2011-12-05 2019-07-31 인셉트, 엘엘씨 Medical organogel processes and compositions
US20130202656A1 (en) 2012-02-03 2013-08-08 Dynasil Biomedical Corporation Systems and kits for the fabrication of tissue patches
WO2013131520A2 (en) 2012-03-06 2013-09-12 Ferrosan Medical Devices A/S Pressurized container containing haemostatic paste
US8721680B2 (en) 2012-03-23 2014-05-13 Accessclosure, Inc. Apparatus and methods for sealing a vascular puncture
US9757105B2 (en) 2012-03-23 2017-09-12 Accessclosure, Inc. Apparatus and methods for sealing a vascular puncture
US9011884B2 (en) 2012-04-18 2015-04-21 Microvention, Inc. Embolic devices
CN104428014B (en) 2012-05-11 2016-11-09 梅迪卡斯生物科学有限责任公司 Biocompatible hydrogel for detachment of retina is treated
CN104487103A (en) * 2012-05-15 2015-04-01 泰克尼恩研究和发展基金有限公司 Fiber-reinforced hydrogel composites and methods of forming fiber-reinforced hydrogel composites
RU2636240C2 (en) 2012-06-12 2017-11-21 Ферросан Медикал Дивайсиз А/С Dry haemostatic composition
US9867880B2 (en) 2012-06-13 2018-01-16 Atrium Medical Corporation Cured oil-hydrogel biomaterial compositions for controlled drug delivery
US9642956B2 (en) 2012-08-27 2017-05-09 Biomet Biologics, Llc Apparatus and method for separating and concentrating fluids containing multiple components
US20140081356A1 (en) * 2012-09-20 2014-03-20 Georgia Tech Research Corporation Thermal capacitors for minimizing complications and side effects from thermal medicine
US9364199B2 (en) 2013-03-14 2016-06-14 Covidien Lp Medical devices
CA2903823C (en) 2013-03-14 2021-08-31 Medicus Biosciences Llc Solid polyglycol-based biocompatible pre-formulation
US10624865B2 (en) 2013-03-14 2020-04-21 Pathak Holdings Llc Methods, compositions, and devices for drug/live cell microarrays
US20140271589A1 (en) 2013-03-15 2014-09-18 Biomet Biologics, Llc Treatment of collagen defects using protein solutions
US10208095B2 (en) 2013-03-15 2019-02-19 Biomet Manufacturing, Llc Methods for making cytokine compositions from tissues using non-centrifugal methods
US9950035B2 (en) 2013-03-15 2018-04-24 Biomet Biologics, Llc Methods and non-immunogenic compositions for treating inflammatory disorders
US9895418B2 (en) 2013-03-15 2018-02-20 Biomet Biologics, Llc Treatment of peripheral vascular disease using protein solutions
US20140271600A1 (en) * 2013-03-15 2014-09-18 Gabrielle Giet Non-cellular bandage, method of using the same, and method of preparing the same
US10143725B2 (en) 2013-03-15 2018-12-04 Biomet Biologics, Llc Treatment of pain using protein solutions
JP6390873B2 (en) 2013-06-21 2018-09-19 フェッローサン メディカル ディバイス エー/エス Dry composition expanded under reduced pressure and syringe for holding the same
US10842969B2 (en) 2013-10-25 2020-11-24 Mercator Medsystems, Inc. Systems and methods of treating malacia by local delivery of hydrogel to augment tissue
AU2014240353A1 (en) 2013-11-12 2015-05-28 Covidien Lp Degradable implantable battery
RU2678592C1 (en) 2013-12-11 2019-01-30 Ферросан Медикал Дивайсиз А/С Dry composition comprising extrusion enhancer
US10085729B2 (en) 2014-03-06 2018-10-02 Ethicon, Inc. Methods and devices for forming biomedical coatings using variable mixing ratios of multi-part compositions
US10124090B2 (en) 2014-04-03 2018-11-13 Terumo Corporation Embolic devices
EP3129464B1 (en) * 2014-04-10 2019-08-28 The Trustees of Columbia University in the City of New York Methods, compositions, and systems for activation and expansion of cells
US10092663B2 (en) 2014-04-29 2018-10-09 Terumo Corporation Polymers
EP3137124B1 (en) 2014-04-29 2019-01-09 Microvention, Inc. Polymers including active agents
US9295752B1 (en) 2014-09-30 2016-03-29 Covidien Lp Bioadhesive for occluding vessels
BR112017007466B1 (en) 2014-10-13 2021-03-02 Ferrosan Medical Devices A/S method for preparing a dry composition, method for reconstituting the dry composition, paste, dry composition, container, homeostatic kit, and, using a dry composition
JP6747650B2 (en) 2014-12-24 2020-08-26 フェロサン メディカル デバイシーズ エイ/エス Syringe for holding and mixing the first substance and the second substance
CN107921237A (en) 2015-04-27 2018-04-17 反射医学公司 Sympathetic nerve cardiopulmonary neural modulation system and method
WO2016201250A1 (en) 2015-06-11 2016-12-15 Microvention, Inc. Expansile device for implantation
US10918796B2 (en) 2015-07-03 2021-02-16 Ferrosan Medical Devices A/S Syringe for mixing two components and for retaining a vacuum in a storage condition
EP3324902A1 (en) 2015-07-22 2018-05-30 Incept, LLC Coated punctal plug
US9833538B2 (en) 2015-08-07 2017-12-05 Xcede Technologies, Inc. Adhesive compositions and related methods
EP3331577A4 (en) 2015-08-07 2019-07-17 Xcede Technologies, Inc. Adhesive compositions and related methods
US9540548B1 (en) 2015-08-07 2017-01-10 Xcede Technologies, Inc. Adhesive compositions and related methods
US11246879B2 (en) 2016-02-09 2022-02-15 Tulai Therapeutics, Inc. Methods, agents, and devices for local neuromodulation of autonomic nerves
AU2017290353B2 (en) 2016-06-29 2023-05-18 Tulavi Therapeutics, Inc. Treatment of sepsis and related inflammatory conditions by local neuromodulation of the autonomic nervous system
JP2020532565A (en) 2017-09-05 2020-11-12 トルク セラピューティクス, インコーポレイテッド Reversible linker and its use
AU2018328209A1 (en) 2017-09-05 2020-04-23 Torque Therapeutics, Inc. Therapeutic protein compositions and methods of making and using the same
KR20210008479A (en) 2018-05-09 2021-01-22 훼로산 메디칼 디바이스 에이/에스 How to prepare a hemostatic composition
WO2020010164A1 (en) 2018-07-02 2020-01-09 Corinne Bright Methods and devices for in situ formed nerve cap
US20220125996A1 (en) * 2019-01-15 2022-04-28 University Of Massachusetts Eggshell Particle Containing Hydrogels And Prepolymer Compositions For Biomedical Applications
US11739166B2 (en) 2020-07-02 2023-08-29 Davol Inc. Reactive polysaccharide-based hemostatic agent
CN115920118B (en) * 2022-10-12 2023-07-04 浙江大学 Double-crosslinked fibrin gel, kit and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4849458A (en) * 1988-06-17 1989-07-18 Matrix Medica, Inc. Segmented polyether polyurethane
US5192743A (en) * 1992-01-16 1993-03-09 Genentech, Inc. Reconstitutable lyophilized protein formulation
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers

Family Cites Families (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2533004A (en) * 1943-10-27 1950-12-05 John D Ferry Fibrin clots and methods for preparing the same
US3520949A (en) * 1966-07-26 1970-07-21 Nat Patent Dev Corp Hydrophilic polymers,articles and methods of making same
IL47468A (en) * 1975-06-12 1979-05-31 Rehovot Res Prod Process for the cross-linking of proteins using water soluble cross-linking agents
AT359652B (en) * 1979-02-15 1980-11-25 Immuno Ag METHOD FOR PRODUCING A TISSUE ADHESIVE
AT366916B (en) * 1980-04-02 1982-05-25 Immuno Ag DEVICE FOR APPLICATING A TISSUE ADHESIVE BASED ON HUMAN OR ANIMAL PROTEINS
US4565784A (en) * 1981-01-26 1986-01-21 Trustees Of Boston University Hydrogels capable of supporting cell growth
AT379311B (en) * 1984-03-29 1985-12-27 Immuno Ag DEVICE FOR APPLICATING A TISSUE ADHESIVE
US4601286A (en) * 1984-04-20 1986-07-22 Kaufman Jack W Article for the protection of living tissues
DE3675588D1 (en) * 1985-06-19 1990-12-20 Ajinomoto Kk HAEMOGLOBIN TIED TO A POLY (ALKENYLENE OXIDE).
AT382783B (en) * 1985-06-20 1987-04-10 Immuno Ag DEVICE FOR APPLICATING A TISSUE ADHESIVE
US4646730A (en) * 1986-05-23 1987-03-03 Johnson & Johnson Products, Inc. Color stabilized hydrogel dressing and process
DE3722904A1 (en) * 1987-01-09 1988-07-21 Harald Maslanka INJECTION DEVICE WITH DOUBLE CANNULA FOR AN ENDOSCOPE
US4937270A (en) * 1987-09-18 1990-06-26 Genzyme Corporation Water insoluble derivatives of hyaluronic acid
US4978336A (en) * 1987-09-29 1990-12-18 Hemaedics, Inc. Biological syringe system
US5024742A (en) * 1988-02-24 1991-06-18 Cedars-Sinai Medical Center Method of crosslinking amino acid containing polymers using photoactivatable chemical crosslinkers
AT397203B (en) * 1988-05-31 1994-02-25 Immuno Ag FABRIC ADHESIVE
US4874368A (en) * 1988-07-25 1989-10-17 Micromedics, Inc. Fibrin glue delivery system
US4902281A (en) * 1988-08-16 1990-02-20 Corus Medical Corporation Fibrinogen dispensing kit
US5041292A (en) * 1988-08-31 1991-08-20 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
US4938763B1 (en) * 1988-10-03 1995-07-04 Atrix Lab Inc Biodegradable in-situ forming implants and method of producing the same
US5475052A (en) * 1988-11-21 1995-12-12 Collagen Corporation Collagen-synthetic polymer matrices prepared using a multiple step reaction
US5304595A (en) * 1988-11-21 1994-04-19 Collagen Corporation Collagen-polymer conjugates
US5510418A (en) * 1988-11-21 1996-04-23 Collagen Corporation Glycosaminoglycan-synthetic polymer conjugates
US5527856A (en) * 1988-11-21 1996-06-18 Collagen Corporation Method of preparing crosslinked biomaterial compositions for use in tissue augmentation
US5936035A (en) * 1988-11-21 1999-08-10 Cohesion Technologies, Inc. Biocompatible adhesive compositions
US5643464A (en) * 1988-11-21 1997-07-01 Collagen Corporation Process for preparing a sterile, dry crosslinking agent
US5800541A (en) * 1988-11-21 1998-09-01 Collagen Corporation Collagen-synthetic polymer matrices prepared using a multiple step reaction
US5162430A (en) * 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
US5550187A (en) * 1988-11-21 1996-08-27 Collagen Corporation Method of preparing crosslinked biomaterial compositions for use in tissue augmentation
US5614587A (en) * 1988-11-21 1997-03-25 Collagen Corporation Collagen-based bioadhesive compositions
US5565519A (en) * 1988-11-21 1996-10-15 Collagen Corporation Clear, chemically modified collagen-synthetic polymer conjugates for ophthalmic applications
US4932270A (en) * 1989-03-21 1990-06-12 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Torque sensor having a spoked sensor element support structure
US5226877A (en) * 1989-06-23 1993-07-13 Epstein Gordon H Method and apparatus for preparing fibrinogen adhesive from whole blood
US5104909A (en) * 1989-09-21 1992-04-14 W. R. Grace & Co.-Conn. Water-absorbent, high capacity polyurethane foams
US5116315A (en) * 1989-10-03 1992-05-26 Hemaedics, Inc. Biological syringe system
US5059424A (en) * 1989-11-01 1991-10-22 Ndm Acquisition Corp. Hydrogel wound dressing product
US5318524A (en) * 1990-01-03 1994-06-07 Cryolife, Inc. Fibrin sealant delivery kit
US5219328A (en) 1990-01-03 1993-06-15 Cryolife, Inc. Fibrin sealant delivery method
US5030215A (en) * 1990-01-03 1991-07-09 Cryolife, Inc. Preparation of fibrinogen/factor XIII precipitate
US5219564A (en) * 1990-07-06 1993-06-15 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
US5246698A (en) * 1990-07-09 1993-09-21 Biomatrix, Inc. Biocompatible viscoelastic gel slurries, their preparation and use
US5292362A (en) * 1990-07-27 1994-03-08 The Trustees Of Columbia University In The City Of New York Tissue bonding and sealing composition and method of using the same
US5391183A (en) * 1990-09-21 1995-02-21 Datascope Investment Corp Device and method sealing puncture wounds
US5626863A (en) * 1992-02-28 1997-05-06 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5529914A (en) * 1990-10-15 1996-06-25 The Board Of Regents The Univeristy Of Texas System Gels for encapsulation of biological materials
US5143662A (en) * 1991-02-12 1992-09-01 United States Surgical Corporation Process for preparing particles of bioabsorbable polymer
US5296518A (en) * 1991-05-24 1994-03-22 Hampshire Chemical Corp. Hydrophilic polyurethaneurea foams containing no toxic leachable additives and method to produce such foams
US5368563A (en) * 1991-12-18 1994-11-29 Micromedics, Inc. Sprayer assembly for physiologic glue
FR2687681B1 (en) 1992-02-20 1995-10-13 Transgene Sa POLYETHYLENEGLYCOL-HIRUDINE CONJUGATES, THEIR PREPARATION PROCESS AND THEIR USE FOR THE TREATMENT OF THROMBOSES.
WO1993017669A1 (en) * 1992-02-28 1993-09-16 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5573934A (en) * 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
US5762620A (en) * 1992-04-02 1998-06-09 Ndm Acquisition Corp. Wound dressing containing a partially dehydrated hydrogel
IT1260154B (en) * 1992-07-03 1996-03-28 Lanfranco Callegaro HYALURONIC ACID AND ITS DERIVATIVES IN INTERPENETRATING POLYMERS (IPN)
US5514379A (en) 1992-08-07 1996-05-07 The General Hospital Corporation Hydrogel compositions and methods of use
US5582596A (en) * 1992-09-26 1996-12-10 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Applicator for applying a biocompatible adhesive
US5844023A (en) * 1992-11-06 1998-12-01 Bio-Tec Biologische Naturverpackungen Gmbh Biologically degradable polymer mixture
CA2150956A1 (en) 1992-12-10 1994-06-23 Robert G. L. Shorr Glycolipid enzyme-polymer conjugates
AU675252B2 (en) * 1992-12-18 1997-01-30 Tremco, Inc. Fast-curing, high strength, two-part sealants using acetoacetate-amine cure chemistry
US5395923A (en) * 1993-02-23 1995-03-07 Haemacure-Biotech, Inc. Process for the obtention of a biological adhesive made of concentrated coagulation factors by "salting-out"
US5749968A (en) * 1993-03-01 1998-05-12 Focal, Inc. Device for priming for improved adherence of gels to substrates
KR960015447B1 (en) * 1993-03-16 1996-11-14 주식회사 삼양사 Biodegradable polymer
ZA941881B (en) * 1993-04-02 1995-09-18 Lilly Co Eli Manifold medication injection apparatus and method
US5951583A (en) * 1993-05-25 1999-09-14 Vascular Solutions, Inc. Thrombin and collagen procoagulant and process for making the same
US5773025A (en) * 1993-09-09 1998-06-30 Edward Mendell Co., Inc. Sustained release heterodisperse hydrogel systems--amorphous drugs
US5446090A (en) * 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
CA2140053C (en) * 1994-02-09 2000-04-04 Joel S. Rosenblatt Collagen-based injectable drug delivery system and its use
US5474540A (en) * 1994-03-25 1995-12-12 Micromedics, Inc. Fluid separation control attachment for physiologic glue applicator
US5672622A (en) * 1994-04-21 1997-09-30 Berlex Laboratories, Inc. Treatment of multiple sclerosis
KR0141431B1 (en) * 1994-05-17 1998-07-01 김상웅 Biodegradable hydrogel copolymer
US5629384A (en) * 1994-05-17 1997-05-13 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
US5419491A (en) * 1994-05-23 1995-05-30 Mattson Spray Equipment, Inc. Two component fluid spray gun and method
US5583114A (en) 1994-07-27 1996-12-10 Minnesota Mining And Manufacturing Company Adhesive sealant composition
US5585007A (en) * 1994-12-07 1996-12-17 Plasmaseal Corporation Plasma concentrate and tissue sealant methods and apparatuses for making concentrated plasma and/or tissue sealant
US5605541A (en) * 1994-12-07 1997-02-25 E. R. Squibb And Sons, Inc. Fibrin sealant applicatoor
US5810885A (en) * 1994-12-28 1998-09-22 Omrix Biopharm Sa Device for applying one or several fluids
US5932462A (en) * 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
US5900245A (en) * 1996-03-22 1999-05-04 Focal, Inc. Compliant tissue sealants
US5656035A (en) * 1995-04-25 1997-08-12 Avoy; Donald R. Refillable fibrinogen dispensing kit
US5817303A (en) * 1995-05-05 1998-10-06 Protein Polymer Technologies, Inc. Bonding together tissue with adhesive containing polyfunctional crosslinking agent and protein polymer
WO1996041818A1 (en) * 1995-06-09 1996-12-27 Drohan William N Chitin hydrogels, methods of their production and use
ATE342295T1 (en) * 1995-07-28 2006-11-15 Genzyme Corp BIODEGRADABLE MULTIBLOKHYDROGENS AND THEIR USE AS CARRIERS FOR CONTROLLED RELEASE PHARMACOLOGICALLY ACTIVE MATERIALS AND TISSUE CONTACT MATERIALS
US6458889B1 (en) * 1995-12-18 2002-10-01 Cohesion Technologies, Inc. Compositions and systems for forming crosslinked biomaterials and associated methods of preparation and use
US5752974A (en) * 1995-12-18 1998-05-19 Collagen Corporation Injectable or implantable biomaterials for filling or blocking lumens and voids of the body
EP1704878B1 (en) * 1995-12-18 2013-04-10 AngioDevice International GmbH Crosslinked polymer compositions and methods for their use
US5702361A (en) * 1996-01-31 1997-12-30 Micro Therapeutics, Inc. Method for embolizing blood vessels
US5855618A (en) * 1996-09-13 1999-01-05 Meadox Medicals, Inc. Polyurethanes grafted with polyethylene oxide chains containing covalently bonded heparin
US20020064546A1 (en) 1996-09-13 2002-05-30 J. Milton Harris Degradable poly(ethylene glycol) hydrogels with controlled half-life and precursors therefor
US6214966B1 (en) * 1996-09-26 2001-04-10 Shearwater Corporation Soluble, degradable poly(ethylene glycol) derivatives for controllable release of bound molecules into solution
US6258351B1 (en) 1996-11-06 2001-07-10 Shearwater Corporation Delivery of poly(ethylene glycol)-modified molecules from degradable hydrogels
AU6169998A (en) 1997-02-14 1998-09-08 Chandrashekar Pathak Biocompatible polymers and methods for their use
US6371975B2 (en) * 1998-11-06 2002-04-16 Neomend, Inc. Compositions, systems, and methods for creating in situ, chemically cross-linked, mechanical barriers
US6162241A (en) * 1997-08-06 2000-12-19 Focal, Inc. Hemostatic tissue sealants
WO1999010022A2 (en) 1997-08-27 1999-03-04 California Institute Of Technology Methods and compositions to prevent formation of adhesions in biological tissues
US6150505A (en) * 1997-09-19 2000-11-21 Hadasit Medical Research Services & Development Ltd. Fibrin microbeads prepared from fibrinogen, thrombin and factor XIII
AR014621A1 (en) * 1997-12-12 2001-03-28 Macromed Inc A POLY (ETHYLENE GLYCOL) IN THE FORM OF A STAR, HETEROFUNCTIONAL, BIOCOMPATIBLE, METHOD FOR OBTAINING AND METHOD TO CONJUG WITH A PROTEIN
US6153531A (en) 1997-12-22 2000-11-28 Philips Electronics North America Corporation Method for preventing electrochemical erosion of interconnect structures
ES2211033T3 (en) 1998-01-07 2004-07-01 Debio Recherche Pharmaceutique S.A. DEGRADABLE HETEROBIFUNCTIONAL POLYETHYLENGLYCOL ACRYLATES AND GELS AND CONJUGATES DERIVED FROM SUCH ACRYLATES
US6251382B1 (en) * 1998-04-17 2001-06-26 Enzon, Inc. Biodegradable high molecular weight polymeric linkers and their conjugates
US6156531A (en) * 1998-07-20 2000-12-05 Sulzer Carbomedics Inc. Cross-linking tissue with a compound having a C8 to C40 aliphatic chain
WO2000012018A1 (en) 1998-08-26 2000-03-09 Advanced Closure Systems, Inc. Compositions, systems, and methods for creating in situ, chemically cross-linked, mechanical barriers or covering structures
US6110484A (en) * 1998-11-24 2000-08-29 Cohesion Technologies, Inc. Collagen-polymer matrices with differential biodegradability
US6312725B1 (en) * 1999-04-16 2001-11-06 Cohesion Technologies, Inc. Rapid gelling biocompatible polymer composition
US6613325B1 (en) 1999-06-01 2003-09-02 Bristol-Myers Squibb Company Prevention of post surgical adhesions using a fibrin monomer sealant

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4849458A (en) * 1988-06-17 1989-07-18 Matrix Medica, Inc. Segmented polyether polyurethane
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5192743A (en) * 1992-01-16 1993-03-09 Genentech, Inc. Reconstitutable lyophilized protein formulation

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6965014B1 (en) * 1996-01-16 2005-11-15 Baxter International Inc. Fibrin material and method for producing and using the same
EP1032367A4 (en) * 1997-11-17 2005-02-02 Haemacure Corp Fibrin sealants or adhesives comprising a hyaluronic acid derivative material
EP1032367A2 (en) * 1997-11-17 2000-09-06 Haemacure Corporation Fibrin sealants or adhesives comprising a hyaluronic acid derivative material
US6514534B1 (en) 1998-08-14 2003-02-04 Incept Llc Methods for forming regional tissue adherent barriers and drug delivery systems
US7025990B2 (en) 1998-08-14 2006-04-11 Incept Llc Methods for forming regional tissue adherent barriers and drug delivery systems
US8409605B2 (en) 1998-11-06 2013-04-02 Neomend, Inc. Biocompatible material composition adaptable to diverse therapeutic indications
US8383144B2 (en) 1998-11-06 2013-02-26 Neomend, Inc. Tissue adhering compositions
US6899889B1 (en) * 1998-11-06 2005-05-31 Neomend, Inc. Biocompatible material composition adaptable to diverse therapeutic indications
US8409249B2 (en) 1998-11-06 2013-04-02 Neomend, Inc. Systems, methods, and compositions for achieving closure of suture sites
US7247314B2 (en) 1998-11-06 2007-07-24 Neomend, Inc Biocompatible material composition adaptable to diverse therapeutic indications
JP2001278917A (en) * 2000-03-29 2001-10-10 Toagosei Co Ltd Composition
EP1695724A1 (en) * 2001-05-09 2006-08-30 Baxter International Inc. Fibrin material and method for producing and using the same
US8529960B2 (en) 2002-03-18 2013-09-10 Carnell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
WO2004009207A1 (en) * 2002-07-18 2004-01-29 Hanuman Llc Plasma concentrating apparatus and method
US6905612B2 (en) 2003-03-21 2005-06-14 Hanuman Llc Plasma concentrate apparatus and method
US7741427B2 (en) 2004-06-16 2010-06-22 Straumann Holding Ag Barrier membrane
US8227460B2 (en) 2004-06-16 2012-07-24 Straumann Holding Ag Methylene blue
US8044172B2 (en) 2004-06-16 2011-10-25 Straumann Holding Ag Barrier membrane
US7282584B2 (en) 2004-06-16 2007-10-16 Straumann Holding Ag Methylene blue
US7919112B2 (en) 2004-08-26 2011-04-05 Pathak Holdings, Llc Implantable tissue compositions and method
US10149670B2 (en) 2004-11-05 2018-12-11 Access Closure, Inc. Apparatus and methods for sealing a vascular puncture
US9386969B2 (en) 2004-11-05 2016-07-12 Incept, Llc Methods for sealing a vascular puncture
US9687216B2 (en) 2004-11-05 2017-06-27 Incept, Llc Methods for sealing a vascular puncture
US8986730B2 (en) 2004-11-05 2015-03-24 Incept, Llc Methods for sealing a vascular puncture
US11298444B2 (en) 2005-04-01 2022-04-12 Trivascular, Inc. Non-degradable, low swelling, water soluble radiopaque hydrogel polymer
US9011687B2 (en) 2005-04-27 2015-04-21 Biomet Biologics, Llc Method and apparatus for producing autologous clotting components
US10940231B2 (en) 2006-03-29 2021-03-09 Incept, Llc Superabsorbent, freeze dried hydrogels for medical applications
US8795709B2 (en) 2006-03-29 2014-08-05 Incept Llc Superabsorbent, freeze dried hydrogels for medical applications
US7872068B2 (en) 2006-05-30 2011-01-18 Incept Llc Materials formable in situ within a medical device
US8044137B2 (en) 2006-05-30 2011-10-25 Incept Llc Materials formable in situ within a medical device
US8911789B2 (en) 2006-10-17 2014-12-16 Carnegie Mellon University Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US8293530B2 (en) 2006-10-17 2012-10-23 Carnegie Mellon University Method and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US8529961B2 (en) 2006-10-17 2013-09-10 Carmell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US8529959B2 (en) 2006-10-17 2013-09-10 Carmell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US8529958B2 (en) 2006-10-17 2013-09-10 Carmell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US9364503B2 (en) 2006-10-17 2016-06-14 Carmell Therapeutics Corporation Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
WO2008134774A3 (en) * 2007-04-27 2009-11-26 Biomet Manufacturing Corp. Blood products with binding agent
US20120076750A1 (en) * 2007-04-27 2012-03-29 Hillary Enyart Fibrin based glue with functionalized hydrophilic polymer protein binding agent
US8092837B2 (en) 2007-04-27 2012-01-10 Biomet Manufacturing Corp Fibrin based glue with functionalized hydrophilic polymer protein binding agent
US11324828B2 (en) 2007-07-09 2022-05-10 Incept, Llc Hydrogel polymeric compositions and methods
US9125807B2 (en) 2007-07-09 2015-09-08 Incept Llc Adhesive hydrogels for ophthalmic drug delivery
US10251954B2 (en) 2007-07-09 2019-04-09 Incept, Llc Hydrogel polymeric compositions and methods
US9775906B2 (en) 2007-07-09 2017-10-03 Incept Llc Hydrogel polymeric compositions and methods
US9370485B2 (en) 2007-07-09 2016-06-21 Incept, Llc Hydrogel polymeric compositions and methods
WO2009014776A1 (en) * 2007-07-23 2009-01-29 Carnegie Mellon University Methods and apparatus for manufacturing plasma based plastics and bioplastics produced therefrom
US7862538B2 (en) 2008-02-04 2011-01-04 Incept Llc Surgical delivery system for medical sealant
US10111979B2 (en) 2009-01-27 2018-10-30 Reddress Ltd. Wound dressings, methods and apparatus for making same and storage and use thereof
US9180142B2 (en) 2009-01-27 2015-11-10 Reddress Ltd. Wound dressings, methods and apparatus for making same and storage and use thereof
US8409606B2 (en) 2009-02-12 2013-04-02 Incept, Llc Drug delivery through hydrogel plugs
US9463004B2 (en) 2009-05-04 2016-10-11 Incept, Llc. Biomaterials for track and puncture closure
US11154624B2 (en) 2009-12-15 2021-10-26 Incept, Llc Echolucent implant compositions and methods
US9669117B2 (en) 2009-12-15 2017-06-06 Incept, Llc Implants and biodegradable tissue markers
US8383161B2 (en) 2009-12-15 2013-02-26 Incept, Llc Radioopaque covalently crosslinked hydrogel particle implants
US9308283B2 (en) 2009-12-15 2016-04-12 Incept, Llc Implants and biodegradable fiducial markers
US10272164B2 (en) 2009-12-15 2019-04-30 Incept, Llc Implants and biodegradable tissue markers
US11786612B2 (en) 2009-12-15 2023-10-17 Incept, Llc Implant and biodegradable tissue marker compositions and methods
US10786581B2 (en) 2009-12-15 2020-09-29 Incept, Llc Implants and biodegradable tissue markers
US8852646B2 (en) 2009-12-15 2014-10-07 Incept, Llc Particulate implants and biodegradable fiducial markers
US11083802B2 (en) 2009-12-15 2021-08-10 Incept, Llc Echolucent implant compositions and methods
US11160883B2 (en) 2009-12-15 2021-11-02 Incept, Llc Echolucent implant composition and methods
US8961501B2 (en) 2010-09-17 2015-02-24 Incept, Llc Method for applying flowable hydrogels to a cornea
EP3113722A4 (en) * 2014-03-07 2017-12-06 Endologix, Inc. Forming hydrogels and materials therefor
US11129927B2 (en) 2014-03-07 2021-09-28 Endologix Llc Method for forming hydrogels and materials therefor
US10350331B2 (en) 2014-03-07 2019-07-16 Endologix, Inc. Method for forming hydrogels and materials therefor
US11759553B2 (en) 2014-03-07 2023-09-19 Endologix Llc Method for forming hydrogels and materials therefor
US9550028B2 (en) 2014-05-06 2017-01-24 Biomet Biologics, LLC. Single step desiccating bead-in-syringe concentrating device
WO2016084097A1 (en) * 2014-11-13 2016-06-02 Hemarus Therapeutics Ltd. Process for producing fibrinogen and thrombin
CN114096286A (en) * 2019-07-12 2022-02-25 加特技术公司 Biocompatible flexible hemostatic tablet
CN114096286B (en) * 2019-07-12 2023-06-02 加特技术公司 Biocompatible flexible hemostatic tablet

Also Published As

Publication number Publication date
US20040002456A1 (en) 2004-01-01
US20100069613A1 (en) 2010-03-18
US8557535B2 (en) 2013-10-15
AU4648697A (en) 1998-04-14
US6887974B2 (en) 2005-05-03
US20020114775A1 (en) 2002-08-22
US7605232B2 (en) 2009-10-20
US7057019B2 (en) 2006-06-06
US7211651B2 (en) 2007-05-01
US20030077272A1 (en) 2003-04-24
US20070197776A1 (en) 2007-08-23

Similar Documents

Publication Publication Date Title
US7211651B2 (en) Proteinaceous gels having visualization agents and methods of use thereof
EP2023941B1 (en) Protein crosslinkers, crosslinking methods and applications thereof
US6537569B2 (en) Radiation cross-linked hydrogels
KR102143252B1 (en) Hemostatic composition
EP2822533B1 (en) Biomaterials for delivery of blood extracts and methods of using same
US20020187182A1 (en) Biocompatible fleece for hemostasis and tissue engineering
US20060127873A1 (en) Composition for cytocompatible, injectable, self-gelling chitosan solutions for encapsulating and delivering live cells or biologically active factors
CN106178085A (en) Rapidly acting dry sealant and use and preparation method
WO2014035725A1 (en) Blood plasma hydrogels for tissue regeneration and wound healing applications
JP6877360B2 (en) Hemostatic composition
EP3903808A1 (en) Hemostatic composition and receptacle comprising same
CN1138483A (en) Fibre proteinogen compound and its prepn. method
RU2462255C1 (en) Organ-specific regenerant gi
CN116870245A (en) Preparation method and application of biodegradable polymer nano hemostatic dressing
JP2002220344A (en) Fibrinogen preparation with excellent solubility
AU2002253939A1 (en) Radiation cross-linked hydrogels

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH KE LS MW SD SZ UG ZW AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 1998514977

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 09147897

Country of ref document: US

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA