US20140256617A1 - In Situ Forming Hydrogel and Method Using Same - Google Patents

In Situ Forming Hydrogel and Method Using Same Download PDF

Info

Publication number
US20140256617A1
US20140256617A1 US14/351,549 US201214351549A US2014256617A1 US 20140256617 A1 US20140256617 A1 US 20140256617A1 US 201214351549 A US201214351549 A US 201214351549A US 2014256617 A1 US2014256617 A1 US 2014256617A1
Authority
US
United States
Prior art keywords
hydrogel
repeat units
polymer
lcst
terpolymer
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
US14/351,549
Inventor
Derek Overstreet
Brent Vernon
Ryan McLemore
Alex McLaren
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.)
Banner Health
Arizona State University ASU
Original Assignee
Banner Health
Arizona State University ASU
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 Banner Health, Arizona State University ASU filed Critical Banner Health
Priority to US14/351,549 priority Critical patent/US20140256617A1/en
Assigned to ARIZONA BOARD OF REGENTS, A BODY CORPORATE OF THE STATE OF ARIZONA ACTING FOR AND ON BEHALF OF ARIZONA STATE UNIVERSITY, BANNER HEALTH reassignment ARIZONA BOARD OF REGENTS, A BODY CORPORATE OF THE STATE OF ARIZONA ACTING FOR AND ON BEHALF OF ARIZONA STATE UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VERNON, BRENT, MCLAREN, Alex, MCLEMORE, Ryan, OVERSTREET, Derek
Publication of US20140256617A1 publication Critical patent/US20140256617A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/14Peptides containing saccharide radicals; Derivatives thereof, e.g. bleomycin, phleomycin, muramylpeptides or vancomycin
    • 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
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0024Solid, semi-solid or solidifying implants, which are implanted or injected in body tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action
    • A61L2300/404Biocides, antimicrobial agents, antiseptic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2400/00Materials characterised by their function or physical properties
    • A61L2400/06Flowable or injectable implant compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/24Materials or treatment for tissue regeneration for joint reconstruction

Definitions

  • In situ forming hydrogels are useful for a variety of biological and biomedical applications including drug delivery, embolization, cell encapsulation and culture, and tissue regeneration.
  • a hydrogel is presented.
  • An aqueous solution of a polymer comprises a first lower critical solution temperature (“LCST”).
  • LCST first lower critical solution temperature
  • the hydrogel is configured to be converted in vivo into a modified hydrogel comprising a modified polymer, wherein an aqueous solution of the modified polymer comprises a second lower critical solution temperature (“LCST”), and wherein the second LCST is greater than the first LCST.
  • LCST lower critical solution temperature
  • Applicants' hydrogel provides an in situ-forming and degradable platform for the controlled local release of drugs when dissolved in physiologically compatible aqueous solutions.
  • Applicants' hydrogel can be used as injectable or space-filling carriers for localized delivery of antibiotics or other drugs for treatment or prevention of orthopaedic infections such as in total hip arthroplasty.
  • Applicants' hydrogel can be administered to nearly any location in the body where a local pathology might arise. After gelation, the resulting gel begins to release one or more medicaments.
  • the hydrogel can a,so be administered to some locations in the body for long-term systemic delivery. Eventually, all the entrapped one or more medicaments are released and the gel dissolves.
  • the various embodiments of Applicants' hydrogel comprise a polymer with a lower critical solution temperature (LCST) in aqueous solution that is less than body temperature.
  • LCST critical solution temperature
  • the polymer is soluble in water.
  • that aqueous polymer solution forms a soft and solid water-swollen hydrogel.
  • pendent ester groups hydrolyze to form a modified hydrogel, wherein the modified hydrogel comprises a higher LCST.
  • the modified hydrogel dissolves, leaves the site of administration, and is excreted from the body via the urine.
  • FIG. 1 graphically illustrates relative absorbance at 450 nm wavelength of 0.25 wt % solutions of Applicants' hydrogel I;
  • FIG. 2 shows a LCST of 5 wt % solutions of poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA, 2.3 mol % JAAm determined by cloud point as a function of time, wherein NIPAAm is N-isopropylacrylamide, DBLA is (R)- ⁇ -Acryloyloxy- ⁇ , ⁇ -dimethyl- ⁇ -butyrolactone, and JAAm is Jeffamine M-1000 acrylamide;
  • FIG. 3 shows deswelling of a control polymer 30 wt % poly(NIPAAm-co-DBLA) at various times after gelation in excess PBS (pH 7.4) at 37° C.;
  • FIG. 4 shows swelling and degradation behavior of Applicants' terpolymer I based gel comprising poly(NIPAAm-co-DBLA-co-JAAm) in excess PBS (pH 7.4);
  • FIG. 5 graphically illustrates the mass of vancomycin released from 1 mL gels loaded with 50,000 micrograms vancomycin HCl and 1 mL bone cement samples loaded with 142,000 micrograms vancomycin HCl;
  • FIG. 6 graphically illustrates an amount of vancomycin released as a fraction of the drug loaded
  • FIG. 7 graphically illustrates a fraction of loaded vancomycin released from Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) gels loaded with either 50 mg or 5 mg per gel; and
  • FIG. 8 summarizes Applicants' method to deliver a medicament to an injection site within the body of an animal, including a human.
  • In situ forming biomaterials which transition from liquid to solid under physiological conditions are promising for a variety of medical applications, such as tissue repair, embolization, and drug delivery. Administering these materials by injection provides reduced infection risk, scar formation, and treatment cost relative to implantation. In situ forming materials can provide local, controlled delivery of therapeutics directly to the desired area with reduced risk of side effects compared with commonly performed systemic delivery. In addition to ease of administration and delivery properties, materials that form in situ are ideal for use in spaces that are irregular or difficult to access with a solid implant, since the shape of an in situ forming implant is defined by its local environment.
  • NIPAAm N-isopropylacrylamide
  • LCST critical solution temperature
  • poly(NIPAAm) As a solution of poly(NIPAAm) is heated above its LCST (near 32° C.), polymer-polymer interactions between the hydrophobic isopropyl groups become more favorable than the polymer-water interactions with NIPAAm side group. Upon this transition, the polymer chains collapse to form a semisolid hydrogel at sufficiently high polymer concentrations for chain entanglement.
  • Various properties such as polymer LCST, chemical reactivity, sensitivity to various stimuli (pH, enzymes), or gel swelling can be controlled by incorporating small molar fractions ( ⁇ 10%) of comonomers with NIPAAm in order to provide a material for a particular application. For example, hydrophobic comonomers decrease the LCST while hydrophilic comonomers increase the LCST.
  • JEFFAMINE M-1000 is a random copolymer of ethylene oxide (EO) and propylene oxide (PO) in a 19:3 EO:PO ratio with one terminal amine and one terminal methoxy group.
  • EO ethylene oxide
  • PO propylene oxide
  • hydrogel represents a more clinically viable class of injectable materials for drug delivery by imparting swelling and drug release control via JEFFAMINE M-1000 in a resorbable polymer.
  • TAA total hip arthroplasty
  • total hip arthroplasty is a very common procedure, with more than 200,000 performed in the US each year. By 2030, this number is estimated to increase to 572,000.
  • the incidence of prosthetic joint infection in total hip and total knee arthroplasties is approximately 1.5-2.5% for primary interventions and higher for revision procedures performed secondary to infection. While the incidence of infection is relatively low, the high number of operations overall combined with the high cost of treating an infected arthroplasty (over $50,000 in Year 1995 dollars) make this an area of great need.
  • R1 and R2 are independently selected from the group consisting of H, alkyl, phenyl, benzyl, 2-cyanoprop-2-yl, 4-cyanopentanoic acid-4-ylethyl-2-propionate, sulfate, 2-[2-methoxypropan-2-yl)oxy]propan-2-yl, and a dithioester derived from a RAFT chain transfer agent such as 4-cyano-4-(ethylsulfanylthiocarbonyl)sulfanylpentanoic acid.
  • pendent group R8 comprises a linkage with the polymer backbone selected from the group consisting of an ester, amide, thioamide, thiourea, anhydride, thioester, thiourea, and alkyl. In certain embodiments, pendent group R8 comprises at least one ester or anhydride.
  • linkage R8 is selected from the group consisting of ester, amide, thioamide, thiourea, anhydride, thioester, thiourea, and alkyl. In certain embodiments, either linkage R8 or pendent group R6 comprises at least one ester or anhydride.
  • R6 is selected from the group consisting of alkyl phenyl, polyesters, and lactones. In certain embodiments, R6 comprises a substituted butyrolactone.
  • R7 comprises an amide linkage. In certain embodiments, R7 comprises a thioamide linkage. In certain embodiments, R7 comprises a urea linkage. In certain embodiments. R7 comprises a thiourea linkage. In certain embodiments, R7 comprises an ester linkage. In certain embodiments, R7 comprises an anhydride linkage.
  • the water soluble polymer comprises a polyether.
  • the water soluble polymer comprises polyether II formed by ring opening polymerization of ethylene oxide, wherein R9 is selected from the group consisting of H, methyl, methoxy, and hydroxyl.
  • n is between about 5 and about 2500.
  • the water soluble polymer comprises polyether III formed by ring opening polymerization of propylene oxide.
  • n is between about 15 and about 250.
  • the water soluble polymer comprises polyether IV formed by co-polymerization of ethylene oxide and propylene oxide.
  • r is between about 5 and about 2500, and p is between about 1 and about 1000.
  • the water soluble polymer comprises polyether V formed by ring opening polymerization of tetrahydrofuran. In certain embodiments, n is between about 10 and about 50.
  • the water soluble polymer comprises a water-soluble polymer of one or more of the following: vinyl alcohol, acrylic acid, methacrylic acid, 2-hydroxyethyl methacrylate, N-2 hydroxypropylmethacrylamide, vinylpyrrolidone, or a monosaccharide.
  • graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XI using monomer XII.
  • graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XIII using monomer XIV.
  • graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XV using monomer XVI.
  • graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XVII using monomer XVIII.
  • graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XIX using monomer XX.
  • graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XXI using monomer XXII.
  • a is between about 10 and about 1000, b is between about 1 and about 250, and c is between about 1 and about 500.
  • Terpolymer I can be synthesized via a number of different procedures.
  • Applicants' graft terpolymer I can be prepared by free radical polymerization.
  • Graft terpolymer I can also be prepared by reversible addition-fragmention chain transfer (“RAFT”) polymerization.
  • RAFT reversible addition-fragmention chain transfer
  • a RAFT polymerization can be performed by adding a quantity of a RAFT agent (thiocarbonylthio compounds) to a conventional free radical polymerization.
  • a RAFT agent thiocarbonylthio compounds
  • free radical polymerization usually the same monomers, initiators, solvents and temperatures can be used. Because of the low concentration of the RAFT agent in the system, the concentration of the initiator is usually lower than in conventional radical polymerization.
  • Radical initiators such as azobisisobutyronitrile (AIBN) and 4,4′-Azobis(4-cyanovaleric acid) (ACVA) which is also called 4,4′-Azobis(4-cyanopentanoic acid) are widely used as the initiator in RAFT.
  • RAFT polymerization is known for its compatibility with a wide range of monomers, including for example acrylates and acrylamides.
  • Graft terpolymer I as either a random copolymer or a block copolymer, can also be prepared by atom transfer radical polymerization (“ATRP”).
  • ATRP atom transfer radical polymerization
  • NIPAAM N-isopropylacrylamide
  • ATRP atom transfer radical polymerization
  • ECP ethyl 2-chloropropionate
  • Me 6 TREN CuCl/tris(2-dimethylaminoethyl)amine
  • An embodiment of Applicant's hydrogel I comprises a random terpolymer formed using three monomers, namely N-isopropylacrylamide (NIPAAm), JEFFAMINE M-1000 acrylamide (JAAm), and acryloyloxy- ⁇ , ⁇ -dimethyl- ⁇ -butyrolactone (DBLA).
  • NIPAAm N-isopropylacrylamide
  • JAAm JEFFAMINE M-1000 acrylamide
  • DBLA acryloyloxy- ⁇ , ⁇ -dimethyl- ⁇ -butyrolactone
  • NIPAAm monomer was recrystallized from hexane.
  • Azobisisobutyronitrile (AIBN) was recrystallized from methanol.
  • HPLC grade tetrahydrofuran (THF) was used for low molecular weight polymerizations and as the mobile phase for molecular weight and polydispersity determination.
  • JEFFAMINE M-1000 polyetheramine was donated by Huntsman Corporation (The Woodlands, Tex., USA).
  • JEFFAMINE M-1000 acrylamide was synthesized from JEFFAMINE M-1000 polyetheramine by acryloylation with acryloyl chloride.
  • JEFFAMINE M-1000 (20 g, 20 mmol) was dissolved at 10 w/v % in dichloromethane (DCM) along with triethylamine (3.3 mL, 24 mmol) and maintained at 0° C. under nitrogen atmosphere.
  • Acryloyl chloride (1.95 mL, 24 mmol) was then added dropwise into the solution under stirring and the reaction was allowed to proceed for 1 hour at 0° C. and then at room temperature for at least 5 hours under nitrogen atmosphere.
  • Hydrogel I prepared using NIPAAm, DBL, and JAAm were synthesized by radical polymerization with 80% dioxane/20% THF as the solvent, but other solvents may be used, including methanol, ethanol, water, benzene, acetone, isopropanol, pure dioxane, pure tetrahydrofuran, or others. All three monomers (NIPAAm, DBLA, and JAAm) were dissolved in the solvent. Monomer solution (10 w/v %) was bubbled with nitrogen for at least 10 minutes prior to addition of initiator to reduce dissolved oxygen. After this time, AIBN (initiator) was added ( ⁇ 0.007 mol AIBN/mol of total monomer) and the reaction allowed to proceed for 24 hr at 65° C. under a slight positive pressure of nitrogen.
  • Applicants' hydrogel was collected by precipitation into 10 to 15-fold excess of a solvent in which the polymer is not soluble, such as chilled diethyl ether or hexane. Following precipitation, the product was collected by filtration and dried overnight under vacuum. The product was then dissolved in deionized water and can optionally be dialyzed against 3500 MWCO at 4° C. for 20 hours with the excess water replaced 3 times in order to purify the material, removing any low molecular weight impurities. Materials were then lyophilized and can be stored at ⁇ 20° C. under nitrogen atmosphere until use.
  • a solvent in which the polymer is not soluble such as chilled diethyl ether or hexane.
  • the materials can be sterilized (such as by ethylene oxide gas sterilization) and then dissolved in 150 mM phosphate buffered saline (pH 7.4). After dissolution of the polymer, drug powder (finely ground) is added directly to the polymer solution and vortex mixed to distribute the drug evenly throughout the solution.
  • the solutions can be stored in a freezer until use and thawed at the time of use. The solution then can be injected, brushed, poured, sprayed, or otherwise administered as a liquid at the desired site. The solution can also be allowed to warm to above its initial LCST and then implanted as a soft hydrogel at the desired site.
  • desired sites for delivery of this material might include the surface of a joint prosthesis, the intramedullary canal before implantation of a prosthesis, an infected area, or any location deemed to have a risk of future infection.
  • An example formulation might be a 30 wt % polymer solution with approximately 50 mg vancomycin hydrochloride loaded per mL of polymer solution.
  • Hydrogels are defined as three-dimensional (3D), water-swollen polymer networks formed as a result of physical or chemical cross-linking. Because of their high water content and mechanical resemblance to natural tissues, hydrogels show promising biocompatibility and potential for medical applications. Injectable hydrogel formulations are especially attractive due to their minimally invasive delivery procedure, providing reduced healing time, reduced scarring, decreased risk of infection, and ease of delivery compared with surgically implanted materials.
  • Polymeric hydrogels which undergo crosslinking during or after administration to a joint space may represent an improvement because such hydrogels undergoes an increase in viscosity within the joint space following administration.
  • the hydrogel precursor may be administered as a relatively less viscous liquid, then forming a much more viscous or viscoelastic gel within a timeframe of less than one hour. Additionally, this higher viscosity imparted by crosslinking in situ could lead to prolonged residence time in the joint and therefore a longer treatment duration achieved with a single administration.
  • Some in situ crosslinking hydrogels are viscoelastic, providing properties that can allow for the gel to remain intact within the joint by yielding during joint motion, conforming to the shape of the joint space in a way that is analogous to a highly viscous liquid. Because of their rheological properties, viscoelastic hydrogels are especially well-suited to conform to the surface of a prosthetic joint (or other surfaces present in a joint) to provide effective and sustained local concentrations of drug within the joint.
  • Applicants utilize a crosslinked hydrogel injected into a joint space.
  • crosslinked hydrogel Applicants mean a physically or chemically crosslinked polymer-containing material containing over 50 weight percent of an aqueous solvent.
  • the gel is crosslinked prior to injection.
  • the gel is crosslinked after injection in the joint space.
  • Applicants' method administers a hydrogel in vivo to a joint space of an animal.
  • the hydrogel forms by physical crosslinking (for example, by heating above an LCST).
  • the crosslinked hydrogel forms by supramolecular self-assembly.
  • the crosslinked hydrogel forms by covalent crosslinking.
  • the crosslinked hydrogel is administered by transcutaneous injection. In certain embodiments, the crosslinked hydrogel is administered to the joint space during a surgical procedure.
  • the crosslinked hydrogel contains no medicament. In certain embodiments, the crosslinked hydrogel comprises a medicament. In certain embodiments, the medicament comprises an antimicrobial.
  • the crosslinked hydrogel is administered to the hip joint in conjunction with total hip arthroplasty. In certain embodiments, the crosslinked hydrogel is administered to the knee joint in conjunction with total knee arthroplasty. In certain embodiments, the joint space is the hip joint. In certain embodiments, the joint space is the knee joint.
  • Applicants' injected hydrogel I in solution, becomes a soft semi-solid filler which steadily delivers antibiotics directly at the surface of the implant which may do so without disrupting the tit between the bone and the prosthesis or generating wear debris from joint motion.
  • the drug concentration near the implant can be maintained at a high level in order to prevent infection, while the systemic exposure to the drug (and the corresponding side effects) can remain low.
  • PMMA poly(methyl methacrylate)
  • the drug(s) are finely ground and mixed into the cement before implantation.
  • Local delivery of drugs drug (drugs administered near the implant site) is required in order to provide high drug concentration at the desired site with low concentration elsewhere in the body.
  • Systemic administration (such as by intravenous infusion) does not effectively target the implant surface and can cause toxicity and side effects at a dose that would be therapeutic at the implant surface.
  • drug release is not sustained. Drug must be maintained at the site of infection at as a high concentration for as long of a time as possible in order to destroy a bacterial biofilm, which can require 1000 times as much drug concentration to inhibit growth as an individual planktonic bacterium. Most of the drug release from cement occurs within the first 1-2 days, with very low release thereafter.
  • bone cement is not degradable. While bone cement is known to be biocompatible, it cannot be used with cementless hip stems and is not re-dosable without an invasive procedure.
  • bone cement is not indicated for use with many cementless hip prosthesis designs.
  • aqueous solution of Applicants' injectable hydrogel composition I overcomes the deficiencies of prior art bone cement.
  • Applicants' hydrogel I provides efficient drug release. Because the material eventually dissolves, all of the drug is eventually delivered and utilized. Local delivery provides the highest drug concentration directly where it is needed—at the implant surface,
  • Applicants' hydrogel provides sustained drug release. Throughout the 7-10 day period during which the material dissolves, drug is released at a relatively steady rate, which may provide improved performance at preventing or managing prosthetic joint infections.
  • hydrogel provides local delivery to the implant surface of cementless stems without disrupting the quality of fit between the hip prosthesis and the femur.
  • Applicants' hydrogel provides degradability. Previous work by Applicants' and colleagues has shown no difference in immune response from that of native tissue following gel dissolution. The degradability of the material also allows for it to be re-dosable by transcutaneous injection in some cases.
  • FIG. 1 graphically illustrates relative absorbance of 0.25 wt % solutions of Applicants' hydrogel I comprising poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA, 2.3 mol % JAAm incubated at 37° C. in Phosphate Buffer Solution after 1 day ( ⁇ ), 3 days ( ⁇ ), 7 days ( ⁇ ), and 15 days ( ⁇ ).
  • DBLA dimethyl-butyrolactone acrylate and JAAm is JEFFAMINE M-1000 acrylamide.
  • FIG. 2 shows the LCST of 5 wt % solutions of poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA, 2.3 mol % JAAm determined by cloud point as a function of time.
  • the LCST for the same polymer without JAAm remains at 18° C. for the first 2 weeks.
  • human body temperature is about 37° C.
  • FIG. 3 shows deswelling of a control polymer 30 wt % poly(NIPAAm-co-DBLA) at various times after gelation in excess PBS (pH 7.4) at 37° C.
  • the control copolymer comprises no absence of JAAm. Gels began shrinking rapidly beginning on the first day. These gels dissolve in approximately 20 weeks.
  • FIG. 4 shows swelling and degradation behavior of Applicants' terpolymer I based gel comprising poly(NIPAAm-co-DBLA-co-JAAm) in excess PBS (pH 7.4). Gels retained their initial volume throughout the study and underwent bulk degradation. Gels flow when inverted after 7-10 days and dissolve completely within 2-3 weeks.
  • FIG. 5 graphically illustrates the mass of vancomycin released from 1 mL gels loaded with 50,000 micrograms vancomycin HCl and 1 mL bone cement samples loaded with 142,000 micrograms vancomycin HCl. Release is prolonged and increased from Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) as compared to clinically used bone cement.
  • FIG. 6 graphically illustrates an amount of vancomycin released as a fraction of the drug loaded. Most of the drug that is loaded into bone cement is not released whereas Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) release nearly all of the drug loaded into them within 8 days.
  • FIG. 7 graphically illustrates a fraction of loaded vancomycin released from Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) gels loaded with either 50 mg or 5 mg per gel. Some vancomycin is insoluble when loaded into the polymer solution at 50 mg/mL. The similarity between these curves indicates that the polymer, as opposed to the drug solubility, is primarily responsible for controlling the rate of drug release.
  • FIG. 8 summarizes Applicants' method to deliver a medicament to an injection site within the body of an animal, including a human.
  • the injection site comprises the surface of an bone implant.
  • the method provides a medicament and a first hydrogel comprising a first LCST less than the body temperature of the subject animal.
  • body temperature for a human is about 37° C.
  • the first hydrogel of step 810 comprises a LCST less than about 37° C.
  • Applicants mean a material selected from the group consisting of a Nucleic acid, a Protein (including growth factors, bone morphogenetic proteins), a Polypeptide, a Contrast agent for imaging an Anesthetic, an Antineoplastic agent, an Antifungal, an Anti-inflammatory drug (steroids, non-steroidal anti-inflammatory drugs (NSAIDs), and an Antibiotic.
  • the Antibiotic comprises one or more of Aminoglycosides, including gentamicin, amikacin, and tobramycin, Cephalosporins including cefazolin, Vancomycin, and Rifampin
  • the first hydrogel of step 810 comprises a polymeric material comprising a backbone formed from polymerization of one or more substituted acrylamides, one or more monomers containing one or more hydrolyzable linkages, and a monomer comprising a water soluble polymer moiety attached thereto.
  • the first hydrogel of block 810 comprises Applicants' hydrogel I.
  • the first hydrogel of block 810 comprises Applicant's hydrogel formed from N-isopropylacrylamide, JEFFAMINE M-1000 acrylamide (JAAm), and acryloyloxy- ⁇ , ⁇ -dimethyl- ⁇ -butyrolactone.
  • the first hydrogel of step 810 comprises Applicant's hydrogel comprising poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA and 2.3 mol % JAAm.
  • an aqueous solution of the medicament and the first hydrogel of block 810 is injected into a selected animal at a temperature less than the first LCST.
  • the injection site comprises a tissue space wherein a subsequently formed gel will substantially completely fill that tissue space.
  • the first hydrogel of block 810 is utilized in conjunction with implantation of an artificial joint. In certain of these embodiments, the injection of block 820 is performed after implantation such that the injected hydrogel is disposed adjacent a surface of the implanted artificial joint.
  • the hydrogel of block 810 is coated onto a surface of an artificial joint prior to implantation.
  • the “injection” of block 820 comprises implantation of the artificial joint comprising a surface coated with the first hydrogel of block 810 .
  • the first hydrogel of block 810 injected into the body of an animal in block 820 is warmed in vivo to a temperature greater than the first LCST.
  • the warming of block 830 is performed by the body heat of the animal.
  • the warming of block 830 is performed by disposing a heating object, such as for example and without limitation, a heat lamp, a heating pad, hot compress, and the like, directed in near proximity to the injection site.
  • the first hydrogel of block 810 injected into the body of an animal in block 820 and warmed in vivo to a temperature greater than the first LCST in block 830 forms in vivo a water-insoluble gel.
  • the water-insoluble gel of block 840 is formed in, and substantially fills, a tissue space.
  • the water-insoluble gel of block 840 is disposed on, and in near vicinity to, a surface of a joint implant.
  • the water-insoluble gel of block 840 releases the medicament of block 810 into tissues adjacent the injection site of block 820 .
  • the release of medicament is sustained over time.
  • the release is approximately proportional to the square root of time over the first 60% of release, with a slower rate of release thereafter.
  • the first hydrogel of block 810 which was injected in block 820 is chemically modified to form a second hydrogel having a second LCST, wherein the second LCST is greater than body temperature.
  • the pendent ester moieties of the plurality of repeat units IV are hydrolyzed in vivo to convert first terpolymer I injected in block 820 and gelled in block 840 to a second terpolymer XXII, wherein R9 comprises a pendent hydroxyl group or a pendent carboxylic acid group resulting from a hydrolysis of an acid moiety or an anhydride moiety, respectively, in pendent group R8.
  • the pendent substituted butyrolactone moieties of the plurality of repeat units IV are hydrolyzed in vivo to convert first terpolymer I injected in block 820 and gelled in block 840 to a second terpolymer XXIII, wherein R9 comprises a carboxylic acid moiety.
  • the first hydrogel of block 810 is converted into a second hydrogel XXII/XXIII that is water soluble at body temperature.
  • the water soluble second hydrogel dissolves in body fluids.
  • step 880 the second hydrogel of step 870 is excreted from the body.
  • a first hydrogel having a LCST less than body temperature and a medicament, in aqueous solution are injected into the body of an animal.
  • the first hydrogel is warmed to a temperature greater than its LCST, and a water-insoluble gel is formed.
  • the medicament is released over time from the water-insoluble gel.
  • the first hydrogel is converted in vivo into a second hydrogel having a LCST greater than body temperature.
  • This second hydrogel is water soluble, and is carried by the animal's circulatory system to its kidneys. The second hydrogel is then excreted from the body.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Dermatology (AREA)
  • Engineering & Computer Science (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Transplantation (AREA)
  • Biomedical Technology (AREA)
  • Inorganic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Dispersion Chemistry (AREA)
  • Neurosurgery (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Materials For Medical Uses (AREA)

Abstract

A hydrogel (I), comprising a polymer backbone wherein an aqueous solution of the hydrogel comprises a first lower critical solution temperature, and wherein the hydrogel is configured to be converted in vivo into a modified hydrogel, and wherein an aqueous solution of the modified hydrogel comprises a second lower critical solution temperature, and wherein the second lower critical solution temperature is greater than the first lower critical solution temperature.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is based on, claims a priority benefit from, and incorporates herein by reference U.S. Patent Application No. 61/546,402, filed Oct. 12, 2011, and entitled, “In Situ Forming Hydrogel and Method Using Same.”
  • BACKGROUND OF THE INVENTION
  • In situ forming hydrogels are useful for a variety of biological and biomedical applications including drug delivery, embolization, cell encapsulation and culture, and tissue regeneration.
  • For most applications where in situ forming hydrogels are used, resistance to shrinking is an important consideration. Usually, the ideal case is that the material transitions quickly from liquid to solid with almost no change in volume. Shrinking or swelling inherently causes changes in the hydrogel's mechanical properties, porosity, and size. Wound healing and embolization applications require retention of the hydrogel's original size at the injection site and good contact with the surrounding tissue. For controlled drug delivery, a fast sol-to-gel transition without syneresis could reduce the high initial burst release of hydrophilic drugs typical of many in situ forming materials. For successful use as synthetic extracellular matrices in vitro or in vivo, gels must retain a high volume fraction of water in order to support cell growth.
  • SUMMARY OF THE INVENTION
  • A hydrogel is presented. An aqueous solution of a polymer comprises a first lower critical solution temperature (“LCST”). When the polymer is heated above its first LCST, a hydrogel is formed. The hydrogel is configured to be converted in vivo into a modified hydrogel comprising a modified polymer, wherein an aqueous solution of the modified polymer comprises a second lower critical solution temperature (“LCST”), and wherein the second LCST is greater than the first LCST.
  • Applicants' hydrogel provides an in situ-forming and degradable platform for the controlled local release of drugs when dissolved in physiologically compatible aqueous solutions. Applicants' hydrogel can be used as injectable or space-filling carriers for localized delivery of antibiotics or other drugs for treatment or prevention of orthopaedic infections such as in total hip arthroplasty.
  • More generally, Applicants' hydrogel can be administered to nearly any location in the body where a local pathology might arise. After gelation, the resulting gel begins to release one or more medicaments. The hydrogel can a,so be administered to some locations in the body for long-term systemic delivery. Eventually, all the entrapped one or more medicaments are released and the gel dissolves.
  • The various embodiments of Applicants' hydrogel comprise a polymer with a lower critical solution temperature (LCST) in aqueous solution that is less than body temperature. At temperatures lower than the LCST, the polymer is soluble in water. When heated above the LCST, that aqueous polymer solution forms a soft and solid water-swollen hydrogel. This allows for the material to be applied or injected as a liquid and set up to form a soft solid at body temperature. In the time following injection, pendent ester groups hydrolyze to form a modified hydrogel, wherein the modified hydrogel comprises a higher LCST. As the LCST of the modified hydrogel increases above body temperature, the modified hydrogel dissolves, leaves the site of administration, and is excreted from the body via the urine.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 graphically illustrates relative absorbance at 450 nm wavelength of 0.25 wt % solutions of Applicants' hydrogel I;
  • FIG. 2 shows a LCST of 5 wt % solutions of poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA, 2.3 mol % JAAm determined by cloud point as a function of time, wherein NIPAAm is N-isopropylacrylamide, DBLA is (R)-α-Acryloyloxy-β,β-dimethyl-γ-butyrolactone, and JAAm is Jeffamine M-1000 acrylamide;
  • FIG. 3 shows deswelling of a control polymer 30 wt % poly(NIPAAm-co-DBLA) at various times after gelation in excess PBS (pH 7.4) at 37° C.;
  • FIG. 4 shows swelling and degradation behavior of Applicants' terpolymer I based gel comprising poly(NIPAAm-co-DBLA-co-JAAm) in excess PBS (pH 7.4);
  • FIG. 5 graphically illustrates the mass of vancomycin released from 1 mL gels loaded with 50,000 micrograms vancomycin HCl and 1 mL bone cement samples loaded with 142,000 micrograms vancomycin HCl;
  • FIG. 6 graphically illustrates an amount of vancomycin released as a fraction of the drug loaded;
  • FIG. 7 graphically illustrates a fraction of loaded vancomycin released from Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) gels loaded with either 50 mg or 5 mg per gel; and
  • FIG. 8 summarizes Applicants' method to deliver a medicament to an injection site within the body of an animal, including a human.
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • Referring to the foregoing paragraphs, this invention is described in preferred embodiments in the following description with reference to the FIGs., in which like numerals represent the same or similar elements. Reference throughout this specification to “one embodiment,” “an embodiment,” or similar language means that a particular feature, structure, or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, appearances of the phrases “in one embodiment,” “in an embodiment,” and similar language throughout this specification may, but do not necessarily, all refer to the same embodiment.
  • The described features, structures, or characteristics of the invention may be combined in any suitable manner in one or more embodiments. In the above description, numerous specific details are recited to provide a thorough understanding of embodiments of the invention. One skilled in the relevant art will recognize, however, that the invention may be practiced without one or more of the specific details, or with other methods, components, materials, and so forth. In other instances, well-known structures, materials, or operations are not shown or described in detail to avoid obscuring aspects of the invention.
  • In situ forming biomaterials which transition from liquid to solid under physiological conditions are promising for a variety of medical applications, such as tissue repair, embolization, and drug delivery. Administering these materials by injection provides reduced infection risk, scar formation, and treatment cost relative to implantation. In situ forming materials can provide local, controlled delivery of therapeutics directly to the desired area with reduced risk of side effects compared with commonly performed systemic delivery. In addition to ease of administration and delivery properties, materials that form in situ are ideal for use in spaces that are irregular or difficult to access with a solid implant, since the shape of an in situ forming implant is defined by its local environment.
  • Aqueous solutions of temperature-responsive polymers based on N-isopropylacrylamide (NIPAAm) are particularly useful for drug delivery applications due to “outside-in” gelation which rapidly takes place in physiological conditions without low molecular weight additives or chemical reaction. NIPAAm-based polymers have a sharp lower critical solution temperature (LCST) which is relatively independent of molecular weight and concentration.
  • As a solution of poly(NIPAAm) is heated above its LCST (near 32° C.), polymer-polymer interactions between the hydrophobic isopropyl groups become more favorable than the polymer-water interactions with NIPAAm side group. Upon this transition, the polymer chains collapse to form a semisolid hydrogel at sufficiently high polymer concentrations for chain entanglement. Various properties such as polymer LCST, chemical reactivity, sensitivity to various stimuli (pH, enzymes), or gel swelling can be controlled by incorporating small molar fractions (<10%) of comonomers with NIPAAm in order to provide a material for a particular application. For example, hydrophobic comonomers decrease the LCST while hydrophilic comonomers increase the LCST.
  • Effective controlled drug delivery from poly(NIPAAm) is limited primarily by two factors: its lack of degradability and the high degree of burst release from the gels within hours after gelation. Poly(NIPAAm)-based gels crosslinked with enzyme-degradable peptides or natural biopolymers have been reported i a the literature. While these gels may weaken or erode over time, the insoluble poly(NIPAAm) portions of the material will remain permanently at the injection site unless the LCST increases above body temperature, increasing the risk of chronic inflammatory response.
  • Fast solute release observed from poly(NIPAAm) homopolymer gels is thought to be due to changes in swelling and heterogeneity in the water distribution of the hydrophobic polymer-rich phase. A variety of drugs (including proteins) tend to release from NIPAAm-based hydrogels within one day.
  • Applicants have found that chemically inert hydrophilic grafts of JEFFAMINE M-1000 covalently bound to a poly(NIPAAm) backbone yield physical gels which provide slow release and controlled gel swelling with minimal LCST effects. JEFFAMINE M-1000 is a random copolymer of ethylene oxide (EO) and propylene oxide (PO) in a 19:3 EO:PO ratio with one terminal amine and one terminal methoxy group. Applicants' hydrogel represents a more clinically viable class of injectable materials for drug delivery by imparting swelling and drug release control via JEFFAMINE M-1000 in a resorbable polymer.
  • Applicants' have further discovered that use of substituted acrylate comonomer (R)-α-acryloyloxy-β,β-dimethyl-γ-butyrolactone (DBLA) increases the LCST of Applicants' hydrogel I as a result of a ring-opening mechanism which converts hydrogel I into a modified hydrogel and a provides an increased LCST without the loss of low molecular weight byproducts. In this design, only a small molar fraction (<10%) of side groups on the polymer become ionized at physiological pH to cause dissolution of the polymer, which may provide improved biocompatibility over using degradable polyesters such as poly(lactic acid).
  • One application where Applicants' in situ forming materials can meet clinical need is in the prevention and management of orthopaedic infections. For example, total hip arthroplasty (THA) is a very common procedure, with more than 200,000 performed in the US each year. By 2030, this number is estimated to increase to 572,000. The incidence of prosthetic joint infection in total hip and total knee arthroplasties is approximately 1.5-2.5% for primary interventions and higher for revision procedures performed secondary to infection. While the incidence of infection is relatively low, the high number of operations overall combined with the high cost of treating an infected arthroplasty (over $50,000 in Year 1995 dollars) make this an area of great need. As the majority of primary THAs are now cementless (where the hip prosthesis is fit directly into the intramedullary space), there is no opportunity for delivery of antibiotics to the surface of the prosthesis via antibiotic-loaded acrylic bone cement. In addition to its role as a fixative and structural element, bone cement is commonly used in orthopaedic surgery as a carrier for antibiotics and antifungal agents, despite the fact that only a small fraction of the entrapped drug is released, and the elution rate is very low after the first day.
  • It would be of great benefit to be able to locally deliver drugs using a resorbable material around orthopaedic implants during or even after these common procedures. Toward this goal, Applicants have developed a polymer formulation based on NIPAAm, JAAm, and DBLA which has properties suitable for an in situ forming hydrogel for orthopaedic applications—specifically, the following: 1) efficient and sustained antibiotic release compared to bone cement, 2) soft yet cohesive physical structure so it can act as a “filler” capable of fitting into a variety of small or irregular locations where cement may not be able to access, and 3) degradability. The method of use for this class of materials is novel in itself because there are currently no such soft and degradable vehicles for drug delivery available on the market. These materials provide an advantage for some medical conditions limited to small areas of the body because they can be easily applied/injected and they then will thicken/gel and entirely fill the irregular space into which they are placed.
  • R1 and R2 are independently selected from the group consisting of H, alkyl, phenyl, benzyl, 2-cyanoprop-2-yl, 4-cyanopentanoic acid-4-ylethyl-2-propionate, sulfate, 2-[2-methoxypropan-2-yl)oxy]propan-2-yl, and a dithioester derived from a RAFT chain transfer agent such as 4-cyano-4-(ethylsulfanylthiocarbonyl)sulfanylpentanoic acid.
  • In certain embodiments, pendent group R8 comprises a linkage with the polymer backbone selected from the group consisting of an ester, amide, thioamide, thiourea, anhydride, thioester, thiourea, and alkyl. In certain embodiments, pendent group R8 comprises at least one ester or anhydride.
  • In certain embodiments, linkage R8 is selected from the group consisting of ester, amide, thioamide, thiourea, anhydride, thioester, thiourea, and alkyl. In certain embodiments, either linkage R8 or pendent group R6 comprises at least one ester or anhydride.
  • In certain embodiments, R6 is selected from the group consisting of alkyl phenyl, polyesters, and lactones. In certain embodiments, R6 comprises a substituted butyrolactone.
  • Figure US20140256617A1-20140911-C00001
  • In certain embodiments, R7 comprises an amide linkage. In certain embodiments, R7 comprises a thioamide linkage. In certain embodiments, R7 comprises a urea linkage. In certain embodiments. R7 comprises a thiourea linkage. In certain embodiments, R7 comprises an ester linkage. In certain embodiments, R7 comprises an anhydride linkage.
  • In certain embodiments, the water soluble polymer comprises a polyether. In certain embodiments, the water soluble polymer comprises polyether II formed by ring opening polymerization of ethylene oxide, wherein R9 is selected from the group consisting of H, methyl, methoxy, and hydroxyl. In certain embodiments, n is between about 5 and about 2500.
  • Figure US20140256617A1-20140911-C00002
  • In certain embodiments, the water soluble polymer comprises polyether III formed by ring opening polymerization of propylene oxide. In certain embodiments, n is between about 15 and about 250.
  • Figure US20140256617A1-20140911-C00003
  • In certain embodiments, the water soluble polymer comprises polyether IV formed by co-polymerization of ethylene oxide and propylene oxide. In certain embodiments r is between about 5 and about 2500, and p is between about 1 and about 1000.
  • Figure US20140256617A1-20140911-C00004
  • In certain embodiments, the water soluble polymer comprises polyether V formed by ring opening polymerization of tetrahydrofuran. In certain embodiments, n is between about 10 and about 50.
  • Figure US20140256617A1-20140911-C00005
  • In certain embodiments, the water soluble polymer comprises a water-soluble polymer of one or more of the following: vinyl alcohol, acrylic acid, methacrylic acid, 2-hydroxyethyl methacrylate, N-2 hydroxypropylmethacrylamide, vinylpyrrolidone, or a monosaccharide.
  • In certain embodiments, graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XI using monomer XII.
  • Figure US20140256617A1-20140911-C00006
  • In certain embodiments, graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XIII using monomer XIV.
  • Figure US20140256617A1-20140911-C00007
  • In certain embodiments, graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XV using monomer XVI.
  • Figure US20140256617A1-20140911-C00008
  • In certain embodiments, graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XVII using monomer XVIII.
  • Figure US20140256617A1-20140911-C00009
  • In certain embodiments, graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XIX using monomer XX.
  • Figure US20140256617A1-20140911-C00010
  • In certain embodiments, graft terpolymer I comprises a plurality of repeat units VI formed using monomer VII, a plurality of repeat units IX formed using monomer X, and a plurality of repeat units XXI using monomer XXII.
  • Figure US20140256617A1-20140911-C00011
  • In certain embodiments, a is between about 10 and about 1000, b is between about 1 and about 250, and c is between about 1 and about 500.
  • Terpolymer I can be synthesized via a number of different procedures. For example, Applicants' graft terpolymer I can be prepared by free radical polymerization.
  • Graft terpolymer I can also be prepared by reversible addition-fragmention chain transfer (“RAFT”) polymerization. Those skilled in the art will appreciate that a RAFT polymerization can be performed by adding a quantity of a RAFT agent (thiocarbonylthio compounds) to a conventional free radical polymerization. Usually the same monomers, initiators, solvents and temperatures can be used. Because of the low concentration of the RAFT agent in the system, the concentration of the initiator is usually lower than in conventional radical polymerization. Radical initiators such as azobisisobutyronitrile (AIBN) and 4,4′-Azobis(4-cyanovaleric acid) (ACVA) which is also called 4,4′-Azobis(4-cyanopentanoic acid) are widely used as the initiator in RAFT. RAFT polymerization is known for its compatibility with a wide range of monomers, including for example acrylates and acrylamides.
  • Graft terpolymer I, as either a random copolymer or a block copolymer, can also be prepared by atom transfer radical polymerization (“ATRP”). Controlled polymerization of N-isopropylacrylamide (NIPAAM) by atom (ATRP) can be effected using ethyl 2-chloropropionate (ECP) as initiator and CuCl/tris(2-dimethylaminoethyl)amine (Me6TREN) as a catalytic system. The living character of the polymerization allows preparation of block copolymers.
  • The following examples are presented to further illustrate to persons skilled in the art how to make and use the invention. These examples are not intended as a limitation, however, upon the scope of the invention.
  • EXAMPLES
  • An embodiment of Applicant's hydrogel I comprises a random terpolymer formed using three monomers, namely N-isopropylacrylamide (NIPAAm), JEFFAMINE M-1000 acrylamide (JAAm), and acryloyloxy-β,β-dimethyl-γ-butyrolactone (DBLA). JAAm is synthesized prior to synthesis of the entire polymer. All materials were reagent grade and can be obtained from Sigma-Aldrich unless otherwise noted.
  • NIPAAm monomer was recrystallized from hexane. Azobisisobutyronitrile (AIBN) was recrystallized from methanol. HPLC grade tetrahydrofuran (THF) was used for low molecular weight polymerizations and as the mobile phase for molecular weight and polydispersity determination. JEFFAMINE M-1000 polyetheramine was donated by Huntsman Corporation (The Woodlands, Tex., USA).
  • JEFFAMINE M-1000 acrylamide (JAAm) was synthesized from JEFFAMINE M-1000 polyetheramine by acryloylation with acryloyl chloride. JEFFAMINE M-1000 (20 g, 20 mmol) was dissolved at 10 w/v % in dichloromethane (DCM) along with triethylamine (3.3 mL, 24 mmol) and maintained at 0° C. under nitrogen atmosphere. Acryloyl chloride (1.95 mL, 24 mmol) was then added dropwise into the solution under stirring and the reaction was allowed to proceed for 1 hour at 0° C. and then at room temperature for at least 5 hours under nitrogen atmosphere. Following the reaction, DCM was evaporated and the remaining residue was dissolved in 0.1 N sodium bicarbonate (200 mL). The product was extracted into DCM and the organic phase purified by column chromatography. After evaporation of DCM under mild heating at reduced pressure, JAAm was obtained as a viscous clear to light yellow liquid. JAAm was solidified by cooling on ice, vacuum dried overnight, then collected and stored at −20° C.
  • Hydrogel I prepared using NIPAAm, DBL, and JAAm were synthesized by radical polymerization with 80% dioxane/20% THF as the solvent, but other solvents may be used, including methanol, ethanol, water, benzene, acetone, isopropanol, pure dioxane, pure tetrahydrofuran, or others. All three monomers (NIPAAm, DBLA, and JAAm) were dissolved in the solvent. Monomer solution (10 w/v %) was bubbled with nitrogen for at least 10 minutes prior to addition of initiator to reduce dissolved oxygen. After this time, AIBN (initiator) was added (˜0.007 mol AIBN/mol of total monomer) and the reaction allowed to proceed for 24 hr at 65° C. under a slight positive pressure of nitrogen.
  • Applicants' hydrogel was collected by precipitation into 10 to 15-fold excess of a solvent in which the polymer is not soluble, such as chilled diethyl ether or hexane. Following precipitation, the product was collected by filtration and dried overnight under vacuum. The product was then dissolved in deionized water and can optionally be dialyzed against 3500 MWCO at 4° C. for 20 hours with the excess water replaced 3 times in order to purify the material, removing any low molecular weight impurities. Materials were then lyophilized and can be stored at −20° C. under nitrogen atmosphere until use.
  • To prepare a solution for antibiotic delivery, the materials can be sterilized (such as by ethylene oxide gas sterilization) and then dissolved in 150 mM phosphate buffered saline (pH 7.4). After dissolution of the polymer, drug powder (finely ground) is added directly to the polymer solution and vortex mixed to distribute the drug evenly throughout the solution. The solutions can be stored in a freezer until use and thawed at the time of use. The solution then can be injected, brushed, poured, sprayed, or otherwise administered as a liquid at the desired site. The solution can also be allowed to warm to above its initial LCST and then implanted as a soft hydrogel at the desired site. Examples of desired sites for delivery of this material might include the surface of a joint prosthesis, the intramedullary canal before implantation of a prosthesis, an infected area, or any location deemed to have a risk of future infection. An example formulation might be a 30 wt % polymer solution with approximately 50 mg vancomycin hydrochloride loaded per mL of polymer solution.
  • Hydrogels are defined as three-dimensional (3D), water-swollen polymer networks formed as a result of physical or chemical cross-linking. Because of their high water content and mechanical resemblance to natural tissues, hydrogels show promising biocompatibility and potential for medical applications. Injectable hydrogel formulations are especially attractive due to their minimally invasive delivery procedure, providing reduced healing time, reduced scarring, decreased risk of infection, and ease of delivery compared with surgically implanted materials.
  • Injection of viscoelastic fluids, most commonly lightly pre-crosslinked hyaluronic acid, into joints has been performed for over 20 years in the treatment of osteoarthritis. These viscosupplementation treatments are said to derive their effectiveness in part from the high viscosity of the injected polymer solution.
  • Polymeric hydrogels which undergo crosslinking during or after administration to a joint space may represent an improvement because such hydrogels undergoes an increase in viscosity within the joint space following administration. Thus the hydrogel precursor may be administered as a relatively less viscous liquid, then forming a much more viscous or viscoelastic gel within a timeframe of less than one hour. Additionally, this higher viscosity imparted by crosslinking in situ could lead to prolonged residence time in the joint and therefore a longer treatment duration achieved with a single administration.
  • Some in situ crosslinking hydrogels (including the Applicants' terpolymer hydrogels) are viscoelastic, providing properties that can allow for the gel to remain intact within the joint by yielding during joint motion, conforming to the shape of the joint space in a way that is analogous to a highly viscous liquid. Because of their rheological properties, viscoelastic hydrogels are especially well-suited to conform to the surface of a prosthetic joint (or other surfaces present in a joint) to provide effective and sustained local concentrations of drug within the joint.
  • In certain embodiments, Applicants utilize a crosslinked hydrogel injected into a joint space. By “crosslinked hydrogel” Applicants mean a physically or chemically crosslinked polymer-containing material containing over 50 weight percent of an aqueous solvent. In certain embodiments, the gel is crosslinked prior to injection. In certain embodiments, the gel is crosslinked after injection in the joint space.
  • Applicants' method administers a hydrogel in vivo to a joint space of an animal. In certain embodiments, the hydrogel forms by physical crosslinking (for example, by heating above an LCST). In certain embodiments, the crosslinked hydrogel forms by supramolecular self-assembly. In certain embodiments, the crosslinked hydrogel forms by covalent crosslinking.
  • In certain embodiments, the crosslinked hydrogel is administered by transcutaneous injection. In certain embodiments, the crosslinked hydrogel is administered to the joint space during a surgical procedure.
  • In certain embodiments, the crosslinked hydrogel contains no medicament. In certain embodiments, the crosslinked hydrogel comprises a medicament. In certain embodiments, the medicament comprises an antimicrobial.
  • In certain embodiments, the crosslinked hydrogel is administered to the hip joint in conjunction with total hip arthroplasty. In certain embodiments, the crosslinked hydrogel is administered to the knee joint in conjunction with total knee arthroplasty. In certain embodiments, the joint space is the hip joint. In certain embodiments, the joint space is the knee joint.
  • TABLE 1
    Selected properties of degradable temperature-responsive
    polymers with and without JAAm.
    Feed ratio Composition* Molecular
    (mol %) (mol %) weight (g/mol) Initial LCST Degradation time
    Polymer DBLA JAAm DBLA JAAm Mn Mw (° C.) (37° C., pH 7.4)
    poly(NIPAAm- 7.2 5.6 0.0 13,100 32,100 18  ~20 weeks**
    co-DBLA)
    poly(NIPAAm- 7.1 4.1 4.7 2.9 13,100 28,200 27 8-10 days
    co-DBLA-co-
    JAAm)
    *Measured by 1H NMR,
    **estimated using accelerated degradation study (ISO 10993)
  • Approximately 800,000 arthroplasties overall were performed in the United States in 2003 alone. Approximately 1.5-2.5% of these fail due to infection, which causes loosening of the implant. Now that the vast majority of hip prostheses rely on cementless fixation of the femoral component, there is no clinically available material which allows for delivery of drugs at the implant surface where most infections arise. While cemented fixation is common in total knee arthroplasty, most of the implant surface is not in contact with the cement, and effective local concentrations are not achieved. Even in situations where bone cement can be used, the rate of drug release is very slow after the first 1-2 days, which may limit its effectiveness in preventing or managing infection.
  • Once a prosthetic joint is implanted and reaches body temperature, Applicants' injected hydrogel I, in solution, becomes a soft semi-solid filler which steadily delivers antibiotics directly at the surface of the implant which may do so without disrupting the tit between the bone and the prosthesis or generating wear debris from joint motion. By delivering the antibiotics directly at the implant surface, the drug concentration near the implant can be maintained at a high level in order to prevent infection, while the systemic exposure to the drug (and the corresponding side effects) can remain low.
  • The only existing product widely used clinically for either prevention or management of orthopaedic infections is poly(methyl methacrylate) (PMMA) bone cement, which is marketed and sold in a variety of formulations. The drug(s) (antibiotics or antifungals) are finely ground and mixed into the cement before implantation. Local delivery of drugs (drugs administered near the implant site) is required in order to provide high drug concentration at the desired site with low concentration elsewhere in the body. Systemic administration (such as by intravenous infusion) does not effectively target the implant surface and can cause toxicity and side effects at a dose that would be therapeutic at the implant surface.
  • There are four major problems with using bone cement for antibiotic delivery. First, the drug release is inefficient, meaning that a low fraction (˜10% for prophylactic “low” doses and ˜40% for “high” doses) of the drug mixed into the cement is ever released. The remaining drug gets trapped deep inside the cement. This is wasteful because most of the drug purchased is never released from the cement. As a result, most of the entrapped drug is not released in a concentration sufficient to provide therapeutic benefit.
  • Second, drug release is not sustained. Drug must be maintained at the site of infection at as a high concentration for as long of a time as possible in order to destroy a bacterial biofilm, which can require 1000 times as much drug concentration to inhibit growth as an individual planktonic bacterium. Most of the drug release from cement occurs within the first 1-2 days, with very low release thereafter.
  • Third, bone cement is not degradable. While bone cement is known to be biocompatible, it cannot be used with cementless hip stems and is not re-dosable without an invasive procedure.
  • Finally, bone cement is not indicated for use with many cementless hip prosthesis designs.
  • An aqueous solution of Applicants' injectable hydrogel composition I overcomes the deficiencies of prior art bone cement. Applicants' hydrogel I provides efficient drug release. Because the material eventually dissolves, all of the drug is eventually delivered and utilized. Local delivery provides the highest drug concentration directly where it is needed—at the implant surface,
  • Applicants' hydrogel provides sustained drug release. Throughout the 7-10 day period during which the material dissolves, drug is released at a relatively steady rate, which may provide improved performance at preventing or managing prosthetic joint infections.
  • Applicants' hydrogel provides local delivery to the implant surface of cementless stems without disrupting the quality of fit between the hip prosthesis and the femur.
  • Applicants' hydrogel provides degradability. Previous work by Applicants' and colleagues has shown no difference in immune response from that of native tissue following gel dissolution. The degradability of the material also allows for it to be re-dosable by transcutaneous injection in some cases.
  • FIG. 1 graphically illustrates relative absorbance of 0.25 wt % solutions of Applicants' hydrogel I comprising poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA, 2.3 mol % JAAm incubated at 37° C. in Phosphate Buffer Solution after 1 day (♦), 3 days (▪), 7 days (▴), and 15 days (). DBLA dimethyl-butyrolactone acrylate and JAAm is JEFFAMINE M-1000 acrylamide.
  • FIG. 2 shows the LCST of 5 wt % solutions of poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA, 2.3 mol % JAAm determined by cloud point as a function of time. As a comparison, the LCST for the same polymer without JAAm remains at 18° C. for the first 2 weeks. As those skilled in the art will appreciate, human body temperature is about 37° C. When the LCST of Applicants' injected hydrogel I increases to above 37° C., material injected into the body will dissolve.
  • FIG. 3 shows deswelling of a control polymer 30 wt % poly(NIPAAm-co-DBLA) at various times after gelation in excess PBS (pH 7.4) at 37° C. The control copolymer comprises no absence of JAAm. Gels began shrinking rapidly beginning on the first day. These gels dissolve in approximately 20 weeks.
  • FIG. 4 shows swelling and degradation behavior of Applicants' terpolymer I based gel comprising poly(NIPAAm-co-DBLA-co-JAAm) in excess PBS (pH 7.4). Gels retained their initial volume throughout the study and underwent bulk degradation. Gels flow when inverted after 7-10 days and dissolve completely within 2-3 weeks.
  • FIG. 5 graphically illustrates the mass of vancomycin released from 1 mL gels loaded with 50,000 micrograms vancomycin HCl and 1 mL bone cement samples loaded with 142,000 micrograms vancomycin HCl. Release is prolonged and increased from Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) as compared to clinically used bone cement.
  • FIG. 6 graphically illustrates an amount of vancomycin released as a fraction of the drug loaded. Most of the drug that is loaded into bone cement is not released whereas Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) release nearly all of the drug loaded into them within 8 days.
  • FIG. 7 graphically illustrates a fraction of loaded vancomycin released from Applicants' injectable and degradable terpolymer I based gels comprising poly(NIPAAm-co-DBLA-co-JAAm) gels loaded with either 50 mg or 5 mg per gel. Some vancomycin is insoluble when loaded into the polymer solution at 50 mg/mL. The similarity between these curves indicates that the polymer, as opposed to the drug solubility, is primarily responsible for controlling the rate of drug release.
  • FIG. 8 summarizes Applicants' method to deliver a medicament to an injection site within the body of an animal, including a human. In certain embodiments, the injection site comprises the surface of an bone implant. Referring now to FIG. 8, in step 810 the method provides a medicament and a first hydrogel comprising a first LCST less than the body temperature of the subject animal. Those skilled in the art will appreciate, that body temperature for a human is about 37° C. In these human injection embodiments, the first hydrogel of step 810 comprises a LCST less than about 37° C.
  • By medicament, Applicants mean a material selected from the group consisting of a Nucleic acid, a Protein (including growth factors, bone morphogenetic proteins), a Polypeptide, a Contrast agent for imaging an Anesthetic, an Antineoplastic agent, an Antifungal, an Anti-inflammatory drug (steroids, non-steroidal anti-inflammatory drugs (NSAIDs), and an Antibiotic. In certain embodiments, the Antibiotic comprises one or more of Aminoglycosides, including gentamicin, amikacin, and tobramycin, Cephalosporins including cefazolin, Vancomycin, and Rifampin
  • In certain embodiments, the first hydrogel of step 810 comprises a polymeric material comprising a backbone formed from polymerization of one or more substituted acrylamides, one or more monomers containing one or more hydrolyzable linkages, and a monomer comprising a water soluble polymer moiety attached thereto.
  • In certain embodiments, the first hydrogel of block 810 comprises Applicants' hydrogel I. In certain embodiments, the first hydrogel of block 810 comprises Applicant's hydrogel formed from N-isopropylacrylamide, JEFFAMINE M-1000 acrylamide (JAAm), and acryloyloxy-β,β-dimethyl-γ-butyrolactone. In certain embodiments, the first hydrogel of step 810 comprises Applicant's hydrogel comprising poly(NIPAAm-co-DBLA-co-JAAm) with 5.9 mol % DBLA and 2.3 mol % JAAm.
  • In block 820, an aqueous solution of the medicament and the first hydrogel of block 810 is injected into a selected animal at a temperature less than the first LCST. In certain embodiments, the injection site comprises a tissue space wherein a subsequently formed gel will substantially completely fill that tissue space. In certain embodiments, the first hydrogel of block 810 is utilized in conjunction with implantation of an artificial joint. In certain of these embodiments, the injection of block 820 is performed after implantation such that the injected hydrogel is disposed adjacent a surface of the implanted artificial joint.
  • In certain embodiments, the hydrogel of block 810 is coated onto a surface of an artificial joint prior to implantation. In these embodiments, the “injection” of block 820 comprises implantation of the artificial joint comprising a surface coated with the first hydrogel of block 810.
  • In block 830, the first hydrogel of block 810 injected into the body of an animal in block 820 is warmed in vivo to a temperature greater than the first LCST. In certain embodiments, the warming of block 830 is performed by the body heat of the animal. In certain embodiments, the warming of block 830 is performed by disposing a heating object, such as for example and without limitation, a heat lamp, a heating pad, hot compress, and the like, directed in near proximity to the injection site.
  • In block 840, the first hydrogel of block 810 injected into the body of an animal in block 820 and warmed in vivo to a temperature greater than the first LCST in block 830, forms in vivo a water-insoluble gel. In certain embodiments, the water-insoluble gel of block 840 is formed in, and substantially fills, a tissue space. In certain embodiments, the water-insoluble gel of block 840 is disposed on, and in near vicinity to, a surface of a joint implant.
  • In block 850, the water-insoluble gel of block 840 releases the medicament of block 810 into tissues adjacent the injection site of block 820. In certain embodiments, the release of medicament is sustained over time. In certain embodiments, the aggregate amount of medicament released from the water-insoluble gel of block 840 into the injection site of block 820 plotted on a Y axis of a graph against time plotted on an X axis of the graph over time can be approximately modeled by a linear equation of the type y=mx+b, wherein m is slope of the straight line and b is the intercept of the straight line with the Y axis. As a general matter, the intercept is 0.
  • In other embodiments, the release is approximately proportional to the square root of time over the first 60% of release, with a slower rate of release thereafter.
  • In block 860, the first hydrogel of block 810 which was injected in block 820 is chemically modified to form a second hydrogel having a second LCST, wherein the second LCST is greater than body temperature. For example, in certain embodiments, the pendent ester moieties of the plurality of repeat units IV are hydrolyzed in vivo to convert first terpolymer I injected in block 820 and gelled in block 840 to a second terpolymer XXII, wherein R9 comprises a pendent hydroxyl group or a pendent carboxylic acid group resulting from a hydrolysis of an acid moiety or an anhydride moiety, respectively, in pendent group R8.
  • Figure US20140256617A1-20140911-C00012
  • In certain embodiments wherein R6 comprises a substituted butyrolactone moiety and R8 is an ester linkage, the pendent substituted butyrolactone moieties of the plurality of repeat units IV are hydrolyzed in vivo to convert first terpolymer I injected in block 820 and gelled in block 840 to a second terpolymer XXIII, wherein R9 comprises a carboxylic acid moiety.
  • Figure US20140256617A1-20140911-C00013
  • In certain embodiments, in block 860 the first hydrogel of block 810 is converted into a second hydrogel XXII/XXIII that is water soluble at body temperature.
  • In block 870, the water soluble second hydrogel dissolves in body fluids.
  • In block 880, the second hydrogel of step 870 is excreted from the body.
  • Using the method of FIG. 8, a first hydrogel having a LCST less than body temperature and a medicament, in aqueous solution, are injected into the body of an animal. The first hydrogel is warmed to a temperature greater than its LCST, and a water-insoluble gel is formed. The medicament is released over time from the water-insoluble gel. Thereafter, the first hydrogel is converted in vivo into a second hydrogel having a LCST greater than body temperature. This second hydrogel is water soluble, and is carried by the animal's circulatory system to its kidneys. The second hydrogel is then excreted from the body.
  • While the preferred embodiments of the present invention have been illustrated in detail, it should be apparent that modifications and adaptations to those embodiments may occur to one skilled in the art without departing from the scope of the present invention as set forth in the following claims.

Claims (41)

1. A hydrogel, comprising:
a polymer backbone comprising a plurality of repeat units, wherein one or more of the plurality of repeat units comprises a pendent water soluble polymer attached thereto;
wherein:
an aqueous solution of a polymer comprises a first lower critical solution temperature (“LCST”);
said solution forms said hydrogel when heated above that first LCST;
said hydrogel is configured to be converted in vivo into a modified hydrogel;
an aqueous solution of said modified hydrogel comprises a second lower critical solution temperature (“LCST”);
said second LCST is greater than said first LCST.
2. (canceled)
3. The hydrogel of claim 1, wherein said polymer backbone comprises a terpolymer.
4. The hydrogel of claim 3, wherein said terpolymer comprises a second plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00014
wherein R3 and R4 are each independently selected from the group consisting of H and alkyl.
5. The hydrogel of claim 4, wherein said terpolymer comprises a third plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00015
wherein linkage R8 is selected from the group consisting of ester, amide, thioamide, thiourea, anhydride, thioester, thiourea, and alkyl;
wherein R6 is selected from the group consisting of alkyl, phenyl, polyesters, and lactones; and
wherein either R8 or R6 comprises at least one ester or anhydride.
6. The hydrogel of claim 5, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00016
7. The hydrogel of claim 5, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00017
8. The hydrogel of claim 5, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00018
9. The hydrogel of claim 5, wherein said terpolymer further comprises a first plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00019
10. The hydrogel of claim 5, wherein said terpolymer further comprises a first plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00020
11. The hydrogel of claim 5, wherein said terpolymer further comprises a first plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00021
12. The hydrogel of claim 5, wherein the water soluble polymer comprises a polyether.
13. The hydrogel of claim 12, wherein the polyether comprises a fourth plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00022
14. The hydrogel of claim 12, wherein the polyether comprises a fourth plurality of repeat units having a structure
Figure US20140256617A1-20140911-C00023
15. A method to deliver a medicament in vivo, comprising:
providing a medicament and a hydrogel, wherein said hydrogel comprises a polymer with a first lower critical solution temperature (“LCST”); wherein one or more of the plurality of repeat units of said polymer comprises a pendent water soluble polymer attached thereto by a linkage selected from the group consisting of amide, thioamide, urea, thiourea, ester, and anhydride;
administering at an administration site said medicament and said hydrogel into an animal, wherein said animal comprises a body temperature
wherein said body temperature is greater than said LCST;
releasing in vivo said medicament from said hydrogel;
converting in vivo said hydrogel into a modified polymer, wherein an aqueous solution of said modified polymer comprises a second lower critical solution temperature (“LCST”), and wherein said second LCST is greater than said body temperature;
excreting said modified polymer by said animal.
16. The method of claim 15, wherein said polymer comprises a first plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00024
wherein R7 is selected from the group consisting of an amide linkage, a thioamide linkage, a urea linkage, a thiourea linkage, an anhydride linkage, and an ester linkage.
17. The method of claim 16, wherein said polymer backbone comprises a terpolymer.
18. The method of claim 17, wherein said terpolymer comprises a second plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00025
wherein R3 and R4 are each independently selected from the group consisting of H, methyl, ethyl, and phenyl.
19. The method of claim 18, wherein said terpolymer comprises a third plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00026
wherein linkage R8 is selected from the group consisting of ester, amide, thioamide, thiourea, anhydride, thioester, thiourea, and alkyl;
wherein R6 is selected from the group consisting of alkyl, phenyl, polyesters, and lactones; and
wherein either R8 or R6 comprises at least one ester or anhydride.
20. The method of claim 19, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00027
21. The method of claim 19, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00028
22. The method of claim 19, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00029
23. The method of claim 19, wherein said first plurality of repeat units comprise a structure:
Figure US20140256617A1-20140911-C00030
24. The method of claim 19, wherein said terpolymer further comprises a first plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00031
25. The method of claim 19, wherein said terpolymer further comprises a first plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00032
26. The method of claim 16, wherein the water soluble polymer comprises a polyether.
27. The hydrogel of claim 26, wherein the polyether comprises a fourth plurality of repeat units having a structure:
Figure US20140256617A1-20140911-C00033
28. The hydrogel of claim 26, wherein the polyether comprises a fourth plurality of repeat units having a structure
Figure US20140256617A1-20140911-C00034
29. A method to administer a hydrogel in vivo to an animal to an administration site, including a human, wherein:
said hydrogel comprises a polymer;
said hydrogel contains over 50% by weight of an aqueous solvent;
said polymer forms physical crosslinks during the use of said hydrogel in response to an environmental condition.
30-41. (canceled)
42. The method of claim 29, wherein said polymer forms physical crosslinks in response to a change in temperature.
43. The method of claim 29, wherein said administration site comprises a joint space.
44. The method of claim 29, wherein said administration site comprises a knee joint.
45. The method of claim 29, wherein said administration site comprises a hip joint.
46. The method of claim 29, wherein said administration site comprises a surface of an implanted medical device.
47. The method of claim 34, wherein said implanted medical device comprises a prosthetic joint.
48. The method of claim 46, wherein said implanted medical device is used in orthopaedic surgery.
49. The method of claim 46, wherein said hydrogel is administered to said implanted medical device after implantation of said implanted medical device
50. The method of claim 29, wherein said hydrogel comprises one or more medicaments.
51. The method of claim 29, wherein said hydrogel contains no medicament.
52. The method of claim 29, wherein said hydrogel comprises an antimicrobial.
US14/351,549 2011-10-12 2012-10-12 In Situ Forming Hydrogel and Method Using Same Abandoned US20140256617A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/351,549 US20140256617A1 (en) 2011-10-12 2012-10-12 In Situ Forming Hydrogel and Method Using Same

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161546402P 2011-10-12 2011-10-12
PCT/US2012/060124 WO2013056170A1 (en) 2011-10-12 2012-10-12 In situ forming hydrogel and method using same
US14/351,549 US20140256617A1 (en) 2011-10-12 2012-10-12 In Situ Forming Hydrogel and Method Using Same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/060124 A-371-Of-International WO2013056170A1 (en) 2011-10-12 2012-10-12 In situ forming hydrogel and method using same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/840,621 Continuation US10780145B2 (en) 2011-10-12 2017-12-13 In situ forming hydrogel and method using same

Publications (1)

Publication Number Publication Date
US20140256617A1 true US20140256617A1 (en) 2014-09-11

Family

ID=48082544

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/351,549 Abandoned US20140256617A1 (en) 2011-10-12 2012-10-12 In Situ Forming Hydrogel and Method Using Same
US15/840,621 Active 2032-12-13 US10780145B2 (en) 2011-10-12 2017-12-13 In situ forming hydrogel and method using same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/840,621 Active 2032-12-13 US10780145B2 (en) 2011-10-12 2017-12-13 In situ forming hydrogel and method using same

Country Status (4)

Country Link
US (2) US20140256617A1 (en)
EP (1) EP2765991A4 (en)
CA (1) CA2852068A1 (en)
WO (1) WO2013056170A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10265439B2 (en) 2015-09-03 2019-04-23 Arizona Board Of Regents On Behalf Of Arizona State University Injectable cell-laden biohybrid hydrogels for cardiac regeneration and related applications
US10624865B2 (en) 2013-03-14 2020-04-21 Pathak Holdings Llc Methods, compositions, and devices for drug/live cell microarrays
CN114712303A (en) * 2016-11-16 2022-07-08 佩尔西卡制药有限公司 Antibiotic formulations for lower back pain

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11173210B2 (en) 2017-10-24 2021-11-16 Sonoran Biosciences, Inc. Temperature-responsive degradable hydrogels
EP3700589A4 (en) * 2017-10-24 2021-07-28 Sonoran Biosciences, Inc. Temperature-responsive degradable hydrogels

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7425581B2 (en) * 1999-07-30 2008-09-16 Universiteit Utrecht Temperature sensitive polymers
US7718193B2 (en) * 2006-03-16 2010-05-18 University Of Washington Temperature- and pH-responsive polymer compositions
WO2008045904A2 (en) * 2006-10-10 2008-04-17 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Thermoresponsive, biodegradable, elastomeric material

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Cui et. al. (Biomacromolecules (2007) 8:1280-1286). *
Lee et. al. (Polymer International (2005) 54:418-422). *
Li et. al. (Biomacromolecules (2009) 10:3306-3316). *
Yoshioka et. al. (Journal of Macromolecular Science, Part A: Pure and Applied Chemistry (1994) A31(1):113-120). *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10624865B2 (en) 2013-03-14 2020-04-21 Pathak Holdings Llc Methods, compositions, and devices for drug/live cell microarrays
US11045433B2 (en) 2013-03-14 2021-06-29 Pathak Holdings Llc Method of making an in situ sustained biodegradable drug delivery implant by filling an artificial tissue cavity
US11969398B2 (en) 2013-03-14 2024-04-30 Pathak Holdings Llc Methods, compositions, and devices for making sustained biodegradable implants in tissue
US10265439B2 (en) 2015-09-03 2019-04-23 Arizona Board Of Regents On Behalf Of Arizona State University Injectable cell-laden biohybrid hydrogels for cardiac regeneration and related applications
CN114712303A (en) * 2016-11-16 2022-07-08 佩尔西卡制药有限公司 Antibiotic formulations for lower back pain

Also Published As

Publication number Publication date
EP2765991A4 (en) 2015-06-24
CA2852068A1 (en) 2013-04-18
EP2765991A1 (en) 2014-08-20
US10780145B2 (en) 2020-09-22
WO2013056170A1 (en) 2013-04-18
US20180153956A1 (en) 2018-06-07

Similar Documents

Publication Publication Date Title
US10780145B2 (en) In situ forming hydrogel and method using same
Klouda Thermoresponsive hydrogels in biomedical applications: A seven-year update
US11191842B2 (en) Injectable thermoresponsive polyelectrolytes
Moon et al. Temperature-responsive compounds as in situ gelling biomedical materials
US10064948B2 (en) Implantable bio-resorbable polymer charged with fragile macromolecules
US20100285094A1 (en) Polymeric compositions and methods of making and using thereof
US20080187568A1 (en) Polymerization with precipitation of proteins for elution in physiological solution
US20050164980A1 (en) Hyaluronic acid modification product
US8747870B2 (en) Polymeric compositions and methods of making and using thereof
US20040185086A1 (en) Polysaccharide-based polymerizable hydrogels
EP2427226B1 (en) Implant filling material and method
US20150099808A1 (en) Amphiphilic copolymers and compositions containing such polymers
Das et al. Recent advances in hydrogels for biomedical applications
Raj et al. Oral insulin–a perspective
JP2004506688A (en) Treatment of injuries and other indications
Olteanu Applications of functionalized chitosan
Overstreet Temperature-responsive hydrogels with controlled water content and their development toward drug delivery and embolization applications
US20090081260A1 (en) Naturally resorbable polyacrylamide gel, production process and uses
US20060241754A1 (en) Implant filling material and method
Kumar et al. Polymer implants for gene and drug delivery
Dipen et al. TREND OF INJECTABLE HYDROGEL IN FORMULATION AND RESERACH.

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARIZONA BOARD OF REGENTS, A BODY CORPORATE OF THE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OVERSTREET, DEREK;VERNON, BRENT;MCLEMORE, RYAN;AND OTHERS;SIGNING DATES FROM 20121129 TO 20121130;REEL/FRAME:032661/0222

Owner name: BANNER HEALTH, ARIZONA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OVERSTREET, DEREK;VERNON, BRENT;MCLEMORE, RYAN;AND OTHERS;SIGNING DATES FROM 20121129 TO 20121130;REEL/FRAME:032661/0222

STCB Information on status: application discontinuation

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