CA2745860A1 - Surgical compositions - Google Patents

Surgical compositions Download PDF

Info

Publication number
CA2745860A1
CA2745860A1 CA2745860A CA2745860A CA2745860A1 CA 2745860 A1 CA2745860 A1 CA 2745860A1 CA 2745860 A CA2745860 A CA 2745860A CA 2745860 A CA2745860 A CA 2745860A CA 2745860 A1 CA2745860 A1 CA 2745860A1
Authority
CA
Canada
Prior art keywords
cells
solution
hydrogel
tissue
natural component
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2745860A
Other languages
French (fr)
Inventor
Saumya Banerjee
Joshua Stopek
Timothy Sargeant
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Covidien LP
Original Assignee
Tyco Healthcare Group LP
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tyco Healthcare Group LP filed Critical Tyco Healthcare Group LP
Publication of CA2745860A1 publication Critical patent/CA2745860A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/52Hydrogels or hydrocolloids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/18Macromolecular materials obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0012Cell encapsulation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/64Animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/30Synthetic polymers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • C12N2533/80Hyaluronan

Abstract

The present disclosure relates to methods of cell encapsulation using multi-component hydrogels. The hydrogels may include a natural component having nucleophilic functional groups as well as an electrophilic component. In embodiments, at least one of the components may be branched, having drugs, antibodies, enzymes, and the like incorporated therein, which may react with at least one of the other components of the hydrogel.

Description

SURGICAL COMPOSITIONS
[0001]

BACKGROUND
[0002] The present disclosure relates to hydrogels. Specifically, the disclosure relates to multi-component hydrogels which may include bioactive agents therein.
[0003] Biocompatible cross-linked polymers having electrophilic and nucleophilic functional groups are known for their use in the formation of hydrogels. These hydrogels may be used in a variety of surgical applications. Hydrogels may be utilized, for example, as a sealant for wounds, as a glue for adhering surgical implants to tissue, as drug delivery devices, as coatings, combinations thereof, and the like.
[0004] Biocompatible polymers, which may be used as tissue scaffolds capable of providing cells with growth and development components, remain desirable.
SUMMARY
[0005] The present disclosure includes a method of cell encapsulation comprising creating a first solution including suspending cells at a desired concentration in a first buffer salt solution, creating a second solution including dissolving a multi-arm polyethylene glycol at a desired concentration in a second buffer salt solution, mixing the first and second solution at about a 1:1 ratio to create a third solution, co-injecting the third solution with a natural component selected from the group consisting of collagen, serum, gelatin, hyaluronic acid, and combinations thereof.

DETAILED DESCRIPTION
[0006] The present disclosure provides hydrogels which include an electrophilic precursor, sometimes referred to herein as an electrophilic crosslinker, and a nucleophilic component. In embodiments, the nucleophilic component is a natural component, which may be cross-linked by the electrophilic cross-linker to form a hydrogel. In embodiments, the hydrogel may be biodegradable. The term "biodegradable" as used herein is defined to include both bioabsorbable and bioresorbable materials.
By biodegradable, it is meant that the materials decompose, or lose structural integrity under body conditions (e.g. enzymatic degradation or hydrolysis) or are broken down (physically or chemically) under physiologic conditions in the body such that the degradation products are excretable or absorbable by the body.
[0007] The hydrogel may be formed in situ and may be utilized as a drug delivery device or tissue scaffold, combinations thereof, and the like. In embodiments, the hydrogel may deliver and/or release biological factors/molecules and/or cells at the site of implantation.
Thus, where the hydrogel of the present disclosure is utilized as a tissue scaffold, it may assist in native tissue regrowth by providing the surrounding tissue with needed nutrients and bioactive agents. In some embodiments, as discussed further below, the hydrogel itself may include a natural component such as collagen, gelatin, hyaluronic acid, combinations thereof, and the like, and thus the natural component may be released at the site of implantation as the hydrogel degrades. The term "natural component" as used herein includes polymers, compositions of matter, materials, combinations thereof, and the like, which can be found in nature or derived from compositions/organisms found in nature. Natural components also may include compositions which are found in nature but can be synthesized by man, for example, using methods to create natural/synthetic/biologic recombinant materials, as well as methods capable of producing proteins with the same sequences as those found in nature, and/or methods capable of producing materials with the same structure and components as natural materials, such as synthetic hyaluronic acid, which is commercially available, for example, from Sigma Aldrich.

[00081 The components for forming hydrogels on or in tissues include, for example, in situ forming materials. The in situ forming material may include a single precursor or multiple precursors that form "in situ", meaning formation occurs at a tissue in a living animal or human body. In general, this may be accomplished by having a precursor that can be activated at the time of application to create, in embodiments, a hydrogel.

Activation can be through a variety of methods including, but not limited to, environmental changes such as pH, ionicity, temperature, etc. In other embodiments, the components for forming a hydrogel may be contacted outside the body and then introduced into a patient as an implant such as a (pre-formed) tissue scaffold.

100091 As noted above, in situ forming materials may be made from one or more precursors. The precursor may be, e.g., a monomer or a macromer. One type of precursor may have a functional group that is an electrophile or nucleophile.
Electrophiles react with nucleophiles to form covalent bonds. Covalent crosslinks or bonds refer to chemical groups formed by reaction of functional groups on different polymers that serve to covalently bind the different polymers to each other.
In certain embodiments, a first set of electrophilic functional groups on a first precursor may react with a second set of nucleophilic functional groups on a second precursor.
When the precursors are mixed in an environment that permits reaction (e.g., as relating to pH or solvent), the functional groups react with each other to form covalent bonds.
The precursors become crosslinked when at least some of the precursors can react with more than one other precursor. For instance, a precursor with two functional groups of a first type may be reacted with a crosslinking precursor that has at least three functional groups of a second type capable of reacting with the first type of functional groups.

[0010] In embodiments the hydrogel may be formed from a single precursor or multiple precursors. For example, where the hydrogel is formed from multiple precursors, for example two precursors, the precursors may be referred to as a first and second hydrogel precursor. The terms "first hydrogel precursor" and "second hydrogel precursor" each mean a polymer, functional polymer, macromolecule, small molecule, or crosslinker that can take part in a reaction to form a network of crosslinked molecules, e.g., a hydrogel.
The term "functional group" as used herein refers to electrophilic or nucleophilic groups capable of reacting with each other to form a bond. Electrophilic functional groups include, for example, N-hydroxysuccinimides ("NHS"), sulfosuccinimides, carbonyldiimidazole, sulfonyl chloride, aryl halides, sulfosuccinimidyl esters, N-hydroxysuccinimidyl esters, succinimidyl esters such as succinimidyl succinates and/or succinimidyl propionates, isocyanates, thiocyanates, carbodiimides, benzotriazole carbonates, epoxides, aldehydes, maleimides, imidoesters, combinations thereof, and the like. In embodiments, the electrophilic functional group is a succinimidyl ester.

[0011] The electrophilic hydrogel precursors may have biologically inert and water soluble cores, as well as non-water soluble cores. When the core is a polymeric region that is water soluble, suitable polymers that may be used include: polyethers, for example. polyalkylene oxides such as polyethylene glycol("PEG"), polyethylene oxide ("PEO"), polyethylene oxide-co-polypropylene oxide ("PPO"), co-polyethylene oxide block or random copolymers, and polyvinyl alcohol ("PVA"); poly(vinyl pyrrolidinone) ("PVP"); poly(amino acids); poly (saccharides), such as dextran, chitosan, alginates, carboxymethylcellulose, oxidized cellulose, hydroxyethylcellulose, hydroxymethylcellulose; hyaluronic acid; and proteins such as albumin, collagen, casein, and gelatin. In embodiments, combinations of the foregoing polymers may be utilized.
[0012] The polyethers, and more particularly poly(oxyalkylenes) or poly(ethylene glycol) or polyethylene glycol, may be utilized in some embodiments. When the core is small in molecular nature, any of a variety of hydrophilic functionalities can be used to make the first and second hydrogel precursors water soluble. For example. functional groups like hydroxyl, amine, sulfonate and carboxylate, which are water soluble, may be used to make the precursor water soluble. For example, the n-hydroxysuccinimide ("NHS") ester of subaric acid is insoluble in water, but by adding a sulfonate group to the succinimide ring, the NHS ester of subaric acid may be made water soluble, without affecting its reactivity towards amine groups.

[0013] Each of the first and second hydrogel precursors may be multifunctional, meaning that it may include two or more electrophilic or nucleophilic functional groups, such that, for example, a nucleophilic functional group on the first hydrogel precursor may react with an electrophilic functional group on the second hydrogel precursor to form a covalent bond. At least one of the first or second hydrogel precursors includes more than two functional groups, so that, as a result of electrophilic-nucleophilic reactions, the precursors combine to form cross-linked polymeric products.

[0014] The macromolecule having the electrophilic functional group may be multi-armed. For example, the macromolecule may be a multi-armed PEG having four, six, eight, or more arms extending from a core. The core may be the same or different from the macromolecule forming the arms. For example, the core may be PEG and the multiple arms may also be PEG. In embodiments, the core may be a natural polymer.
[0015] The molecular weight (MW) of the electrophilic crosslinker may be from about 2,000 to about 100,000; in embodiments from about 10,000 to about 40,000.
Multi-arm precursors may have a molecular weight that varies depending on the number of arms.
For example, an arm having a 1000 MW of PEG has enough CH2CH2O groups to total at least 1000 MW. The combined molecular weight of an individual arm may be from about 250 to about 5,000; in embodiments from about 1,000 to about 3,000; in embodiments from about 1,250 to about 2,500. In embodiments, the electrophilic crosslinker may be a multi-arm PEG functionalized with multiple NHS groups having, for example, four, six or eight arms and a molecular weight from about 5.000 to about 25,000. Other examples of suitable precursors are described in U.S. Patent Nos.
6,152,943; 6.165,201; 6,179,862; 6,514.534; 6,566,406; 6,605,294; 6,673,093;
6,703.047;

6,818,018; 7,009,034; and 7,347,850, the entire disclosures of each of which are incorporated herein by reference.

[0016] The electrophilic precursor may be a cross-linker that provides an electrophilic functional group capable of bonding with nucleophiles on another component, in embodiments a natural component. The natural component may be endogenous to the patient to which the electrophilic crosslinker is applied, or may be exogenously applied.
[0017] In embodiments, one of the precursors may be a natural component possessing nucleophilic groups. Nucleophilic groups which may be present include, for example, NH2, -SH, -OH, -PH2, and -CO-NH-NH2.

[0018] The natural component may be, for example, collagen, gelatin, blood (including serum, which may be whole serum or extracts therefrom), hyaluronic acid, proteins, albumin, other serum proteins, serum concentrates, platelet rich plasma (prp), combinations thereof, and the like. Additional suitable natural components which may be utilized or added to another natural component, sometimes referred to herein as a bioactive agent, include, for example, stem cells, DNA. RNA, enzymes, growth factors, peptides, polypeptides, antibodies, other nitrogenous natural molecules, combinations thereof, and the like. Other natural components may include derivatives of the foregoing, for example modified hyaluronic acid, dextran. other polysaccharide, polymers and/or polypeptides, including aminated polysaccharides which may be naturally derived, synthetic, or biologically derived. For example, in embodiments hyaluronic acid may be modified to make it nucleophilic.

[0019] In embodiments, any of the above natural components may be synthetically prepared, e.g., synthetic hyaluronic acid, utilizing methods within the purview of those skilled in the art. Similarly, in embodiments the natural component could be a natural or synthetic long chain aminated polymer. The natural component may also be modified, i.e., aminated to create a nucleophilic polymer.

[0020] The natural component may provide cellular building blocks or cellular nutrients to the tissue that it contacts in situ. For example, serum contains proteins, glucose, clotting factors, mineral ions, and hormones which may be useful in the formation or regeneration of tissue.

[0021] In embodiments, the natural component includes whole serum. In embodiments, the natural component is autologous, i.e., collagen, serum, blood, and the like, from the body where the hydrogel is (or is to be) formed. In this manner, the person or animal in which the hydrogel is to be used may provide the natural component for use in formation of the hydrogel. In such embodiments, the resulting hydrogel is semi-autologous, including a synthetic electrophilic precursor and an autologous/endogenous nucleophilic precursor.

[0022] In embodiments, a multifunctional nucleophilic polymer, such as a natural component having multiple amine groups, may be used as a first hydrogel precursor and a multifunctional electrophilic polymer, such as a multi-arm PEG
functionalized with multiple NHS groups, may be used as a second hydrogel precursor. In embodiments, the precursors may be in solution(s), which may be combined to permit formation of the hydrogel. Any solutions utilized as part of the in situ forming material system should not contain harmful or toxic solvents. In embodiments, the precursor(s) may be substantially soluble in a solvent such as water to allow application in a physiologically-compatible solution, such as buffered isotonic saline.
8 [0023] In embodiments, a hydrogel may be formed from a combination of collagen and gelatin as the natural component, with a multi-functional PEG utilized as a crosslinker.
In embodiments, the collagen or gelatin may be placed in solution, utilizing a suitable solvent. Such a buffer may have a pH from about 8 to about 12, in embodiments from about 8.2 to about 9. Examples of such buffers include, but are not limited to, borate buffers, and the like.

[0024] In a second solution, an electrophilic crosslinker, in embodiments a multi-arm PEG functionalized with electrophilic groups such as n-hydroxysuccinimide, may be prepared in a buffer such as Hanks Balanced Salt Solution, Dulbecco's Modified Eagle's Medium, Phosphate Buffered Saline, water, phosphate buffer, combinations thereof, and the like. The electrophilic crosslinker, in embodiments a multi-arm PEG
functionalized with n-hydroxysuccinimide groups, may be present in a solution including the above buffer at a concentration from about 0.02 grams/ml to about 0.5 grams/ml, in embodiments from about 0.05 grams/ml to about 0.3 grams/ml.

[0025] The two components may be introduced in situ, wherein the electrophilic groups on the multi-arm PEG crosslink the amine nucleophilic components of the collagen and/or Gelatin. The ratio of natural component to electrophilic component (i.e., collagen:PEG) may be from about 0.1:1 to about 100:1, in embodiments from about 1:1 to about 10:1.

[0026] The natural components; e.g., collagen, gelatin, and/or hyaluronic acid, may together be present at a concentration of at least about 1.5 percent by weight of the hydrogel, in embodiments from about 1.5 percent by weight to about 20 percent by weight of the hydrogel. in other embodiments from about 2 percent by weight to about 10
9 percent by weight of the hydrogel. In certain embodiments, collagen may be present from about 0.5 percent to about 7 percent by weight of the hydrogel, in further embodiments, from about 1 percent to about 4 percent by weight of the hydrogel. In another embodiment, gelatin may be present from about 1 percent to about 20 percent by weight of the hydrogel, in further embodiments, from about 2 percent to about
10 percent by weight of the hydrogel. In yet another embodiment, hyaluronic acid and collagen combined as the natural component(s) may be present from about 0.5 percent to about 8 percent by weight of the hydrogel, in further embodiments, from about 1 percent to about percent by weight of the hydrogel. It is also envisioned that the hyaluronic acid may not be present as a "structural" component, but as more of a bioactive agent. For example, hyaluronic acid may be present in solution/gel in concentrations as low as 0.001 percent by weight of the solution/gel and have biologic activity.

[0027] The electrophilic crosslinker may be present in amounts of from about 0.5 percent by weight to about 20 percent by weight of the hydrogel, in embodiments from about 1.5 percent by weight to about 15 percent by weight of the hydrogel.

[0028] In situ forming materials may be formed either through covalent, ionic or hydrophobic bonds. Physical (non-covalent) crosslinks may result from complexation, hydrogen bonding, desolvation, Van der Waals interactions, ionic bonding.
combinations thereof, and the like, and may be initiated by mixing two precursors that are physically separated until combined in situ, or as a consequence of a prevalent condition in the physiological environment, including: temperature, pH, ionic strength, combinations thereof and the like. Chemical (covalent) crosslinking may be accomplished by any of a number of mechanisms, including: free radical polymerization, condensation polymerization, anionic or cationic polymerization, step growth polymerization, electrophile-nucleophile reactions, combinations thereof, and the like.

[0029] In some embodiments, in situ forming material systems may include biocompatible multi-precursor systems that spontaneously crosslink when the precursors are mixed, but wherein the two or more precursors are individually stable for the duration of the deposition process. In other embodiments, in situ forming materials may include a single precursor that crosslinks with endogenous materials and/or tissues.

[0030] The crosslinking density of the resulting biocompatible crosslinked polymer may be controlled by the overall molecular weight of the crosslinker and natural component and the number of functional groups available per molecule. A lower molecular weight between crosslinks, such as 600 daltons (Da), will give much higher crosslinking density as compared to a higher molecular weight, such as 10,000 Da. Elastic gels may be obtained with higher molecular weight natural components with molecular weights of more than 3000 Da.

[0031] The crosslinking density may also be controlled by the overall percent solids of the crosslinker and natural component solutions. Increasing the percent solids increases the probability that an electrophilic group will combine with a nucleophilic group prior to inactivation by hydrolysis. Yet another method to control crosslink density is by adjusting the stoichiometry of nucleophilic groups to electrophilic groups. A
one to one ratio may lead to the highest crosslink density, however, other ratios of reactive functional groups (e.g., electrophile:nucleophile) are envisioned to suit a desired formulation.

1l [0032] The natural component containing a nucleophilic functional group and the electrophilic functional precursor react to form a hydrogel. The reaction may occur in situ or outside of the body for future implantation therein or thereon. In embodiments, a hydrogel of the present disclosure may be produced by combining precursors that crosslink quickly after application to a surface, e.g., on a tissue of a patient, to form an in situ forming material.

[0033] The hydrogel thus produced may be bioabsorbable, so that it does not have to be retrieved from the body. Absorbable materials are absorbed by biological tissues and disappear in vivo at the end of a given period, which can vary, for example, from one day to several months, depending on the chemical nature of the material.
Absorbable materials include both natural and synthetic biodegradable polymers, as well as bioerodible polymers.

[0034] In embodiments, one or more precursors having biodegradable linkages present in between functional groups may be included to make the hydrogel biodegradable or absorbable. In some embodiments, these linkages may be, for example, esters, which may be hydrolytically degraded in physiological solution. The use of such linkages is in contrast to protein linkages that may be degraded by proteolytic action. A
biodegradable linkage optionally also may form part of a water soluble core of one or more of the precursors. Alternatively, or in addition, functional groups of precursors may be chosen such that the product of the reaction between them results in a biodegradable linkage.

For each approach, biodegradable linkages may be chosen such that the resulting biodegradable biocompatible crosslinked polymer degrades or is absorbed in a desired period of time. Generally, biodegradable linkages may be selected that degrade the hydrogel under physiological conditions into non-toxic or low toxicity products.
[00351 The gels of the present disclosure may degrade due to hydrolysis or enzymatic degradation of the biodegradable region, whether part of the natural component or introduced into a synthetic electrophilic crosslinker. The degradation of gels containing synthetic peptide sequences will depend on the specific enzyme and its concentration. In some cases, a specific enzyme may be added during the crosslinking reaction to accelerate the degradation process. In the absence of any degradable enzymes, the crosslinked polymer may degrade solely by hydrolysis of the biodegradable segment. In embodiments in which polyglycolate is used as the biodegradable segment, the crosslinked polymer may degrade in from about 1 day to about 30 days depending on the crosslinking density of the network. Similarly, in embodiments in which a polycaprolactone based crosslinked network is used, degradation may occur over a period of time from about I month to about 8 months. The degradation time generally varies according to the type of degradable segment used, in the following order:
polyglycolate<polylactate<polytrimethylene carbonate<polycaprolactone. Thus, it is possible to construct a hydrogel with a desired degradation profile, from a few days to months, using a proper degradable segment.

[00361 Where utilized, the hydrophobicity generated by biodegradable blocks such as oligohydroxy acid blocks or the hydrophobicity of PPO blocks in PLURONICTM or TETRONICTM polymers utilized to form the electrophilic crosslinker may be helpful in dissolving small organic drug molecules. Other properties which will be affected by incorporation of biodegradable or hydrophobic blocks include: water absorption, mechanical properties and thermosensitivity.

[0037] Properties of the in situ forming material system may be selected according to the intended application. For example, if the in situ forming material is to be used to temporarily occlude a reproductive organ, such as a fallopian tube, it may be desirable that the in situ forming material system undergo significant swelling and be biodegradable. Alternatively, the in situ forming material may have thrombotic properties, or its precursors may react with blood or other body fluids to form a coagulum. In embodiments, the precursors react with blood or body fluids, which may be endogenous or exogenously applied, thereby forming a tissue scaffold.

[0038] Other applications may require different characteristics of the in situ forming material system. Generally, the materials should be selected on the basis of exhibited biocompatibility and lack of toxicity.

[0039] Certain properties of the in situ forming material can be useful, including adhesion to a variety of tissues, desirable setting times to enable a surgeon to accurately and conveniently place the in situ forming materials, high water content for biocompatibility, which may be relevant for hydrogels, mechanical strength for use in sealants, and/or toughness to resist destruction after placement. Synthetic materials that are readily sterilized and avoid the dangers of disease transmission involved in the use of natural materials may thus be used. Indeed, certain in situ polymerizable hydrogels made using synthetic precursors are within the purview of those skilled in the art, e.g., as used in commercially available products such as FOCAL SEAL' (Genzyme, Inc.), COSEAL
"
(Angiotech Pharmaceuticals), and DURASEAL (Confluent Surgical, Inc). Other known hydrogels include, for example, those disclosed in U.S. Patent Nos. 6,656,200;
5,874,500;
5,543,441; 5,514,379; 5,410,016; 5,162,430; 5,324,775; 5,752,974; and 5,550,187.

[0040] As noted above, in embodiments a branched multi-arm PEG, sometimes referred to herein as a PEG star, may be included to form a hydrogel of the present disclosure. A
PEG star may be functionalized so that its arms include biofunctional groups such as amino acids, peptides, antibodies, enzymes, drugs, combinations thereof, or other moieties such as bioactive agents in its cores, its arms, or at the ends of its arms. The biofunctional groups may also be incorporated into the backbone of the PEG, or attached to a reactive group contained within the PEG backbone. The binding can be covalent or non-covalent, including electrostatic, thiol mediated, peptide mediated, or using known reactive chemistries, for example, biotin with avidin.

[0041] Amino acids incorporated into a PEG star may be natural or synthetic, and can be used singly or as part of a peptide. Sequences may be utilized for cellular adhesion, cell differentiation, combinations thereof, and the like, and may be useful for binding other biological molecules such as growth factors, drugs, cytokines, DNA, antibodies, enzymes, combinations thereof, and the like. Such amino acids may be released upon enzymatic degradation of the PEG star.

[0042] These PEG stars may also include functional groups as described above to permit their incorporation into a hydrogel. The PEG star may be utilized as the electrophilic crosslinker or, in embodiments, be utilized as a separate component in addition to the electrophilic crosslinker described above. In embodiments, the PEG stars may include electrophilic groups that bind to nucleophilic groups. As noted above, the nucleophilic groups may be part of a natural component utilized to form a hydrogel of the present disclosure.

[0043] In some embodiments a biofunctional group may be included in a PEG star by way of a degradable linkage, including an ester linkages formed by the reaction of PEG
carboxylic acids or activated PEG carboxylic acids with alcohol groups on a biofunctional group. In this case, the ester groups may hydrolyze under physiological conditions to release the biofunctional group.

[0044] In embodiments an in situ forming material may also include an initiator. An initiator may be any precursor or group capable of initiating a polymerization reaction for the formation of the in situ forming material.

In situ Polymerization [0045] The precursor(s) may be dissolved to form a solution prior to use. with the solution being delivered to the patient. As used herein, a solution may be homogeneous, heterogeneous, phase separated, or the like. In other embodiments, the precursor(s) may be in an emulsion. Solutions suitable for use in accordance with the present disclosure include those that may be used to form implants in lumens or voids. Where two solutions are employed, each solution may contain one precursor of an in situ forming material which forms upon contact. The solutions may be separately stored and mixed when delivered to tissue.

[0046] Electrophilic crosslinkers as described above may be reacted with natural components to generate crosslinked polymeric networks. Generally, aqueous solutions of crosslinkers may be mixed with natural components to produce a crosslinked hydrogel.

The reaction conditions for forming a crosslinked polymeric hydrogel will depend on the nature of the functional groups. In embodiments, reactions are conducted in buffered aqueous solutions at a pH of about 5 to about 12. Buffers include, for example, phosphate buffered saline (PBS), Hanks Balanced Salt Solution, Dulbecco's Modified Eagle's Medium, water, phosphate buffer, sodium borate buffer, triethanol amine buffer, combinations thereof and the like. In some embodiments, organic solvents such as ethanol or isopropanol may be added to improve the reaction speed or to adjust the viscosity of a given formulation.

[0047) When the electrophilic crosslinker is synthetic (for example, based on polyalkylene oxide), it may be desirable to use molar equivalent quantities of the reactants. In some cases, molar excess of an electrophilic crosslinker may be added to compensate for side reactions such as reactions due to hydrolysis of the functional group.
[0048] When choosing the electrophilic crosslinker and natural component, at least one of the precursors may have more than two functional groups per molecule and at least one degradable region, if it is desired that the resultant biocompatible crosslinked polymer be biodegradable.

[0049] Formulations may be prepared that are suited to make precursor crosslinking reactions occur in situ. In general. this may be accomplished by having a precursor that can be activated at the time of application to a tissue to form a crosslinked hydrogel.
Activation can be made before, during, or after application of the precursor to the tissue, provided that the precursor is allowed to conform to the tissue's shape before crosslinking and associated gelation is otherwise too far advanced. Activation includes, for instance, mixing precursors with functional groups that react with each other. Thus, in situ polymerization includes activation of chemical moieties to form covalent bonds to create an insoluble material, e.g., a hydrogel, at a location where the material is to be placed on, within, or both on and within, a patient. In situ polymerizable polymers may be prepared from precursor(s) that can be reacted such that they form a polymer within the patient. Thus precursor(s) with electrophilic functional groups can be mixed or otherwise activated in the presence of precursors with nucleophilic functional groups.
[00501 With respect to coating a tissue, and without limiting the present disclosure to a particular theory of operation, it is believed that reactive precursor species that crosslink quickly after contacting a tissue surface may form a three dimensional structure that is mechanically interlocked with the coated tissue. This interlocking contributes to adherence, intimate contact, and continuous coverage of the coated region of the tissue.
In other embodiments, where electrophilic precursors are used, such precursors may react with free amines in tissue, thereby serving as a means for attaching the hydrogel to tissue.
[00511 The crosslinking reaction leading to gelation can occur, in some embodiments within a time from about 1 second to about 5 minutes. in embodiments from about 3 seconds to about 1 minute; persons of ordinary skill in these arts will immediately appreciate that all ranges and values within these explicitly stated ranges are contemplated. For example, in embodiments, the in situ gelation time of hydrogels according to the present disclosure is less than about 20 seconds, and in some embodiments, less than about 10 seconds, and in yet other embodiments less than about 5 seconds.

Administration [0052] As noted above, the electrophilic crosslinker may be introduced by itself, whereby it crosslinks endogenous natural components. Alternatively, natural components may be removed from a patient or obtained from other sources, and then introduced along with the electrophilic crosslinker to form a hydrogel of the present disclosure, such as, for example, plasma. The precursor(s) may be placed into solution prior to use, with the solution being delivered to the patient. One may use a syringe for delivery of a single precursor, i.e., an electrophilic crosslinker, or a dual syringe or similar device to apply more than one precursor solutions, such as those described in U.S. Patent Nos.
4,874,368;
4,631,055; 4,735,616; 4,359,049; 4,978,336; 5,116,315; 4,902,281; 4,932,942;
6,179,862;
6,673,093; and 6,152,943.

[0053] In other embodiments, the precursors may be combined to create a film or foam, which then may be inplanted into the patient. Methods for creating films and foams are within the purview those skilled in the art, which include but are not limited to spraying, film casting, lyophilization techniques, etc.

Uses [0054] As noted above, the hydrogels of the present disclosure may be utilized as tissue adhesives or sealants. The hydrogel may also be used to correct a tissue defect. As used herein, a "tissue defect" may include any breakdown of tissue from a normal, healthy state. This breakdown may be due to internal factors such as degenerative disease, or external factors such as injury. Any variation from the normal structure of a tissue may be a "tissue defect." In some embodiments, where utilized to correct a tissue defect, a hydrogel of the present disclosure may be referred to as a tissue scaffold.

[0055] The hydrogel compositions may be utilized for cartilage repair, tendon repair, ligament repair, treatment of osteoarthritis, bone fillers, to repair soft tissue defects, as a filler for soft/hard tissue interfaces, and/or for general organ or tissue repair. General organ or tissue repair may include, for example, gastrointestinal, thoracic, cardiac. neural.
or other organ repair.

[0056] In embodiments, where utilized for cartilage repair, a method for using a hydrogel of the present disclosure may include identifying a cartilage defect and then cleaning out the cartilage defect. Once the defect has been cleaned, a hydrogel composition of the present disclosure may be introduced into the defect. As noted above, in embodiments, at least one hydrogel precursor may be introduced therein. In embodiments, one of the precursors, sometimes referred to as a first hydrogel precursor, may be a natural component possessing nucleophilic groups. As noted above, the natural component may be endogenous or exogenously applied. A second precursor possessing electrophilic functional groups may also be applied to the defect utilizing any method or apparatus described above or as within the purview of those skilled in the art. The first hydrogel precursor and the second hydrogel precursor may then be allowed to react to form a hydrogel, which may act as a tissue scaffold at the site of the cartilage defect.

[0057] The hydrogel may be porous or smooth. In certain embodiments, the hydrogel may be porous. The term "porous" as used herein means that the hydrogel may possess defined openings and/or spaces which are present as a surface characteristic or a bulk material property, partially or completely penetrating the hydrogel. Pores may be created using methods within the purview of those skilled in the art including, but not limited to, processes such as sintering, leaching of salt, sugar or starch crystals.
Porous materials may have an open-cell structure, where the pores are connected to each other, forming an interconnected network. Conversely, hydrogel may be closed cell, where the pores are not interconnected.

[0058] The hydrogel may be coated with or include additional bioactive agents.
The term "bioactive agent", as used herein, is used in its broadest sense and includes any substance or mixture of substances that have clinical use. Consequently, bioactive agents may or may not have pharmacological activity per se, e.g., a dye.

[0059] Alternatively a bioactive agent could be any agent which provides a therapeutic or prophylactic effect, a compound that affects or participates in tissue growth, cell growth, cell differentiation, an anti-adhesive compound, a compound that may be able to invoke a biological action such as an immune response, or could play any other role in one or more biological processes. It is envisioned that the bioactive agent may be applied to the hydrogel in any suitable form of matter, e.g., films, powders, liquids, gels and the like.

[0060] As noted above, in embodiments that include a multi-arm PEG or PEG
star, the bioactive agent may be incorporated into the core of the PEG, the arms of the PEG, or combinations thereof. In embodiments, the bioactive agent may be attached to a reactive group in the PEG chain. The bioactive agent may be bound covalently, non-covalently, i.e.. electrostatically, through a thiol-mediated or peptide-mediated bond, or using biotin-avidin chemistries and the like.

[0061] Examples of classes of bioactive agents, which may be utilized in accordance with the present disclosure include, for example, anti-adhesives, antimicrobials, analgesics, antipyretics, anesthetics, antiepileptics, antihistamines, anti-inflammatories, cardiovascular drugs, diagnostic agents, sympathomimetics, cholinomimetics, antimuscarinics, antispasmodics, hormones, growth factors, muscle relaxants, adrenergic neuron blockers, antineoplastics, immunogenic agents, immunosuppressants, gastrointestinal drugs, diuretics, steroids, lipids, lipopolysaccharides, polysaccharides, platelet activating drugs, clotting factors and enzymes. It is also intended that combinations of bioactive agents may be used.

[0062] Anti-adhesive agents can be used to prevent adhesions from forming between the hydrogel and the surrounding tissues opposite the target tissue. In addition, anti-adhesive agents may be used to prevent adhesions from forming between the coated implantable medical device and the packaging material. Some examples of these agents include, but are not limited to hydrophilic polymers such as poly(vinyl pyrrolidone), carboxymethyl cellulose, hyaluronic acid, polyethylene oxide, poly vinyl alcohols, and combinations thereof.

[0063] Suitable antimicrobial agents which may be included as a bioactive agent include, for example, triclosan, also known as 2.4,4'-trichloro-2'-hydroxydiphenyl ether, chlorhexidine and its salts, including chlorhexidine acetate, chlorhexidine gluconate, chlorhexidine hydrochloride, and chlorhexidine sulfate, silver and its salts, including silver acetate, silver benzoate, silver carbonate, silver citrate, silver iodate, silver iodide, silver lactate, silver laurate, silver nitrate, silver oxide, silver palmitate, silver protein, and silver sulfadiazine, polymyxin,. tetracycline, aminoglycosides, such as tobramycin and gentamicin, rifampicin, bacitracin, neomycin, chloramphenicol, miconazole.
quinolones such as oxolinic acid, norfloxacin, nalidixic acid. pefloxacin, enoxacin and ciprofloxacin, penicillins such as oxacillin and pipracil, nonoxynol 9, fusidic acid, cephalosporins, and combinations thereof. In addition, antimicrobial proteins and peptides such as bovine lactoferrin and lactoferricin B may be included as a bioactive agent.

[0064] Other bioactive agents, which may be included as a bioactive agent include: local anesthetics; non-steroidal antifertility agents; parasympathomimetic agents;
psychotherapeutic agents; tranquilizers; decongestants; sedative hypnotics;
steroids;
sulfonamides; sympathomimetic agents; vaccines; vitamins; antimalarials; anti-migraine agents; anti-parkinson agents such as L-dopa; anti-spasmodics; anticholinergic agents (e.g., oxybutynin); antitussives; bronchodilators; cardiovascular agents, such as coronary vasodilators and nitroglycerin; alkaloids; analgesics; narcotics such as codeine, dihydrocodeinone, meperidine, morphine and the like; non-narcotics, such as salicylates, aspirin, acetaminophen, d-propoxyphene and the like; opioid receptor antagonists, such as naltrexone and naloxone; anti-cancer agents; anti-convulsants; anti-emetics;
antihistamines; anti-inflammatory agents, such as hormonal agents, hydrocortisone, prednisolone, prednisone, non-hormonal agents, allopurinol, indomethacin, phenylbutazone and the like; prostaglandins and cytotoxic drugs;
chemotherapeutics, estrogens; antibacterials; antibiotics; anti-fungals; anti-virals;
anticoagulants;
anticonvulsants; antidepressants; antihistamines; and immunological agents.

[0065] Other examples of suitable bioactive agents, which may be included in the hydrogel include, for example, viruses and cells, including stem cells;
peptides, polypeptides and proteins, as well as analogs, muteins, and active fragments thereof;

immunoglobulins; antibodies; cytokines (e.g., lymphokines, monokines, chemokines);
blood clotting factors; hemopoietic factors; interleukins (IL-2, IL-3, IL-4, IL-6);
interferons ((3-IFN, a-IFN and y-IFN); erythropoietin; nucleases; tumor necrosis factor;
colony stimulating factors (e.g., GCSF, GM-CSF, MCSF); insulin; anti-tumor agents and tumor suppressors; blood proteins such as fibrin, thrombin, fibrinogen, synthetic thrombin, synthetic fibrin, synthetic fibrinogen; gonadotropins (e.g., FSH, LH, CG, etc.);
hormones and hormone analogs (e.g., growth hormone); vaccines (e.g., tumoral, bacterial and viral antigens); somatostatin; antigens; blood coagulation factors; growth factors (e.g., nerve growth factor, insulin-like growth factor); bone morphogenic proteins; TGF-B; protein inhibitors; protein antagonists; protein agonists; nucleic acids, such as antisense molecules, DNA, RNA, RNAi; oligonucleotides; polynucleotides; and ribozymes.

[0066] The precursors and/or the hydrogel formed from the precursors may contain visualization agents to improve their visibility during surgical procedures.
Visualization agents may be selected from a variety of non-toxic colored substances, such as dyes, suitable for use in implantable medical devices. Suitable dyes are within the purview of those skilled in the art and may include, for example, a dye for visualizing a thickness of the hydrogel as it is formed in situ, e.g., as described in U.S. Patent No.
7,009,034. In some embodiments, a suitable dye may include, for example, FD&C Blue #1, FD&C
Blue #2, FD&C Blue #3, FD&C Blue #6, D&C Green #6, methylene blue, indocyanine green, other colored dyes, and combinations thereof. It is envisioned that additional visualization agents may be used such as fluorescent compounds (e.g., flurescein or eosin), x-ray contrast agents (e.g., iodinated compounds), ultrasonic contrast agents, and MRI contrast agents (e.g., Gadolinium containing compounds).

[0067] The visualization agent may be present in either a crosslinker or natural component solution. The colored substance may or may not become incorporated into the resulting hydrogel. In embodiments, however, the visualization agent does not have a functional group capable of reacting with the crosslinker or natural component.

[0068] The visualization agent may be used in small quantities, in embodiments less than I% weight/volume, and in other embodiments less that 0.01 % weight/volume and in yet other embodiments less than 0.00 1% weight/volume concentration.

[0069] In embodiments, the bioactive agent may be encapsulated by the hydrogel. For example, the hydrogel may form polymer microspheres around the bioactive agent.
[0070] Where the hydrogel is utilized as a tissue scaffold, it may be desirable to include bioactive agents which promote wound healing and/or tissue growth, including colony stimulating factors, blood proteins, fibrin, thrombin, fibrinogen, hormones and hormone analogs, blood coagulation factors, growth factors, bone morphogenic proteins, TGF-P, IGF, combinations thereof, and the like.

[0071] As the hydrogel hydrolyzes in situ, the bioactive components and any added bioactive agents are released. This may provide nutrients from the natural components, as well as bioactive agents, to the surrounding tissue, thereby promoting growth and/or regeneration of tissue.

[0072] In other embodiments, a method of cell encapsulation is disclosed. The method comprises creating a first solution, such as a cell solution may be combined with a second solution. such as a multi-arm polyethylene glycol (PEG) solution in about a 1:1 ratio (hereinafter a third solution). The third solution may be co-injected into tissue with a natural component. In embodiments, the natural component may include a solution comprising collagen, serum, gelatin, hyaluronic acid and combinations thereof.
The term solution as used herein includes suspensions, emulsions, slurries, and the like.

[0073] In yet other embodiments, a method of cell encapsulation comprises co-injecting into a pre-shaped mold the third solution and the natural component. The mold may be pre-formed to mimic or replicate specific tissue structures such as organs or grafts. The molds may also be shaped to yield an implant or hydrogel which may be used to fill tissue spaces or voids.

[0074] In alternate embodiments. the third solution may be injected into a scaffold or tissue containing the natural component.

[0075] Suitable multi-arm PEGs include those disclosed herein such as 4. 6, and 8-arm PEGs. Additionally, it should be understood that the multi-armed PEGs include electrophilic functional groups. Suitable electrophilic functional groups include those described above.

[0076] Further, buffer concentrations of the natural component solution may vary as a function of the multi-arm PEG MW and the number of arms or branches on the multi-arm PEG. Sodium borate buffer ranges are from about 0.075M to about 0.235 M.

[0077] Suitable cells include those derived from tissue layers, tissue, organs, organ systems, solid and/or hollow, circulatory/bodily fluid-based, etc., including, but not limited to one or more of the following: endo or epithelium; stratum;
adventitia; blood cells (red or white); venous or arterial origin; tissue or blood based inflammatory/immune cells including macrophage/monocytes,T-cells, killer cells, giant cells, neutrophils/polymorphonuclear cells, and the like; lymphatic derived cells;
bone derived cells (osteoblasts, osteoclasts, osteoprogenitor); bone marrow aspirate or derived cells;
stem cells/progenitor cells/adult stem cells; heart/myocardium; blood vessel/artery;
umbilical/fetal tissue/organs/cells; kidney/renal cells; fat; liver; pancreas;
bladder; ureter;
periosteum; fascia; lung; GI tract cells such as esophageal, small/large intestine, colon, rectum, and stomach; neural cells including neurons and glial cells such as (central and/or peripheral nervous system) neurons, astrocytes, schwann cells, oligodendricytes, microglia, olfactory ensheathing cells, dorsal root ganglion cells, neural stem cells and progenitors, brain, cord, or nerve-derived, and optic nerve/retina; eye including various layers of cornea, retina, iris, optic nerve, ciliary muscle, and strabismus;
connective/soft tissue including abdominal wall, pelvic floor, cartilage, meniscus, ligament, tendon, joint capsule, muscle, skin and dermal cells, hair follicles and combinations thereof.

[0078] The aformentioned cells may be a whole population from one or more of the above, or a selected population from one or more of the above. Selected populations may be obtained by a variety of sorting or selection processes, including surface markers, adhesion methods, density methods. genetic methods, size methods, proteomic methods, or any other method used to differentiate cell populations.

[0079] The following Examples are being submitted to illustrate embodiments of the present disclosure. These Examples are intended to be illustrative only and are not intended to limit the scope of the present disclosure. Also, parts and percentages are by weight unless otherwise indicated.

EXAMPLES

[0080] A hydrogel is prepared as follows. Collagen is dissolved in borate buffer (pH
8.75) using a sonicator at about 37 C for approximately 5 minutes to yield a final concentration of from about 3 to about 6 % by weight. The collagen solution is then centrifuged to remove gas/bubbles. Separately, a multi-arm PEG is dissolved in Hanks buffer (at a neutral pH) to yield a final concentration of from about 4 to about 40 % by weight. The two solutions are then simultaneously sprayed in situ at a 1:1 ratio to yield a final gel concentration of from about 1.5 to about 3% by weight collagen, and from about 2.5 to about 20% by weight PEG.

[0081] A hydrogel is prepared as set forth in Example 1, except at a 10:1 ratio collagen:PEG, to yield a final gel concentration of from about 0.5 to about 4%
by weight multiarm PEG and from about 3 to about 6% by weight collagen.

[0082] A hydrogel is prepared as follows. Gelatin is dissolved at a concentration of from about 5 to about 15% by weight in borate buffer at a pH of about 8.25.
Separately, a multi-arm PEG is dissolved in Hanks buffer at a concentration of from about 5 to about 20% by weight. Solutions are simultaneously sprayed in situ at a 1:1 ratio.
yielding a final gel concentration of from about 2.5 to about 7.5% by weight gelatin, and from about 2.5 to about 10% by weight multi-armed PEG.

[0083] A hydrogel is prepared as follows. Hyaluronic acid is dissolved in Hanks buffer at neutral pH, at up to about 2% by weight. A multi-arm PEG is next added to the hyaluronic acid solution at a concentration of from about 5 to about 20% by weight. In a separate vessel, collagen is dissolved in a borate buffer (pH 8.75) at a final solution concentration of from about 3 to about 6% by weight. The 2 solutions are then sprayed at a 1:1 ratio in situ. The final gel concentration is about I% by weight hyaluronic acid, from about 1.5 to about 3% by weight collagen, and from about 2.5 to about 10%
by weight multi-arm PEG.

[0084] The components of the gels of Examples 1, 3 and 4 are summarized below in Table 1.

In Situ Forming Hydrogel Formulations and Conditions xample Material Component 1 Component 2 H Buffers Composition Composition Range an e I Multi-armed PEG: 4aIOkSS, Collagen: bovine PEG in Hanks, PEG & Collagen a20kSG, type 1; 2-6 wt% water or BS, 8al5kSG; saline; DPBS, 0.0239-0.3828 ollagen cell g/mL H>8 culture media 3 Multi-armed PEG: 4a1OkSS, Gelatin: bovine PEG in Hanks, PEG & Gelatin a2OkSG, type B; 5-20 water or PBS, 8a15kSG; % saline; DPBS, 0.0239-0.3828 Gelatin cell g/mL H>8 culture media Multi-armed PEG: 4a2OkSG, Collagen: bovine PEG in Hanks, EG & Collagen 8al5kSG; type 1; 2-6 wt% water or BS, & Hyaluronan 0.0239-0.3828 HA: 74, 130, saline; DPBS, g/mL 770, 910 kDa; Collagen cell p to 2 wt% pH->8 culture media 4al OkSS: 4 arm PEG; Molecular weight (Mw) 10 kDa; SS (succinimidyl succinate)linker 4a2OkSG: 4 arm PEG; Mw 20 kDa; SG (succinimidyl glutarate) linker 8al5kSG: 8 arm PEG; Mw 15 kDa; SG linker *Note, concentrations are for each component prior to mixing at 1:1 100851 A first cell solution was created. Cells were trypsinized, neutralized trypsin, and counted. Cells were then spun down and resuspended in Hank's buffer salt solution to create a first cell solution at a concentration of 4 x 106 cells/mL.

100861 A second solution was created by dissolving multi-armed PEG (200mg/mL) in Hank's buffer salt solution.

[00871 The first solution and second solution were mixed at about a 1:1 ratio to create a third solution.

[0088] Separately, gelatin was solubilized at 10 wt% in 0.235M Sodium Borate buffer @
pH 8.25. The resultant gelatin solution was vortexed, sonicated and spun down to remove bubbles.

[0089] The third solution and the gelation solution were loaded into a dual barrel syringe and co-injected. (The third solution and the gelatin being disposed in separate barrels in the dual barrel syringe.) [0090] It should be noted that while the examples listed above are formed in situ, the materials may be formed ex vivo and then utilized as an implant in situ.

[0091] While the above description contains many specifics, these specifics should not be construed as limitations on the scope of the present disclosure, but merely as exemplifications of preferred embodiments thereof. Those skilled in the art will envision many other possible variations that are within the scope and spirit of the present disclosure.

Claims (17)

WHAT IS CLAIMED IS
1. A method of cell encapsulation comprising:

creating a first solution including suspending cells at a desired concentration in a first buffer salt solution, creating a second solution including dissolving a multi-arm polyethylene glycol at a desired concentration in a second buffer salt solution, mixing the first and second solution at about a 1:1 ratio to create a third solution, co-injecting the third solution with a natural component selected from the group consisting of collagen, serum, gelatin, hyaluronic acid, and combinations thereof.
2. The method of claim 1, wherein the natural component comprises a solution including collagen.
3. The method of claim 1, wherein the natural component comprises a solution including gelatin.
4. The method of claim 1, further comprising recovering a resultant hydrogel.
5. The method of claim 4, wherein the desired concentration of the first solution is about four times a desired final concentration of the cells in the hydrogel.
6. The method of claim 4, wherein the desired concentration of the second solution is about four times a desired final concentration of the multi-arm polyethylene glycol in the hydrogel.
7. The method of claim 1, wherein the cells are derived from tissues selected from the group consisting of endo or epithelium; stratum; adventitia; blood cells (red or white);

venous or arterial origin; tissue or blood based inflammatory/immune cells including macrophage/monocytes,T-cells, killer cells, giant cells, neutrophils/polymorphonuclear cells, and the like; lymphatic derived cells; bone derived cells (osteoblasts, osteoclasts, osteoprogenitor); bone marrow aspirate or derived cells; stem cells/progenitor cells/adult stem cells; heart/myocardium; blood vessel/artery; umbilical/fetal tissue/organs/cells;
kidney/renal cells; fat: liver; pancreas; bladder; ureter; periosteum; fascia;
lung; GI tract cells such as esophageal, small/large intestine, colon, rectum, and stomach;
neural cells including neurons and glial cells such as (central and/or peripheral nervous system) neurons, astrocytes, schwann cells, oligodendricytes, microglia, olfactory ensheathing cells, dorsal root ganglion cells, neural stem cells and progenitors, brain, cord, or nerve-derived, and optic nerve/retina; eye including various layers of cornea, retina, iris, optic nerve, ciliary muscle, and strabismus; connective/soft tissue including abdominal wall, pelvic floor, cartilage, meniscus, ligament, tendon, joint capsule, muscle, skin and dermal cells, hair follicles and combinations thereof.
8. The method of claim 1, wherein the first buffer salt solution comprises Hank's balanced salt solution.
9. The method of claim 1, wherein the second buffer salt solution comprises Hank's balanced salt solution.
10. The method of claim 1, wherein the natural component includes a nucleophilic functional group.
11. The method of claim 1, wherein the natural component comprises endogenous tissue.
12. The method of claim 1, wherein the multi-arm polyethylene glycol comprises and eight-arm n-hydroxysuccinimide group.
13. The method of claim 1, wherein the multi-arm polyethylene glycol comprises and four-arm n-hydroxysuccinimide group.
14. The method of claim 1, wherein the multi-arm polyethylene glycol comprises and six-arm n-hydroxysuccinimide group.
15. The method of claim 1, wherein the natural component comprises a sodium borate buffer solution.
16. The method of claim 1, wherein the natural component comprises a sodium borate buffer solution of from about .075M to about 0.235M.
17. The method of claim 1, further comprising at least one bioactive agent selected from the group consisting of stem cells, DNA, RNA, enzymes, growth factors, peptides, polypeptides, antibodies, and combinations thereof.
CA2745860A 2010-10-08 2011-07-08 Surgical compositions Abandoned CA2745860A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12/900,766 2010-10-08
US12/900,766 US20110081701A1 (en) 2009-10-02 2010-10-08 Surgical compositions

Publications (1)

Publication Number Publication Date
CA2745860A1 true CA2745860A1 (en) 2012-04-08

Family

ID=44677788

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2745860A Abandoned CA2745860A1 (en) 2010-10-08 2011-07-08 Surgical compositions

Country Status (5)

Country Link
US (1) US20110081701A1 (en)
EP (1) EP2457599A3 (en)
JP (1) JP2012081252A (en)
AU (1) AU2011204996A1 (en)
CA (1) CA2745860A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011140517A2 (en) 2010-05-07 2011-11-10 Medicus Biosciences, Llc Methods for treating diseases of the lung
DE102011077809A1 (en) * 2011-06-20 2012-12-20 Charité - Universitätsmedizin Berlin Biocompatible adhesive and process for its preparation
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
EP2846847B1 (en) 2012-05-11 2020-03-25 C.P. Medical Corporation Biocompatible hydrogel treatments for retinal detachment
JP6516721B2 (en) * 2013-03-14 2019-05-22 メディカス バイオサイエンシーズ エルエルシー Biocompatible pre-formulation based on solid polyglycol
DE102014007201B4 (en) * 2014-05-19 2016-03-10 Luphos Gmbh Device and method for geometric measurement of an object
JP7422126B2 (en) * 2018-07-10 2024-01-25 ターガン インコーポレイテッド Methods of preparing and delivering oocyst solutions
CN114642765B (en) * 2021-03-15 2023-04-25 浙江大学 Injectable hydrogel cell scaffold material for treating soft tissue injury and preparation method and application thereof

Family Cites Families (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US435904A (en) * 1890-09-02 sampson
AT366916B (en) 1980-04-02 1982-05-25 Immuno Ag DEVICE FOR APPLICATING A TISSUE ADHESIVE BASED ON HUMAN OR ANIMAL PROTEINS
AT379311B (en) * 1984-03-29 1985-12-27 Immuno Ag DEVICE FOR APPLICATING A TISSUE ADHESIVE
AT382783B (en) * 1985-06-20 1987-04-10 Immuno Ag DEVICE FOR APPLICATING A TISSUE ADHESIVE
DE3722904A1 (en) * 1987-01-09 1988-07-21 Harald Maslanka INJECTION DEVICE WITH DOUBLE CANNULA FOR AN ENDOSCOPE
US4978336A (en) * 1987-09-29 1990-12-18 Hemaedics, Inc. Biological syringe system
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
US5162430A (en) * 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
US5936035A (en) * 1988-11-21 1999-08-10 Cohesion Technologies, Inc. Biocompatible adhesive compositions
US5800541A (en) * 1988-11-21 1998-09-01 Collagen Corporation Collagen-synthetic polymer matrices prepared using a multiple step reaction
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
US5306500A (en) * 1988-11-21 1994-04-26 Collagen Corporation Method of augmenting tissue with collagen-polymer conjugates
US5116315A (en) 1989-10-03 1992-05-26 Hemaedics, Inc. Biological syringe system
US5113315A (en) * 1990-08-07 1992-05-12 Cirqon Technologies Corporation Heat-conductive metal ceramic composite material panel system for improved heat dissipation
US20040195710A1 (en) * 1990-10-15 2004-10-07 Hubbell Jeffrey A. Gels for encapsulation of biological materials
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
US5514379A (en) * 1992-08-07 1996-05-07 The General Hospital Corporation Hydrogel compositions and methods of use
US5919455A (en) * 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
EP0713707A1 (en) * 1994-11-23 1996-05-29 Collagen Corporation In situ crosslinkable, injectable collagen composition for tissue augmention
US6129761A (en) * 1995-06-07 2000-10-10 Reprogenesis, Inc. Injectable hydrogel compositions
US6833408B2 (en) * 1995-12-18 2004-12-21 Cohesion Technologies, Inc. Methods for tissue repair using adhesive materials
US5752974A (en) * 1995-12-18 1998-05-19 Collagen Corporation Injectable or implantable biomaterials for filling or blocking lumens and voids of the body
PT876165E (en) * 1995-12-18 2006-10-31 Angiotech Biomaterials Corp COMPOSITIONS OF RETICULATED POLYMERS AND PROCESSES FOR THEIR USE
US7435425B2 (en) * 2001-07-17 2008-10-14 Baxter International, Inc. Dry hemostatic compositions and methods for their preparation
US7009034B2 (en) * 1996-09-23 2006-03-07 Incept, Llc Biocompatible crosslinked polymers
US6566406B1 (en) * 1998-12-04 2003-05-20 Incept, Llc Biocompatible crosslinked polymers
DK1053019T3 (en) * 1998-01-07 2004-04-13 Debio Rech Pharma Sa Degradable heterobifunctional polyethylene glycol acrylates and gels and conjugates derived therefrom
JP4078032B2 (en) * 1998-03-12 2008-04-23 ネクター セラピューティックス エイエル,コーポレイション Poly (ethylene glycol) derivatives with proximal reactive groups
US6152943A (en) * 1998-08-14 2000-11-28 Incept Llc Methods and apparatus for intraluminal deposition of hydrogels
US6605294B2 (en) * 1998-08-14 2003-08-12 Incept Llc Methods of using in situ hydration of hydrogel articles for sealing or augmentation of tissue or vessels
US6818018B1 (en) * 1998-08-14 2004-11-16 Incept Llc In situ polymerizable hydrogels
CA2340381C (en) * 1998-08-14 2009-01-13 Incept Llc Methods and apparatus for in situ formation of hydrogels
US6514534B1 (en) * 1998-08-14 2003-02-04 Incept Llc Methods for forming regional tissue adherent barriers and drug delivery systems
US6179862B1 (en) * 1998-08-14 2001-01-30 Incept Llc Methods and apparatus for in situ formation of hydrogels
US7347850B2 (en) * 1998-08-14 2008-03-25 Incept Llc Adhesion barriers applicable by minimally invasive surgery and methods of use thereof
US6703047B2 (en) * 2001-02-02 2004-03-09 Incept Llc Dehydrated hydrogel precursor-based, tissue adherent compositions and methods of use
US7279001B2 (en) * 1998-11-06 2007-10-09 Neomend, Inc. Systems, methods, and compositions for achieving closure of vascular puncture sites
US6830756B2 (en) * 1998-11-06 2004-12-14 Neomend, Inc. Systems, methods, and compositions for achieving closure of vascular puncture sites
US8802146B2 (en) * 1998-11-06 2014-08-12 Neomend, Inc. Systems, methods, and compositions for prevention of tissue adhesion
US6899889B1 (en) * 1998-11-06 2005-05-31 Neomend, Inc. Biocompatible material composition adaptable to diverse therapeutic indications
US20080114092A1 (en) * 1998-12-04 2008-05-15 Incept Llc Adhesion barriers applicable by minimally invasive surgery and methods of use thereof
US6662805B2 (en) * 1999-03-24 2003-12-16 The Johns Hopkins University Method for composite cell-based implants
ES2391763T3 (en) * 2000-04-07 2012-11-29 Collagen Matrix, Inc. Embolization device
US7265186B2 (en) * 2001-01-19 2007-09-04 Nektar Therapeutics Al, Corporation Multi-arm block copolymers as drug delivery vehicles
MXPA04004336A (en) * 2001-11-07 2005-05-16 Nektar Therapeutics Al Corp Branched polymers and their conjugates.
CA2493083C (en) * 2002-07-16 2012-11-06 Bio Syntech Canada Inc. Composition for cytocompatible, injectable, self-gelling chitosan solutions for encapsulating and delivering live cells or biologically active factors
CA2511815A1 (en) * 2002-12-26 2004-07-22 Mountain View Pharmaceuticals, Inc. Polymer conjugates of cytokines, chemokines, growth factors, polypeptide hormones and antagonists thereof with preserved receptor-binding activity
US7316811B2 (en) * 2002-12-30 2008-01-08 Nektar Therapeutics Al, Corporation Multi-arm polypeptide-poly (ethylene glycol) block copolymers as drug delivery vehicles
CA2519092C (en) * 2003-03-14 2014-08-05 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
US7351787B2 (en) * 2004-03-05 2008-04-01 Bioartificial Gel Technologies, Inc. Process for the preparation of activated polyethylene glycols
US7919112B2 (en) * 2004-08-26 2011-04-05 Pathak Holdings, Llc Implantable tissue compositions and method
US20060062768A1 (en) * 2004-09-23 2006-03-23 Olexander Hnojewyj Biocompatible hydrogel compositions
US7365127B2 (en) * 2005-02-04 2008-04-29 Enzon Pharmaceuticals, Inc. Process for the preparation of polymer conjugates
US7541541B1 (en) * 2005-11-03 2009-06-02 Taymac Corporation Safety outlet cover
US20070292404A1 (en) * 2006-03-27 2007-12-20 Biosynexus Incorporated Antimicrobial polymer conjugates
EP2023941B1 (en) * 2006-04-24 2016-12-21 Incept, LLC Protein crosslinkers, crosslinking methods and applications thereof
US7960498B2 (en) * 2006-06-30 2011-06-14 Actamax Surgical Materials, Llc Tissue adhesives with modified elasticity
US9271706B2 (en) * 2008-08-12 2016-03-01 Covidien Lp Medical device for wound closure and method of use

Also Published As

Publication number Publication date
US20110081701A1 (en) 2011-04-07
AU2011204996A1 (en) 2012-04-26
EP2457599A3 (en) 2014-09-03
EP2457599A2 (en) 2012-05-30
JP2012081252A (en) 2012-04-26

Similar Documents

Publication Publication Date Title
US8968785B2 (en) Surgical compositions
US20110081701A1 (en) Surgical compositions
US9468684B2 (en) Hydrogel implants with varying degrees of crosslinking
US9855372B2 (en) Auto-sealant matrix for tissue repair
US8591950B2 (en) Hydrogel implants with varying degrees of crosslinking
US8734824B2 (en) Hydrogel implants with varying degrees of crosslinking
US9144611B2 (en) Hydrogel implants with varying degrees of crosslinking
US8591929B2 (en) Hydrogel implants with varying degrees of crosslinking
WO2013032623A2 (en) Hydrogel implants with varying degrees of crosslinking
US20110081398A1 (en) Multi-mechanism surgical compositions
AU2012313983B2 (en) Multilayer implants for delivery of therapeutic agents
US9795717B2 (en) Enzymatically degradable compositions
US20140271613A1 (en) Surgical Compositions

Legal Events

Date Code Title Description
FZDE Discontinued

Effective date: 20150708

FZDE Discontinued

Effective date: 20150708