US20190076373A1 - Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration - Google Patents

Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration Download PDF

Info

Publication number
US20190076373A1
US20190076373A1 US16/033,025 US201816033025A US2019076373A1 US 20190076373 A1 US20190076373 A1 US 20190076373A1 US 201816033025 A US201816033025 A US 201816033025A US 2019076373 A1 US2019076373 A1 US 2019076373A1
Authority
US
United States
Prior art keywords
cryogel
polymer composition
composition
cell
cells
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
US16/033,025
Inventor
Sidi A. Bencherif
David J. Mooney
David Edwards
Roger Warren Sands
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.)
Harvard College
Original Assignee
Harvard College
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 Harvard College filed Critical Harvard College
Priority to US16/033,025 priority Critical patent/US20190076373A1/en
Assigned to PRESIDENT AND FELLOWS OF HARVARD COLLEGE reassignment PRESIDENT AND FELLOWS OF HARVARD COLLEGE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EDWARDS, DAVID A., SANDS, ROGER W., BENCHERIF, SIDI A., MOONEY, DAVID J.
Publication of US20190076373A1 publication Critical patent/US20190076373A1/en
Priority to US18/095,488 priority patent/US20230404936A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7007Drug-containing films, membranes or sheets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/14Macromolecular materials
    • A61L27/20Polysaccharides
    • 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
    • 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
    • A61L27/56Porous materials, e.g. foams or sponges
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L5/00Compositions of polysaccharides or of their derivatives not provided for in groups C08L1/00 or C08L3/00
    • C08L5/04Alginic acid; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/64Medicinal preparations containing antigens or antibodies characterised by the architecture of the carrier-antigen complex, e.g. repetition of carrier-antigen units
    • 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
    • A61L2300/20Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials
    • A61L2300/252Polypeptides, proteins, e.g. glycoproteins, lipoproteins, cytokines
    • 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
    • A61L2300/20Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices containing or releasing organic materials
    • A61L2300/258Genetic materials, DNA, RNA, genes, vectors, e.g. plasmids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/60Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a special physical form
    • A61L2300/64Animal cells
    • 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
    • A61L2400/00Materials characterised by their function or physical properties
    • A61L2400/16Materials with shape-memory or superelastic properties
    • 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
    • A61L2430/34Materials or treatment for tissue regeneration for soft tissue reconstruction

Definitions

  • the invention relates to polymer scaffolds for drug and cell delivery systems.
  • Tissue engineering is an approach for regeneration, replacement, and improvement of the functions of damaged tissues by manipulating materials according to the specific structure or function of the desired tissues.
  • Porous and biodegradable polymer scaffolds are utilized as a structural supporting matrix or as a cell adhesive substrate for cell-based tissue engineering.
  • a major side effect of the surgical implantation of three dimensional scaffolds is the trauma created by physicians while treating patient illness.
  • current technologies for the surgical implantation of three dimensional scaffolds involve incisions that lead to patient pain, bleeding, and bruising. As such, there is a pressing need in the art to develop less invasive structured polymer scaffolds.
  • the present invention provides compositions and a minimally-invasive method of injecting preformed large macroporous polymer-based hydrogels that are loaded with cargo such as cells and/or therapeutics such as small molecule compounds, proteins/peptides (e.g., antigens to which an immune response is desired), or nucleic acids.
  • Hydrogel also called aquagel
  • Hydrogels are highly absorbent (they can contain over 99% water) natural or synthetic polymers that possess a degree of flexibility very similar to natural tissue, due to their significant water content.
  • the invention features a cell-compatible highly crosslinked hydrogel polymer composition comprising a high density of open interconnected pores, wherein the hydrogel is characterized by shape memory following deformation by compression or dehydration.
  • the hydrogel comprises polymers that are modified, e.g., sites on the polymer molecule are modified with a methacrylic acid group (methacrylate (MA)) or an acrylic acid group (acrylate).
  • MA methacrylic acid group
  • acrylate acrylic acid group
  • An exemplary modified alginate is MA-alginate (methacrylated alginate).
  • Methacrylated-alginate 50% corresponds to the degree of methacrylation of alginate. This means that every other repeat unit contains a methacrylated group.
  • the degree of methacrylation can be varied from 1% to 90%.
  • the chemical modification may reduce solubility of the polymer water-solubility.
  • Polymers can also be modified with acrylated groups instead of methacrylated groups. The product would then be referred to as an acrylated-polymer.
  • the degree of methacrylation (or acrylation) can be varied for most polymers. However, some polymers (e.g. PEG) maintain their water-solubility properties even at 100% chemical modification.
  • polymers in the hydrogel are 50-100% crosslinked (covalent bonds). The extent of crosslinking correlates with the durability of the hydrogel. Thus, a high level of crosslinking (90-100%) of the modified polymers is desirable.
  • the highly crosslinked hydrogel polymer composition is characterized by at least 50% polymer crosslinking (e.g., 75%, 80%, 85%, 90%, 95%, 98%).
  • the high level of crosslinking confers mechanical robustness to the structure.
  • the % crosslinking is generally less than 100%.
  • the composition is formed using a free radical polymerization process and a cryogelation process.
  • the cryogel comprises at least 75% pores, e.g., 80%, 85, 90%, 95%, and up to 99% pores.
  • the pores are interconnected. Interconnectivity is important to the function of the composition, as without interconnectivity, water would become trapped within the gel. Interconnectivity of the pores permits passage of water (and other compositions such as cells and compounds) in and out of the structure.
  • the composition In a fully hydrated state, the composition comprises between 90-99% water. In a compressed or dehydrated hydrogel, up to 50%, 60%, 70% of that water is absent.
  • the composition comprises a cell adhesion composition chemically linked, e.g., covalently attached, to the polymer.
  • the cell adhesion composition comprises a peptide comprising an RGD amino acid sequence.
  • the composition comprises a eukaryotic cell in one or more of the open interconnected pores.
  • the eukaryotic cell comprises a live attenuated cancer cell (e.g., irradiated cell acts as cancer antigen).
  • the composition comprises a biomolecule in one or more of the open interconnected pores.
  • Biomolecules include small molecule compounds (e.g., less than 1000 daltons in molecular mass), nucleic acids, proteins or fragments thereof, peptides.
  • biomolecules include granulocyte macrophage-colony stimulating factore (GM-CSF), large nucleic acid compositions such as plasmid DNA, and smaller nucleic acid compositions such as CpG oligodeoxynucleotide (CpG-ODN).
  • GM-CSF granulocyte macrophage-colony stimulating factore
  • CpG-ODN CpG oligodeoxynucleotide
  • cryogel compositions are injectable through a hollow needle.
  • the composition maintains structural integrity and shape memory properties, i.e., after compression or dehydration, the composition regains its shape after it is rehydrated or the shear forces of compression are removed/relieved
  • the composition comprises an alginate-based hydrogel.
  • polymer compositions from which the cryogel is fabricated include hyaluronic acid, gelatin, heparin, dextran, carob gum, PEG, PEG derivatives including PEG-co-PGA and PEG-peptide conjugates.
  • the techniques can be applied to any biocompatible polymers, e.g. collagen, chitosan, carboxymethylcellulose, pullulan, polyvinyl alcohol (PVA), Poly(2-hydroxyethyl methacrylate) (PHEMA), Poly(N-isopropylacrylamide) (PNIPAAm), Poly(acrylic acid) (PAAc), etc.
  • the shape of the cryogel is dictated by a mold and can thus take on any shape desired by the fabricator, e.g., various sizes and shapes (disc, cylinders, squares, strings, etc.) are prepared by cryogenic polymerization.
  • Injectable cryogels can be prepared in the micrometer-scale to millimeter-scale. Volume varies from a few hundred ⁇ m 3 to over 100 mm 3 .
  • An exemplary scaffold composition is between 1 mm 3 and 10 mm 3 in size.
  • the cryogel is defined by volume.
  • the cryogel scaffold composition comprises 25 ⁇ l in volume in a hydrated state. The gels are hydrated in an aqueous medium.
  • Exemplary cryogel compositions are typically in the range of 10-70 ⁇ l in volume and may be larger or smaller depending on the use and site to be treated.
  • the cryogel acts as a sponge.
  • the cryogels are sterilized.
  • the cryogels are hydrated, loaded with cells or other compounds (e.g., small molecules and other compounds, nucleic acids, or proteins/peptides) and loaded into a syringe or other delivery apparatus.
  • the syringes are prefilled and refridgerated until use.
  • the cryogel is dehydrated, e.g., lyophylized, optionally with a drug or other compound loaded in the gel and stored dry or refridgerated.
  • the cryogel-loaded syringe or apparatus Prior to administration, the cryogel-loaded syringe or apparatus is contacted with a solution containing cells and/or other compounds to be delivered.
  • the barrel of the cryogel pre-loaded syringe is filled with a physiologically-compatible solution, e.g., phosphate-buffered saline (PBS).
  • a physiologically-compatible solution e.g., phosphate-buffered saline (PBS).
  • PBS phosphate-buffered saline
  • the cryogel is administered to a desired anatomical site followed by the volume of solution, optionally containing other ingredients, e.g., cells or therapeutic compounds.
  • a 25 ⁇ l cryogel is administered with approximately 200 ⁇ l of solution.
  • the cryogel is then rehydrated and regains its shape integrity in situ.
  • the volume of PBS or other physiologic solution administered following cryogel placement is generally about 10 times the volume of the cryogel itself.
  • a method for repairing, regenerating, or restructuring a tissue comprises administering to a subject the device/cryogel composition described above. If the cryogel contains cells, the cells retain their viability after passage through the syringe or delivery apparatus, cells proliferate in the device/cryogel, then leave the cryogel composition to function outside of the gel and in the bodily tissues of the recipient subject.
  • the cryogel is administered subcutaneously as a dermal filler, thereby restructuring the tissue, e.g., dermal tissue.
  • the cryogel device comprises a stem cell and the composition/device is administered to a damaged or diseased tissue of a subject, thereby repairing or regenerating the tissue, e.g., muscle, bone, kidney, liver, heart, bladder, ocular tissue or other anatomic structures.
  • tissue e.g., muscle, bone, kidney, liver, heart, bladder, ocular tissue or other anatomic structures.
  • cryogel compositions are used in a method for delivering genetic material, e.g., to deliver plasmid DNA.
  • a method for eliciting an immune response is carried out by administering to a subject a cryogel composition as described above that further contains a microbial pathogen or tumor cell to which an immune response is elicited.
  • a vaccine composition is administered prophylactically or therapeutically.
  • Cell viability is minimally affected or unaffected by the shear thinning process, and gel/cell constructs stay fixed at the point of introduction. As such, these gels are useful for the delivery of cells and other compounds to target biological sites in therapeutic methods such as tissue regeneration (cell therapy, drug delivery) efforts.
  • the invention provides a device comprising an injectable scaffold composition with open, interconnected macropores.
  • the scaffold composition is injectable through a hollow needle.
  • the scaffold composition is injectable through a 16-gauge, an 18-gauge, a 20-gauge, a 22-gauge, a 24-gauge, a 26-gauge, a 28-gauge, a 30-gauge, a 32-gauge, or a 34-gauge needle.
  • the scaffold composition Upon compression, the scaffold composition maintains shape memory properties.
  • the scaffold composition also maintains structural integrity in that it is flexible (i.e., not brittle) and does not break under sheer pressure.
  • the scaffold composition is an alginate-based hydrogel.
  • the scaffold composition is between 0.01 mm 3 and 100 mm 3 .
  • the scaffold composition is between 1 mm 3 and 75 mm 3 , between 5 mm 3 and 50 mm 3 , between 10 mm 3 and 25 mm 3 .
  • the scaffold composition is between 1 mm 3 and 10 mm 3 in size.
  • the hydrogel comprises macropores, e.g., pores that are characterized by a diameter of 2 ⁇ m-1 mm.
  • the average pore size comprises 200 ⁇ m.
  • Cells can move into and out of the cryogel via the open interconnected pores as a typical cell comprises a diameter or about 20 ⁇ m.
  • the gel delivery devices are suitable for treatment of human beings, as well as animals such as horses, cats, or dogs.
  • the hydrogel is characterized by shape-memory.
  • the polymer chains of the hydrogel are covalently crosslinked and/or oxidized.
  • Such hydrogels are suitable for minimally-invasive delivery. Prior to delivery into the human body, such a hydrogel is lyophylized and compressed prior to administration to a subject for the regeneration of muscle tissue. Minimally-invasive delivery is characterized by making only a small incision into the body. For example, the hydrogel is administered to a muscle of a subject using a needle or angiocatheter.
  • Injectable cryogels have been designed to pass through a hollow structure, e.g., very fine needles, such as 18-30G needles, as a tissue filler for applications in cosmetic surgery, for tissue augmentation, and tissue repair which may be due to injury caused by disease and external trauma.
  • the injectable cryogels may be molded to a desired shape, in the form of rods, square, disc, spheres, cubes, fibers, foams.
  • the injectable cryogels can be used as scaffolds for cell incorporation.
  • the formed cryogel is mixed with cells to provide tissue engineered products, or can be used as a bio-matrix to aid tissue repair or tissue augmentation.
  • the incorporated cells can be any mammalian cells (e.g. stem cells, fibroblasts, osteoblasts, chrondrocytes, immune cells, etc).
  • Injectable cryogels can also be produced in a form in which pharmaceuticals or other bioactive substances (e.g. growth factors, DNA, enzymes, peptides, drugs, etc) are incorporated for controlled drug delivery.
  • pharmaceuticals or other bioactive substances e.g. growth factors, DNA, enzymes, peptides, drugs, etc.
  • Injectable cryogels may be further functionalized by addition of a functional group chosen from the group consisting of: amino, vinyl, aldehyde, thiol, silane, carboxyl, azide, alkyne.
  • the cryogel may be further functionalized by the addition of a further cross-linker agent (e.g. multiple arms polymers, salts, aldehydes, etc).
  • the solvent may be aqueous, and in particular acidic or alkaline.
  • the aqueous solvent may comprise a water-miscible solvent (e.g. methanol, ethanol, DMF, DMSO, acetone, dioxane, etc).
  • the cryo-crosslinking takes place in a mold and the injectable cryogels may be degradable.
  • the pore size can be controlled by the selection of the main solvent used, the incorporation of a porogen, the freezing temperature applied, the cross-linking conditions (e.g. polymer concentration), and also the type and molecule weight of the polymer used.
  • Exemplary applications include use as a dermal filler, in drug delivery, as a wound dressing, for post surgical adhesion prevention, and for repair and/or regenerative medical applications such as cell therapy, gene therapy, tissue engineering, immunotherapy.
  • FIGS. 1A-1D are a series of photomicrographs showing injectable alginate-based hydrogel systems.
  • Rhodamine-labeled 1% methacrylated (MA)-alginate gels with various sizes and shapes (disc, cylinders, squares, etc.) were prepared by cryogenic polymerization. Square shape injectable scaffolds are shown.
  • Fluorescent macroscopic gels suspended in 0.2 mL of phosphate buffered saline (PBS) were injected via 16-gauge diameter needles (FIG. 1 B) with a complete geometric restoration as illustrated in the microscopy image before ( FIGS. 1A and 1D ) and after injection ( FIGS. 1C and 1D ).
  • PBS phosphate buffered saline
  • FIG. 2 is a line graph demonstrating stress vs. strain curves for conventional nanoporous and macroporous 1% rhodamine-labeled MA-alginate gels subjected to compression tests.
  • alginate cryogels In contrast to the brittle nature of the conventional nanoporous gels, alginate cryogels have the ability to withstand reversibly large deformation while keeping their structural integrity and shape memory properties.
  • FIG. 3A is a fluorescence photograph showing minimally invasive subcutaneous injection of macroporous scaffolds into the lower back of mice.
  • FIG. 3B is a photograph showing hydrogel localization after subcutaneous injection of preformed rhodamine-labeled 1% MA-alginate gels (4 mm ⁇ 4 mm ⁇ 1 mm) in the subcutis of a mouse after 3 days.
  • FIG. 3C is a photograph showing merged phase-contrast and fluorescence of a subcutaneously injected rhodamine-labeled alginate macroporous scaffold with restoration of geometry after placement.
  • FIG. 3D is a photograph of a subcutaneously injected rhodamine-labeled alginate macroporous scaffold with restoration of geometry.
  • FIGS. 4A-4F are a series of photographs showing that injectable pre-seeded scaffolds promote in situ localization of bioluminescent B16 cells.
  • FIG. 4A is a photograph showing alginate cryogel scaffolds (white) and rhodamine-labeled alginate scaffolds (pink).
  • Bioluminescence B16-F10 cells were seeded on 1% RGD-modified MA-Alginate cryogels at a concentration of 200 ⁇ 10 3 cells/scaffold. Luciferase transected melanoma cells were cultured for 6 hr into rhodamine-labeled alginate cryogels before injection into mice.
  • FIG. 4A is a photograph showing alginate cryogel scaffolds (white) and rhodamine-labeled alginate scaffolds (pink).
  • Bioluminescence B16-F10 cells were seeded on 1% RGD-modified MA-Alginate cryogels at a concentration
  • FIG. 4B is a photograph showing optical live imaging to demonstrate that macroporous alginate gels are suitable for homogenous encapsulation and distribution of bioluminescent B16 cells.
  • FIG. 4C is a photograph showing scanning electron microscope (SEM) imaging to demonstrate that macroporous alginate gels are suitable for homogenous encapsulation and distribution of bioluminescent B16 cells.
  • FIG. 4D is a photograph showing live fluorescence imaging of subcutaneous injections of gels.
  • FIG. 4E is a photograph showing live fluorescence imaging of subcutaneous injections of gels at 2 days post-injection.
  • FIG. 4F is a photograph showing live fluorescence imaging of subcutaneous injections of gels at 9 days post-injection. Bioluminescent B16-cells were visualized by live imaging.
  • FIG. 5 is a diagram showing preparation of an autologous alginate-based active cryogel vaccine containing living attenuated B16-F10 melanoma cells for the prophylactic and therapeutic treatments of skin cancer in mice.
  • CpG (adjuvant) & GM-CSF (cytokine) loaded RGD-modified alginate cryogels were seeded with irradiated B16-F10 cells and cultured for 6 h prior animal vaccination via subcutaneous injection.
  • FIG. 6 is a bar graph showing immunity against B16F10 challenge induced by different vaccination protocols. Infection-mimicking microenvironment from injectable alginate-based cryogel conferred potent anti-tumor immunity. A comparison of the survival time in mice treated with Cryogels; (C) antigen+GM-CSF+CpG-ODN (0.2 ⁇ 10 6 irradiated B16F10 melanoma cells+3 ⁇ g GM 100 ⁇ g CpG), antigen+GM-CSF (0.4 ⁇ 10 6 -CSF+(D) 6 irradiated B16F10 melanoma cells+3 ⁇ g GM), (E) antigen+CpG-ODN (0.4 ⁇ 10 6 irradiated B16F10 melanoma cells+100 ⁇ g CpG).
  • mice were also immunized using 0.4 ⁇ 10 6 B16F10 melanoma cells transduced with the murine GM-CSF gene (A) and bolus injections of 0.4 ⁇ 10 6 irradiated B16F10 melanoma cells+3 ⁇ g GM-CSF+100 ⁇ g CpG-ODN (B). Mice were challenged (Day 6) with 10 5 B16-F10 melanoma tumor cells and monitored for the onset of tumor occurrence. Each group contained 10 mice.
  • FIGS. 7A and 7B are line graphs showing that local delivery of cryogel vaccine promotes recruitment of CD11c(+) DCs and proliferation of CD3(+) T cells.
  • FIG. 7A Cell recruitment and expansion at the injection site and secondary lymphoid organs (LN, spleen) in response to cryogel vaccination and challenge. The in vivo proliferative responsiveness of the cells was assessed by cell counting.
  • LN secondary lymphoid organs
  • FIGS. 7A and 7B Cryogel matrices co-delivering GM-CSF, CpG-ODN, and presenting attenuated B16F10 melanoma cells stimulate potent local and systemic CD11 c (+) DCs and CD3(+) T cells in secondary lymphoid organs (LN and spleen) as well as the cryogel scaffolds.
  • FIG. 9B is a photograph that shows a representative localized light emission in response to application of firefly luciferin after 29 d post injection in mice inoculated with PEI/DNA-containing cryogels.
  • FIG. 10 is a line graph showing 1 H NMR for MA-alginate with its characteristic vinylic peaks ( ⁇ 5.3-5.8 ppm).
  • Deuterated chloroform (D 2 O) was used as solvent, and the polymer concentration was 1% wt/v.
  • the efficiency of alginate methacrylation was calculated based on the ratio of the integrals for alginate protons to the methylene protons of methacrylate.
  • MA-alginate macromonomer was found to have approximately a degree of methacrylation (DM) of 49%.
  • FIGS. 11A and 11B are a series of line graphs showing 1 H NMR of uncross-linked ( FIG. 11A ) and cryopolymerized ( FIG. 11B ) 1% wt/v MA-alginate in D 2 O. Cryogelation is induced directly in an NMR tube. 1 mL of macromonomer solution containing the initiator system was transferred into the NMR tube before cryogenic treatment at ⁇ 20° C. for 17 hr. The vinylic peaks (between 5.3-5.8 ppm) disappeared after cryo-crosslinking. The conversion was evaluated by comparing the relative peaks of uncross-linked and cross-linked methylene protons.
  • FIGS. 12A-12C are a series of photographs showing scanning electron microscopic images of free PLGA microspheres ( FIG. 12A ) and PLGA microspheres dispersed in a alginate square-shaped cryogel ( FIGS. 12B and 12C ).
  • FIGS. 13A and 13B are a series of photographs showing that cells injected via the cryogels have a low apoptosis and cell death.
  • a RGD-containing peptide was chemically attached to the cryogels to improve cell adhesion to the 3D-structure alginate-based scaffolds.
  • Cell viability, spreading, and actin cytoskeleton organization process was assessed by confocal microscopy. Cells colonize the porous structure of the alginate-based cryogel and were observed to be growing inside the pores.
  • FIG. 13A live/dead cell viability assay of D1 mesenchymal stem cells (MSC, 1 d incubation post-injection) and
  • FIG. 13B confocal image showing injected D1 MSC (6 d incubation post-injection) in RGD-modified MA-alginate cryogels.
  • compositions and methods described herein reduce the cost and invasiveness of the tissue engineering approach.
  • tissue engineering used devices and polymer scaffolds that required surgical implantation. Implantation of polymer scaffolds at a surgical site requires anesthesia and incisions, each of which treatment methods have undesirable side effects.
  • compositions and methods that allow tissue engineers and surgeons to engage in tissue engineering applications in a less invasive manner, thereby removing the need for surgical implantation.
  • injectable scaffolds were developed to reduce the invasiveness of a tissue engineering system, thereby eliminating the need for, or reduce the size of, any incisions required to implant the material.
  • a system to be injectable it must be capable of flowing through a hollow small-bore needle.
  • Methods of implantation of a preformed scaffold or injection of a liquid for polymerization in situ presented a number of challenges including short response time, proper gelation conditions, appropriate mechanical strength and persistence time, biocompatibility, and the likelihood to protect protein drugs or cells in some adverse environments.
  • deformable fully-crosslinked and pre-shaped porous scaffold that is easily prepared, processed, and injected through the needle of a syringe was developed.
  • the invention described herein provides a minimally-invasive method of injecting preformed macroporous hydrogels that are loaded with cells and/or therapeutics.
  • Cells are implanted and cultured onto the polymeric matrix before or after administration to a subject.
  • FDA-approved polymer-based scaffolds that support the attachment and proliferation of cells, degradable and capable of releasing drugs (e.g., proteins) at a controlled rate in vivo are designed in any desirable size and shape, and injected in situ as a safe, preformed, fully characterized, and sterile controlled delivery device. Described in detail below are biologically active cell-seeded injectable scaffolds with structural integrity within the body that controllably deliver growth factors while providing cellular building blocks to enhance tissue formation.
  • Seeding and organizing cells prior administration of macroscopic injectable matrices enhance in vivo cell engraftment and provide cell support and guidance in the initial tissue formation stage.
  • This invention is useful for clinical applications including artificial extracellular matrix for tissue engineering, dermal filler in cosmetic surgery, controlled release reservoir for drug and cell delivery, and immune cell reprogramming for cancer vaccines. Additional benefits include less injection pain, less bleeding/bruising and higher levels of patient satisfaction.
  • the present invention describes a non invasive strategy to administer large-size macroporous biodegradable hydrogels as a 3-D scaffold and a drug delivery platform.
  • Any biocompatible polymers or monomers undergoing cryopolymerization are utilized. Suitable polymers and monomers include naturally derived polymers (alginate, hyaluronic acid, heparin, gelatin, carob gum, collagen, etc.) and synthetic polymers (poly(ethylene glycol) (PEG), PEGylated glutaminase (PEG-PGA), PEG-poly(L-lactide; PLA), poly(2-hydroxyethyl methacrylate) (pHEMA), PAAm, poly(N-isopropylacrylamide) (PNIPAAm), etc.).
  • PEG poly(ethylene glycol)
  • PEG-PGA PEGylated glutaminase
  • PLA poly(2-hydroxyethyl methacrylate)
  • PAAm poly(N-isopropylacrylamide)
  • the highly elastic macroscopic scaffolds with spongy-like morphology are prepared by cryogelation, a technique used to produce polymeric materials with large interconnected pores, high volume fraction porosity within soft, mechanically stable and high water absorbing capacity.
  • cryogels allow for the injectability of preformed large-size scaffolds through a needle without the need of an invasive implantation.
  • Flowable material can fill any defect due to the sponginess of the network.
  • Elastic deformation of cryogels by external forces (mechanical deformation) led to abrupt gel shrinkage with full shape recovery capability, which is useful in the design of injectable preformed scaffolds for cell delivery in a minimally-invasive fashion for tissue engineering and regenerative medicine.
  • the strategies described herein are for delivery of preformed biomaterials suitable for minimally invasive therapies.
  • Injectable macroscopic biomaterials are useful as surgical tissue adhesives, space-filling injectable materials for hard and soft tissue repair, drug delivery, and tissue engineering.
  • Described herein is an approach of pure alginate scaffolds fabrication, which resulted in the formation of, interconnected, superporous network (pore size in the range of 10 ⁇ m-600 ⁇ m).
  • pore size in the range of 10 ⁇ m-600 ⁇ m.
  • These spongy-like gels are highly flexible and squeezable, capable of releasing up to 70% of their water content without altering the gel microstructure.
  • the gel further includes a large range of purified polymers such as hyaluronic acid, heparin, carob gum, gelatin etc; or a cell adhesive molecule such as fibronectin, or integrin binding peptide.
  • the hydrogel is used as a drug reservoir for the controlled delivery of one or more therapeutic agents.
  • Alginate-based gels have excellent mechanical properties, elongation, and fast shape recovery by elasticity. The shape of the gels, which was deformed by an external force (e.g., shear stress), was recovered by swelling in a very short time ( ⁇ 1 s). This recovery had good persistence and repeatability.
  • the superporous (e.g., greater than 75% porosity) scaffolds described herein offer significant advantages such as injectability and easy and efficient cell encapsulation post-polymerization.
  • the cryogels are characterized by porosities of 80-90% or more. Animal studies were performed to examine the integration of the spongy-like gels with the host tissue show that the alginate-based scaffolds are biocompatible and do not elicit an immune response or rejection when injected in mice.
  • Methacrylated alginate was prepared by reacting high molecular weight alginate with aminoethyl methacrylate (AEMA). To synthesize methacrylated alginate with 100% theoretical methacrylation of uronic acid carboxylate groups, high molecular weight sodium alginate (1 g) was dissolved in a buffer solution (0.6% w/v, pH ⁇ 6.5) of 100 mM MES containing 0.5 M NaCl.
  • N-Hydroxysuccinimide (NHS, 1.3 g) and N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDC, 2.8 g) was added to the reaction mixture to activate the carboxylic acid groups of the alginate.
  • EDC N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride
  • 1 H NMR was used to confirm the chemical modification of alginate and characterize the degree of functionalization of MA-alginate ( FIG. 10 ).
  • any biocompatible water-soluble polymer or monomer can be used to make injectable cryogels.
  • Several monomers/polymers or a combination of polymers have been used to make the injectable cryogel devices described herein, e.g., hyaluronic acid, gelatin, heparin, dextran, carob gum, PEG, PEG derivatives including PEG-co-PGA and PEG-peptide conjugates.
  • the polymers may be a combination of degradable and non-degradable synthetic polymers and natural polymers (polysaccharides, peptides, proteins, DNA).
  • Biocompatible synthetic polymers include Polyethylene glycol (PEG), Polyvinyl alcohol (PVA), Poly(2-hydroxyethyl methacrylate) (PHEMA), Poly(N-isopropylacrylamide) (PNIPAAm), Poly(acrylic acid) (PAAc), Polyesters (e.g. Polylactide, Polyglycolide, Polycaprolactone), and Polyanhydrides.
  • Naturally-occurring polymers include Carbohydrates (e.g. Starch, Cellulose, Dextrose, Alginate, Hyaluronic Acid, Heparin, Dextran, Gellan Gum, etc), Proteins (e.g. Gelatin, Albumin, Collagen), Peptides, and DNA. All compositions are purified prior to fabrication of the hydrogels.
  • injectable cryogels are optionally polymerized using other processes.
  • injectable cryogels can be classified under two main groups according to the nature if their cross-linking mechanism, namely chemically and physically cross-linked gels.
  • Covalent cross-linking processes include radical polymerization (vinyl-vinyl coupling), michael-type addition reaction (vinyl-thiol cross-linking), Condensation (carboxylic acid-alcholol and carboxylic acid-amine cross-linking), Oxidation (thiol-thiol cross-linking), Click chemistry (1,3-dipolar cycloaddition of organic azides and alkynes), Diels-Alder reaction (cycloaddition of dienes and dienophiles), Oxime, Imine and Hydrazone chemistries.
  • Non-covalent cross-linking include Ionic cross-linking (e.g. calcium-crosslinked alginate), Self assembly (phase transition in response to external stimuli, such as Temperature, pH, ion concentration, hydrophobic interactions, light, metabolite, and electric current).
  • Cryogel matrices were synthesised by redox-induced free radical polymerization of MA-alginate in water.
  • Alginate cryogels are synthesized by mixing 10 mg (1% wt/v) of MA-alginate macromonomer in deionized water with TEMED (0.5% wt/v) and APS (0.25% wt/v). The mixture is immediately poured into a pre-cooled Teflon mold and frozen at ⁇ 20° C. After cryo-crosslinking has finished, gels are heated to room temperature to remove ice crystals, and washed with distilled water.
  • RGD-containing peptide composition e.g., ACRL-PEG-G4RGDASSKY (SEQ ID NO:2) as a comonomer (0.8% wt/v) during the polymerization.
  • ACRL Acryloyl
  • RGD-containing peptide composition monomers
  • RGD becomes chemically attached (covalently attached) to the polymer structure.
  • RGD integrin-binding motif was used to promote cell-substrate interactions.
  • NMR spectroscopy was used to characterize vinyl conversion of MA-alginate macromonomer after cryopolymerization. As shown in FIG.
  • cryogels see FIG. 11 .
  • Injectable cryogels can be prepared at different concentrations depending on the MW and the degree of chemical modification of the polymer itself (1% wt/v was chosen as a proof of concept).
  • RGD remains attached to the polymer structure by virtue of covalent bonding (co-polymerization).
  • certain biomolecules are to be released following administration of the cryogel to the subject. In this case, the biomolecules are simply mixed with the polymer prior to the cryogelation process.
  • Cryogels are a class of materials with a highly porous interconnected structure that are produced using a cryotropic gelation (or cryogelation) technique.
  • Cryogelation is a technique in which the polymerization-crosslinking reactions are conducted in quasi-frozen reaction solution.
  • the macromoner (MA-alginate) solution the macromonomers and initiator system (APS/TEMED) expelled from the ice concentrate within the channels between the ice crystals, so that the reactions only take place in these unfrozen liquid channels.
  • APS/TEMED macromonomers and initiator system
  • a porous material is produced whose microstructure is a negative replica of the ice formed. Ice crystals act as porogens.
  • Pore size is tuned by altering the temperature of the cryogelation process. For example, the cryogelation process is typically carried out by quickly freezing the solution at ⁇ 20° C. Lowering the temperature to, e.g., ⁇ 80° C., would result in more ice crystals and lead to smaller
  • cryogels compared to conventional macroporous hydrogels obtained by phase separation is their high mechanical stability. They are very tough, and can withstand high levels of deformations, such as elongation and torsion; they can also be squeezed under mechanical force to drain out their solvent content.
  • the improved mechanical properties of alginate cryogels originate from the high crosslinking density (highly methacrylated alginate polymerizes into cross-linked polymer structures with a relatively high crosslink density) of the unfrozen liquid channels of the reaction system. Thus, after polymerization, the gel channels with high polymer content are perfect materials for building the pore walls.
  • Biomolecules e.g., GM-CSF, CpG nucleic acids
  • GM-CSF CpG nucleic acids
  • these molecules are released from the cryogel by diffusion or gel degradation over time.
  • low molecular weight compositions e.g., CpG oligonucleotides
  • Larger entrapped molecules greater than about 10 kDa, e.g., 10-50 kDa in molecular mass
  • proteins large DNAs, e.g., plasmid DNA
  • Human Recombinant GM-CSF (e.g., available from PeproTech, Catalog #300-03) is encoded by the following polypeptide sequence (SEQ ID NO:1):
  • Injectable delivery systems for therapeutic proteins have attracted wide attention.
  • Conventional hydrogels typically release their hydrophilic contents too rapidly in a large initial burst, and phagocytes may clear microspheres within a relatively short time period after administration.
  • Microsphere/cryogel combination systems achieve a controlled and sustained release of proteins as an injectable delivery system.
  • PLGA microspheres size ⁇ 10-50 ⁇ m
  • GM-CSF model protein
  • the mixing ratio of the components was optimized to retain injectability and shape memory properties of pure alginate cryogels.
  • PLGA microspheres were physically entrapped within the cryogel network (polymeric walls) of cryogels.
  • hybrid cryogel have been created as a carrier for controlled delivery of hydrophobic and/or low molecule weight drugs. The results not only provide a strategy for delivery drugs from an injectable 3-D preformed macroporous scaffolds as a sustained-release drug carrier but also open an avenue for the design of the hybrid injectable hydrogels.
  • hybrid polymer combinations include cryo-ferrogels and polydiacetylene-based cryogels.
  • One class of injectable porous biomaterials for on-demand drug and cell delivery comprises cryo-ferrogels.
  • injectable color-changing biomaterials such as polydiacetylene-based cryogels, which change in response to external stimuli such as mechanical forces.
  • the materials contain mechanophore-molecules (e.g., Polydiacetylene Liposome) that undergo a geometric distortion when a certain amount of force is exerted upon it, leading to a color transition.
  • mechanophore-molecules e.g., Polydiacetylene Liposome
  • Smart polymers that change color when the material becomes overstressed are very useful to identify cell-substrate interactions and to accurately measure deformations.
  • Syringes and needles are typically used to introducing the cryogels into the body.
  • the term “syringe” technically refers to the reservoir (that holds the liquid) and the plunger (which pushes the liquid out of the reservoir).
  • the “needle” is the part that enters the body, e.g., into a vein, under the skin, or into muscle or other tissue.
  • the needle gauge refers to the size of the bore or hole in the needle. The higher the gauge, the thinner the needle (and the smaller the hole). A 28 gauge needle (abbreviated 28G) is therefore thinner than a 25 gauge needle, which is in turn thinner than an 18 gauge needle. Insulin needles are typically 1 ⁇ 2 inch in length and tuberculin needles are typically 5 ⁇ 8 of an inch in length. As inscribed on packaging, needle length appears after the gauge number: “28G 1 ⁇ 2” refers to a 28 gauge needle that is 1 ⁇ 2 inch long.
  • Muscle syringes are typically 1 cc in volumes, but larger volumes are sometimes, e.g., 2 to 5 ccs syringes, depending on the application. Larger volumes and larger bores are appropriate for delivery of cryogels for larger scale muscle repair or regeneration, e.g., after extensive or traumatic laceration of tissue such as injuries incurred in battle or car/plane accidents.
  • Intravenous injectors or needles are used for fine or delicate tissue therapy, e.g., cosmetic dermal filler administration. Such applications typically use shorter needles no larger than 25G.
  • Reversible compatible behavior enables pre-formed cryogels with desired physical properties, as characterized ex-vivo, to be delivered in-vivo via application of a moderate non-destructive shear stress during injection through a syringe. Studies were carried out to evaluate whether the fluid velocity, dynamic pressure, and shear stress resulting from the injection affects cell viability.
  • cryogels are capable of absorbing most of the energy when the scaffolds are compressed, thereby, maintaining high cell viability (92%) and their proliferative potential as shown in FIG. 13 .
  • the shear stress (or compression) applied to cells in the cryogel as they pass through the bore of a needle or other delivery apparatus such as a catheter does not measurably hurt or damage the cells within the cryogel.
  • cell viability was routinely 90% or greater.
  • compositions and methods described herein provide hydrogels for minimally invasive delivery of shape memory scaffolds for in vivo applications.
  • This method has demonstrated highly efficient and reproducible fabrication of injectable shape-defined macroporous scaffolds.
  • the material performance is readily manipulated by altering its composition, formulation, and degradation profile.
  • the formation of specific shapes and structural stability are desirable characteristics for shape-defined materials, and the most important requirement of these types of materials for minimally invasive therapies is the ability to collapse and faithfully reform the scaffold's structure in a stimulus-responsive manner.
  • a combination of mechanical compression and dehydration is sufficient to compress the scaffolds developed in this work, allowing minimally invasive delivery through a conventional-gauge needle.
  • cylindrical (4 mm diameter ⁇ 8 mm height) nanoporous gels reduced their heights by ⁇ 16% when subjected to a vertical load before mechanical fracture.
  • cylindrical macroporous gels give much larger deformation under lower mechanical stress, due to its lower modulus.
  • Macroporous scaffolds attained 90% or more of compression strain without mechanical fracture, demonstrating their ability to maintain their structural integrity after compression, compaction, and minimally invasive delivery. Also, these results confirmed that the scaffolds displayed shape memory in vitro.
  • the large volume change of the macroporous shape-defined gels was caused by reversible collapse of the interconnected pores.
  • the collapsing pores force water contained in the macropores to flow out of the gel.
  • Gel deformation and water convection enhances water transport in and out of the gel.
  • the elastically deformed gel immediately returns to its original, unreformed shape-defined configuration in less than 1 s, as surrounding water was reabsorbed into the gel.
  • Example 3 Shape Memory Injectable Scaffolds as a Controlled Drug Delivery Carrier
  • Covalently crosslinked alginate scaffolds possessing shape memory properties were successfully used as a drug delivery system in vivo.
  • the gels having a predefined size and structure were able to exceptionally maintain their structural features after minimally invasive subcutaneously insertion in mice.
  • Suspended gels in PBS were spontaneously hydrated with full geometric restoration after one single injection per site on the lower back of mice. Injected animals did not demonstrate abnormalities in feeding, grooming, or behavior during the time frame of the experiment, nor did they exhibit signs of distress.
  • the hydrogels maintained their hydrogel shape integrity at the site of injection.
  • Animal studies performed to examine the integration of the spongy-like gels with the host tissue showed that the alginate-based scaffolds were biocompatible and did not elicit an immune response or rejection when injected in mice.
  • rhodamine-labeled scaffolds were surgically removed from mice and analyzed. As shown in FIG. 3B , the scaffold guided in vivo tissue formation around the scaffold indicating the scaffolds could support tissue growth and integration.
  • fluorescent microscopy used to visualize the rhodamine-labeled scaffold noticeably displayed the original geometry, structural integrity, square-defined shape retention of the gels in vivo ( FIG. 3C ).
  • Rhodamine-labeled BSA was also used as a drug delivery model. By providing a drug depot at the site of injection, such devices achieve high local drug concentrations without significant systemic administration. Sustained release of BSA was achieved from the injected square-defined scaffolds as shown in FIG. 3D . Targeted and controlled delivery of rhodamine-labeled BSA in mice was quantified via real-time non-invasive live imaging ( FIG. 3A ). Exemplary compound, BSA, was either physically entrapped or chemically grafted to the scaffold during the cryopolymerization process. As illustrated in FIG. 3E , sustained controlled release of BSA was achieved over of period of 4 months. Surprisingly, the release profiles for both types of BSA were similar indicating that the release is mainly mediated by matrix degradation over protein diffusion.
  • Example 4 Cryogel Compositions Enhance Survivability and Limit Migration of Injected Cells In Vivo
  • compositions and methods described herein are non-invasive methods of cell injection based on cell-scaffold integration.
  • Cell transplantation is a therapeutic option for patients with impaired regional or global function due to cell death.
  • the limited number of transplantation methods of cells is considered a major factor limiting the efficacy of cell therapies.
  • injectable preformed scaffolds offer the possibility of homogeneously distributing cells and molecular signals throughout the scaffold.
  • the scaffolds are injected directly into tissues or cavities, e.g., muscle, bone, skin, fat, organs, even of irregular shape and size, in a minimally invasive manner.
  • the compositions and methods described herein offer significant advantages such as injectability and efficient cell encapsulation post-polymerization while allowing sufficient mechanical strength to withstand biomechanical loading and providing temporary support for the cells.
  • a unique characteristic of these cell/scaffold constructs is that when an appropriate shear stress is applied, the deformable hydrogel is dramatically and reversibly compressed (up to 90% of its volume) resulting in injectable macroporous preformed scaffolds.
  • This property allows gel/cell constructs to be delivered via syringe with high precision to target sites. Homogenous cellular distribution and cell viability are unaffected by the shear thinning process and gel/cell constructs stay fixed at the point of introduction, suggesting that these gels are useful for the delivery of cells to target biological sites in tissue regeneration efforts.
  • melanoma B16 cells Integration of melanoma B16 cells to RGD-modified alginate cryogel scaffolds and their injections into healthy mice was investigated to demonstrate successful syringe-delivery and function of pre-cultured cells while promoting homing, survival, and engraftment of tumorigenic cells.
  • the results presented herein demonstrate that the designed tissue-engineered scaffolds mimic the natural environment where cells normally reside, and as a result tumors are formed after every injection of tumorigenic cell-embedded matrix in healthy BALB/c mice.
  • the inoculation of melanoma cells subcutaneously was monitored via real-time non-invasive live imaging ( FIG. 4D ). The incidence of tumor formation and tumor growth was examined over a period of 9 days.
  • FIGS. 4D-4E The success of the melanoma B16 tumor model is clearly evident as shown in FIGS. 4D-4E .
  • the cell/scaffold construct has fulfilled several criteria: successful syringe-delivery with precision to a target site and cell survival in their current local environment resulting in tumor formation.
  • rhodamine-labeled (1) and rhodamine-labeled RGD-modified (2) cell-seeded alginate cryogels were administered in mice to study the effect of cell-engraftment in cell transplantation and homing.
  • a bolus of free cells (B) was also injected.
  • Rhodamine-labeled scaffolds were successfully injected subcutaneously as shown in FIG. 4D . Except for the bolus injection site, red-emitting rhodamine dyes show intense fluorescent red spots in each side of the mice's back indicating in vivo localization of cell-seeded scaffolds.
  • bioluminescence of cell-seeded scaffolds was measured 30 min after intraperitoneal injection of luciferin.
  • bioluminescence for injected RGD-modified cell-seeded gels was particularly brighter when compared to the plain scaffolds showing the necessity to incorporate RGD to the polymeric network to support cell-engraftment and thus efficient cell transplantation.
  • the absence of bioluminescence suggests minimal cell retention at the injection site, rapid cell migration, and likely limited cell transplants survival.
  • a minimally invasive scaffold-based active vaccine containing host pathogens was developed for the therapeutic treatment of cancer.
  • the immune system needs an external boost from immunotherapies to be able to become more effective in fighting cancer.
  • the active immunotherapy system described herein was designed to stimulate the patient's immune system, with the objective of promoting an antigen-specific antitumor effect using the body's own immune cells.
  • the cryogel-vaccine leads to a durable antitumor response that protects tumor recurrence.
  • Dendritic cells DCs are antigen-presenting cells critically involved in regulating the immune system.
  • the vaccine mediates in situ manipulation of dendritic cell recruitment, activation, and their dispersion to the lymph nodes. Cytosine-guanosine oligonucleotide (CpG-ODN) was used as an adjuvant further stimulate responses to the vaccine.
  • both components can be easily incorporated into the cryogel matrix and released in a sustained fashion to recruit and host DCs, and subsequently present cancer antigens from the irradiated cells (or other cell-associated antigens) and danger signals to activate resident na ⁇ ve DCs and promote their homing to the lymph nodes, which is necessary for a robust anti-cancer immune response.
  • Specific and protective anti-tumor immunity was generated with our minimally invasive alginate-based active vaccine, as 80% survival was achieved in animals that otherwise die from cancer within a couple of months.
  • the data using the cryogel-based prophylactic vaccine for melanoma was shown to induce a very strong immunologic memory, as 100% survival was achieved in the rechallenged animals following 100 days post vaccination.
  • Different tumor cell-associated antigens are used in the cellular cryogel-based vaccine platform, thereby permitting treatment or prophylaxis for a variety of cancers.
  • Active specific immunotherapy involves the priming of the immune system in order to generate a T-cell response against tumor-associated antigens.
  • One example of the active specific approach is adoptive T-cell therapy, which involves the ex vivo cultivation of T cells with demonstrated activity against a specific target cancer antigen. Cells are obtained from the subject, purified, and cultured. Such ex vivo cultivation increases the frequency of these T cells to achieve therapeutic levels. The cells are then infused back into the patient via injectable alginate-based cryogel.
  • Creating an infection-mimicking microenvironment by appropriately presenting exogenous cytokines (e.g., GM-CSF) and danger signals (e.g., CpG-ODN), in concert with cancer antigen provides a means to precisely control the number and timing of DC tr afficking and activation, in situ.
  • exogenous cytokines e.g., GM-CSF
  • danger signals e.g., CpG-ODN
  • Macroporous cryogel matrices were fabricated for controlled release of GM-CSF to recruit and house host DCs, and with an interconnected porous structure that allows for cell infiltration and subsequently present cancer antigens (irradiated B16F10 melanoma cells) and danger signals (CpG-ODN) to activate the resident DCs and dramatically enhance their homing to lymph nodes and proliferation.
  • Matrices were loaded with 3 mg of GM-CSF and injected into the subcutaneous pockets of C57BL/6J mice.
  • FIG. 3B indicates that the cryogel vaccine controls or therapeutically alters immune cell trafficking and activation in the body.
  • a large number of DCs are recruited to the vaccine site.
  • CD11c(+) DCs may home to the inguinal lymph nodes and spleen, present antigens to naive T cells, and stimulate and expand specific T-cell populations that elicit anti-tumor responses
  • the total number of CD11c(+) DCs is inversely proportional to the total number of CD3(+) T cells.
  • FACS analysis of cells infiltrating the vaccine site revealed a significant CD3(+) T cell response peaking at day 13.
  • Local CD3(+) T cell numbers dropped sharply by day 24 and were negligible at day 30.
  • cryogel matrices released approximately 20% of their bioactive GM-CSF load within the first 5 days, followed by slow and sustained release of bioactive GM-CSF over the next 10 days ( FIG. 8 , cryogel A); this release profile was chosen to allow diffusion of the factor through the surrounding tissue to effectively recruit resident DCs.
  • Cryogels can be successfully used for specific spatiotemporal delivery of several drugs, as the incorporation of a second biomolecule (CpG-ODN) did not alter the release profile of GM-CSF over time ( FIG. 8 , cryogel B).
  • CpG-ODN second biomolecule
  • Hybrid cryogel have been created as a potential carrier for controlled delivery of hydrophobic and/or low molecule weight drugs. Our results not only provide a new strategy for delivery drugs from an injectable 3-D preformed macroporous scaffolds as a sustained-release drug carrier but also open an avenue for the design of new hybrid injectable hydrogels.
  • Example 6 Injectable Biodegradable Cryogels as a Gene Delivery System
  • Nonviral gene delivery systems based upon polycation/plasmid DNA complexes are gaining recognition as an alternative to viral gene vectors for their potential in avoiding immunogenicity and toxicity problems inherent in viral systems. Studies were carried out to determine the feasibility of using a controlled release system based on encapsulated condensed plasmid DNA in injectable cryogels to achieve gene transfer in the surrounding tissues after injection.
  • a unique feature of the cryogel-based gene delivery system is the biodegradability of the polymeric system, which can provide a sustained release of DNA at different rates depending on the polymer, cross-link density, mass fraction, and porosity created during the cryogelation process.
  • Encapsulated DNA complexed with polyethylenimine (PEI), a nondegradable cationic polymer known to be an effective gene carrier, and naked PEI/DNA complexes, which were prepared at a ratio of 7:1 (PEI:DNA) were injected subcutaneously on the lower back of na ⁇ ve mice using luciferase as a reporter gene ( FIG. 9 ).
  • PEI:DNA polyethylenimine
  • encapsulated PEI/DNA displayed strong bioluminescence providing the highest transgene expression at ⁇ 10 photons/s, about two-order of magnitude higher than that produced by naked PEI/DNA.
  • the expression levels for naked PEI/DNA were about the same as day 1 but increased by 1 order of magnitude when released in a controllable fashion from the cryogels. Till 29 days, encapsulated PEI/DNA still provided a level of transgene expression at ⁇ 10 7 photons/s, similar to that observed at previous time points. This level was significantly higher than those offered by naked PEI/DNA.
  • cryogels promote gene transfection to surrounding cells in the subcutis of mice, with an efficiency superior in terms of prolonged gene expression to naked DNA.
  • the results establish an injectable delivery system as an effective gene carrier applicable to program or treat targeted cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Transplantation (AREA)
  • Dermatology (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dispersion Chemistry (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Oncology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Surgery (AREA)
  • Medicinal Preparation (AREA)
  • Materials For Medical Uses (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Immobilizing And Processing Of Enzymes And Microorganisms (AREA)
  • Polymers & Plastics (AREA)

Abstract

The invention provides polymer compositions for cell and drug delivery.

Description

    RELATED APPLICATION INFORMATION
  • This application is a continuation of U.S. application Ser. No. 14/112,096, filed Oct. 16, 2013, which is a 371 of International Application No.: PCT/US2012/035505, filed Apr. 27, 2012, which claims the benefit of U.S. Provisional Application Ser. No. 61/480,237, filed Apr. 28, 2011, the entire contents of which are incorporated herein by reference.
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • This invention was made with U.S. Government support under Grant Number R01 DE013349 from the National Institutes of Health. The Government has certain rights in the invention.
  • SEQUENCE LISTING
  • This application incorporates by reference the ASCII text file identified by the name “29297-084N01US_ST25.txt”, containing 2 KB of data, created on Dec. 20, 2013 and filed in computer-readable format (CRF) in the parent application, U.S. Ser. No. 14/112,096, on Dec. 27, 2013.
  • FIELD OF THE INVENTION
  • The invention relates to polymer scaffolds for drug and cell delivery systems.
  • BACKGROUND
  • Tissue engineering is an approach for regeneration, replacement, and improvement of the functions of damaged tissues by manipulating materials according to the specific structure or function of the desired tissues. Porous and biodegradable polymer scaffolds are utilized as a structural supporting matrix or as a cell adhesive substrate for cell-based tissue engineering. A major side effect of the surgical implantation of three dimensional scaffolds is the trauma created by physicians while treating patient illness. For example, current technologies for the surgical implantation of three dimensional scaffolds involve incisions that lead to patient pain, bleeding, and bruising. As such, there is a pressing need in the art to develop less invasive structured polymer scaffolds.
  • SUMMARY OF THE INVENTION
  • The present invention provides compositions and a minimally-invasive method of injecting preformed large macroporous polymer-based hydrogels that are loaded with cargo such as cells and/or therapeutics such as small molecule compounds, proteins/peptides (e.g., antigens to which an immune response is desired), or nucleic acids. Hydrogel (also called aquagel) is a network of polymer chains that are hydrophilic, and are sometimes found as a colloidal gel in which water is the dispersion medium. Hydrogels are highly absorbent (they can contain over 99% water) natural or synthetic polymers that possess a degree of flexibility very similar to natural tissue, due to their significant water content. Unlike conventional hydrogels, a unique characteristic of these cell/scaffold constructs described here is that when an appropriate shear stress is applied, the deformable hydrogel is dramatically and reversibly compressed (up to 90% of its volume) resulting in injectable macroporous preformed scaffolds. This property allows gel/cell constructs to be delivered via syringe with high precision to target sites.
  • Accordingly, the invention features a cell-compatible highly crosslinked hydrogel polymer composition comprising a high density of open interconnected pores, wherein the hydrogel is characterized by shape memory following deformation by compression or dehydration. The hydrogel comprises polymers that are modified, e.g., sites on the polymer molecule are modified with a methacrylic acid group (methacrylate (MA)) or an acrylic acid group (acrylate). An exemplary modified alginate is MA-alginate (methacrylated alginate). In the case of Methacrylated-alginate, 50% corresponds to the degree of methacrylation of alginate. This means that every other repeat unit contains a methacrylated group. The degree of methacrylation can be varied from 1% to 90%. Above 90%, the chemical modification may reduce solubility of the polymer water-solubility. Polymers can also be modified with acrylated groups instead of methacrylated groups. The product would then be referred to as an acrylated-polymer. The degree of methacrylation (or acrylation) can be varied for most polymers. However, some polymers (e.g. PEG) maintain their water-solubility properties even at 100% chemical modification. After crosslinking, polymers normally reach near complete methacrylate group conversion indicating approximately 100% of cross-linking efficiency. For example, the polymers in the hydrogel are 50-100% crosslinked (covalent bonds). The extent of crosslinking correlates with the durability of the hydrogel. Thus, a high level of crosslinking (90-100%) of the modified polymers is desirable.
  • For example, the highly crosslinked hydrogel polymer composition is characterized by at least 50% polymer crosslinking (e.g., 75%, 80%, 85%, 90%, 95%, 98%). The high level of crosslinking confers mechanical robustness to the structure. However, the % crosslinking is generally less than 100%. The composition is formed using a free radical polymerization process and a cryogelation process.
  • The cryogel comprises at least 75% pores, e.g., 80%, 85, 90%, 95%, and up to 99% pores. The pores are interconnected. Interconnectivity is important to the function of the composition, as without interconnectivity, water would become trapped within the gel. Interconnectivity of the pores permits passage of water (and other compositions such as cells and compounds) in and out of the structure. In a fully hydrated state, the composition comprises between 90-99% water. In a compressed or dehydrated hydrogel, up to 50%, 60%, 70% of that water is absent.
  • In some examples, the composition comprises a cell adhesion composition chemically linked, e.g., covalently attached, to the polymer. For example, the cell adhesion composition comprises a peptide comprising an RGD amino acid sequence.
  • For cell therapy, the composition comprises a eukaryotic cell in one or more of the open interconnected pores. For example, the eukaryotic cell comprises a live attenuated cancer cell (e.g., irradiated cell acts as cancer antigen). Optionally, the composition comprises a biomolecule in one or more of the open interconnected pores. Biomolecules include small molecule compounds (e.g., less than 1000 daltons in molecular mass), nucleic acids, proteins or fragments thereof, peptides. Exemplary biomolecules include granulocyte macrophage-colony stimulating factore (GM-CSF), large nucleic acid compositions such as plasmid DNA, and smaller nucleic acid compositions such as CpG oligodeoxynucleotide (CpG-ODN).
  • Preferably, the cryogel compositions are injectable through a hollow needle. Upon compression or dehydration, the composition maintains structural integrity and shape memory properties, i.e., after compression or dehydration, the composition regains its shape after it is rehydrated or the shear forces of compression are removed/relieved
  • In one example, the composition comprises an alginate-based hydrogel. Other examples of polymer compositions from which the cryogel is fabricated include hyaluronic acid, gelatin, heparin, dextran, carob gum, PEG, PEG derivatives including PEG-co-PGA and PEG-peptide conjugates. The techniques can be applied to any biocompatible polymers, e.g. collagen, chitosan, carboxymethylcellulose, pullulan, polyvinyl alcohol (PVA), Poly(2-hydroxyethyl methacrylate) (PHEMA), Poly(N-isopropylacrylamide) (PNIPAAm), Poly(acrylic acid) (PAAc), etc. The shape of the cryogel is dictated by a mold and can thus take on any shape desired by the fabricator, e.g., various sizes and shapes (disc, cylinders, squares, strings, etc.) are prepared by cryogenic polymerization. Injectable cryogels can be prepared in the micrometer-scale to millimeter-scale. Volume varies from a few hundred μm3 to over 100 mm3. An exemplary scaffold composition is between 1 mm3 and 10 mm3 in size. In another example, the cryogel is defined by volume. For example, the cryogel scaffold composition comprises 25 μl in volume in a hydrated state. The gels are hydrated in an aqueous medium. Exemplary cryogel compositions are typically in the range of 10-70 μl in volume and may be larger or smaller depending on the use and site to be treated.
  • The cryogel acts as a sponge. The cryogels are sterilized. In some applications, the cryogels are hydrated, loaded with cells or other compounds (e.g., small molecules and other compounds, nucleic acids, or proteins/peptides) and loaded into a syringe or other delivery apparatus. For example, the syringes are prefilled and refridgerated until use. In another example, the cryogel is dehydrated, e.g., lyophylized, optionally with a drug or other compound loaded in the gel and stored dry or refridgerated. Prior to administration, the cryogel-loaded syringe or apparatus is contacted with a solution containing cells and/or other compounds to be delivered. For example, the barrel of the cryogel pre-loaded syringe is filled with a physiologically-compatible solution, e.g., phosphate-buffered saline (PBS). In practice, the cryogel is administered to a desired anatomical site followed by the volume of solution, optionally containing other ingredients, e.g., cells or therapeutic compounds. For example, a 25 μl cryogel is administered with approximately 200 μl of solution. The cryogel is then rehydrated and regains its shape integrity in situ. The volume of PBS or other physiologic solution administered following cryogel placement is generally about 10 times the volume of the cryogel itself.
  • Also within the invention are methods of using the cryogel compostions. For example, a method for repairing, regenerating, or restructuring a tissue comprises administering to a subject the device/cryogel composition described above. If the cryogel contains cells, the cells retain their viability after passage through the syringe or delivery apparatus, cells proliferate in the device/cryogel, then leave the cryogel composition to function outside of the gel and in the bodily tissues of the recipient subject. For example, the cryogel is administered subcutaneously as a dermal filler, thereby restructuring the tissue, e.g., dermal tissue. In another example, the cryogel device comprises a stem cell and the composition/device is administered to a damaged or diseased tissue of a subject, thereby repairing or regenerating the tissue, e.g., muscle, bone, kidney, liver, heart, bladder, ocular tissue or other anatomic structures.
  • In another example, the cryogel compositions are used in a method for delivering genetic material, e.g., to deliver plasmid DNA.
  • In yet another example, a method for eliciting an immune response, is carried out by administering to a subject a cryogel composition as described above that further contains a microbial pathogen or tumor cell to which an immune response is elicited. Such a vaccine composition is administered prophylactically or therapeutically.
  • Cell viability is minimally affected or unaffected by the shear thinning process, and gel/cell constructs stay fixed at the point of introduction. As such, these gels are useful for the delivery of cells and other compounds to target biological sites in therapeutic methods such as tissue regeneration (cell therapy, drug delivery) efforts.
  • The invention provides a device comprising an injectable scaffold composition with open, interconnected macropores. Preferably, the scaffold composition is injectable through a hollow needle. For example, the scaffold composition is injectable through a 16-gauge, an 18-gauge, a 20-gauge, a 22-gauge, a 24-gauge, a 26-gauge, a 28-gauge, a 30-gauge, a 32-gauge, or a 34-gauge needle. Upon compression, the scaffold composition maintains shape memory properties. The scaffold composition also maintains structural integrity in that it is flexible (i.e., not brittle) and does not break under sheer pressure. In one aspect, the scaffold composition is an alginate-based hydrogel. The scaffold composition is between 0.01 mm3 and 100 mm3. For example, the scaffold composition is between 1 mm3 and 75 mm3, between 5 mm3 and 50 mm3, between 10 mm3 and 25 mm3. Preferably, the scaffold composition is between 1 mm3 and 10 mm3 in size.
  • The hydrogel, if to be used to transplant cells, comprises pores to permit the structure to be seeded with cells and to allow the cells to proliferate and migrate out to the structure to relocate to bodily tissues such as the injured or diseased muscle in need of repair or regeneration. For example, cells are seeded at a concentration of about 1×104 to 1×107 cells/ml and are administered dropwise onto a dried hydrogel device. The dose of the gel/device to be delivered to the subject is scaled depending on the magnitude of the injury or diseased area, e.g., one milliliter of gel for a relatively small defect and up to 50 mls of gel for a large wound. Preferable the hydrogel comprises macropores, e.g., pores that are characterized by a diameter of 2 μm-1 mm. The average pore size comprises 200 μm. Cells can move into and out of the cryogel via the open interconnected pores as a typical cell comprises a diameter or about 20 μm. The gel delivery devices are suitable for treatment of human beings, as well as animals such as horses, cats, or dogs.
  • Preferably, the hydrogel is characterized by shape-memory. The polymer chains of the hydrogel are covalently crosslinked and/or oxidized. Such hydrogels are suitable for minimally-invasive delivery. Prior to delivery into the human body, such a hydrogel is lyophylized and compressed prior to administration to a subject for the regeneration of muscle tissue. Minimally-invasive delivery is characterized by making only a small incision into the body. For example, the hydrogel is administered to a muscle of a subject using a needle or angiocatheter.
  • Injectable cryogels have been designed to pass through a hollow structure, e.g., very fine needles, such as 18-30G needles, as a tissue filler for applications in cosmetic surgery, for tissue augmentation, and tissue repair which may be due to injury caused by disease and external trauma. The injectable cryogels may be molded to a desired shape, in the form of rods, square, disc, spheres, cubes, fibers, foams. In some situations, the injectable cryogels can be used as scaffolds for cell incorporation. The formed cryogel is mixed with cells to provide tissue engineered products, or can be used as a bio-matrix to aid tissue repair or tissue augmentation. The incorporated cells can be any mammalian cells (e.g. stem cells, fibroblasts, osteoblasts, chrondrocytes, immune cells, etc).
  • Injectable cryogels can also be produced in a form in which pharmaceuticals or other bioactive substances (e.g. growth factors, DNA, enzymes, peptides, drugs, etc) are incorporated for controlled drug delivery.
  • Injectable cryogels may be further functionalized by addition of a functional group chosen from the group consisting of: amino, vinyl, aldehyde, thiol, silane, carboxyl, azide, alkyne. Alternatively, the cryogel may be further functionalized by the addition of a further cross-linker agent (e.g. multiple arms polymers, salts, aldehydes, etc). The solvent may be aqueous, and in particular acidic or alkaline. The aqueous solvent may comprise a water-miscible solvent (e.g. methanol, ethanol, DMF, DMSO, acetone, dioxane, etc).
  • The cryo-crosslinking takes place in a mold and the injectable cryogels may be degradable. The pore size can be controlled by the selection of the main solvent used, the incorporation of a porogen, the freezing temperature applied, the cross-linking conditions (e.g. polymer concentration), and also the type and molecule weight of the polymer used.
  • Therapeutic and cosmetic uses are described throughout the specification. Exemplary applications include use as a dermal filler, in drug delivery, as a wound dressing, for post surgical adhesion prevention, and for repair and/or regenerative medical applications such as cell therapy, gene therapy, tissue engineering, immunotherapy.
  • Biomolecules are purified naturally-occurring, synthetically produced, or recombinant compounds, e.g., polypeptides, nucleic acids, small molecules, or other agents. For example, the compositions include GM-CSF, pathogen-associated molecular patterns (PAMPs) such as CpG-ODN, and tumor antigens or other antigens. The compositions described herein are purified. Purified compounds are at least 60% by weight (dry weight) the compound of interest. Preferably, the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight the compound of interest. Purity is measured by any appropriate standard method, for example, by column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.
  • Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All published foreign patents and patent applications cited herein are incorporated herein by reference. Genbank and NCBI submissions indicated by accession number cited herein are incorporated herein by reference. All other published references, documents, manuscripts and scientific literature cited herein are incorporated herein by reference. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-1D are a series of photomicrographs showing injectable alginate-based hydrogel systems. Rhodamine-labeled 1% methacrylated (MA)-alginate gels with various sizes and shapes (disc, cylinders, squares, etc.) were prepared by cryogenic polymerization. Square shape injectable scaffolds are shown. Fluorescent macroscopic gels suspended in 0.2 mL of phosphate buffered saline (PBS) were injected via 16-gauge diameter needles (FIG. 1B) with a complete geometric restoration as illustrated in the microscopy image before (FIGS. 1A and 1D) and after injection (FIGS. 1C and 1D).
  • FIG. 2 is a line graph demonstrating stress vs. strain curves for conventional nanoporous and macroporous 1% rhodamine-labeled MA-alginate gels subjected to compression tests. In contrast to the brittle nature of the conventional nanoporous gels, alginate cryogels have the ability to withstand reversibly large deformation while keeping their structural integrity and shape memory properties.
  • FIG. 3A is a fluorescence photograph showing minimally invasive subcutaneous injection of macroporous scaffolds into the lower back of mice. FIG. 3B is a photograph showing hydrogel localization after subcutaneous injection of preformed rhodamine-labeled 1% MA-alginate gels (4 mm×4 mm×1 mm) in the subcutis of a mouse after 3 days. FIG. 3C is a photograph showing merged phase-contrast and fluorescence of a subcutaneously injected rhodamine-labeled alginate macroporous scaffold with restoration of geometry after placement. FIG. 3D is a photograph of a subcutaneously injected rhodamine-labeled alginate macroporous scaffold with restoration of geometry. Dashed lines denote square-shaped geometry restoration of inserted shape-defined scaffolds. FIG. 3E is a line graph showing in vivo sustained release profiles of crosslinked (chemically anchored) or encapsulated (physically entrapped) rhodamine-labeled bovine serum albumin (BSA) to injected cryogels. Upon dissection 3 days post-injection, rhodamine-labeled gels recovered their square shape features, had soft consistencies, and were integrated into the surrounding tissues. Values represent mean and standard deviation (n=4).
  • FIGS. 4A-4F are a series of photographs showing that injectable pre-seeded scaffolds promote in situ localization of bioluminescent B16 cells. FIG. 4A is a photograph showing alginate cryogel scaffolds (white) and rhodamine-labeled alginate scaffolds (pink). Bioluminescence B16-F10 cells were seeded on 1% RGD-modified MA-Alginate cryogels at a concentration of 200×103 cells/scaffold. Luciferase transected melanoma cells were cultured for 6 hr into rhodamine-labeled alginate cryogels before injection into mice. FIG. 4B is a photograph showing optical live imaging to demonstrate that macroporous alginate gels are suitable for homogenous encapsulation and distribution of bioluminescent B16 cells. FIG. 4C is a photograph showing scanning electron microscope (SEM) imaging to demonstrate that macroporous alginate gels are suitable for homogenous encapsulation and distribution of bioluminescent B16 cells. FIG. 4D is a photograph showing live fluorescence imaging of subcutaneous injections of gels. FIG. 4E is a photograph showing live fluorescence imaging of subcutaneous injections of gels at 2 days post-injection. FIG. 4F is a photograph showing live fluorescence imaging of subcutaneous injections of gels at 9 days post-injection. Bioluminescent B16-cells were visualized by live imaging. Arg-Gly-Asp (RGD; cell-adhering peptide)-Alginate scaffolds significantly promoted target delivery of cells compared to unmodified gels. By contrast, injection of free cells (bolus) did not promote localization of cells (bioluminescent signal absent).
  • FIG. 5 is a diagram showing preparation of an autologous alginate-based active cryogel vaccine containing living attenuated B16-F10 melanoma cells for the prophylactic and therapeutic treatments of skin cancer in mice. CpG (adjuvant) & GM-CSF (cytokine) loaded RGD-modified alginate cryogels were seeded with irradiated B16-F10 cells and cultured for 6 h prior animal vaccination via subcutaneous injection.
  • FIG. 6 is a bar graph showing immunity against B16F10 challenge induced by different vaccination protocols. Infection-mimicking microenvironment from injectable alginate-based cryogel conferred potent anti-tumor immunity. A comparison of the survival time in mice treated with Cryogels; (C) antigen+GM-CSF+CpG-ODN (0.2×106 irradiated B16F10 melanoma cells+3 μg GM 100 μg CpG), antigen+GM-CSF (0.4×106-CSF+(D) 6 irradiated B16F10 melanoma cells+3 μg GM), (E) antigen+CpG-ODN (0.4×106 irradiated B16F10 melanoma cells+100 μg CpG). Animals were also immunized using 0.4×106 B16F10 melanoma cells transduced with the murine GM-CSF gene (A) and bolus injections of 0.4×106 irradiated B16F10 melanoma cells+3 μg GM-CSF+100 μg CpG-ODN (B). Mice were challenged (Day 6) with 105 B16-F10 melanoma tumor cells and monitored for the onset of tumor occurrence. Each group contained 10 mice.
  • FIGS. 7A and 7B are line graphs showing that local delivery of cryogel vaccine promotes recruitment of CD11c(+) DCs and proliferation of CD3(+) T cells. (FIG. 7A) Cell recruitment and expansion at the injection site and secondary lymphoid organs (LN, spleen) in response to cryogel vaccination and challenge. The in vivo proliferative responsiveness of the cells was assessed by cell counting. (FIG. 7B) Cryogel matrices co-delivering GM-CSF, CpG-ODN, and presenting attenuated B16F10 melanoma cells stimulate potent local and systemic CD11c(+) DCs and CD3(+) T cells in secondary lymphoid organs (LN and spleen) as well as the cryogel scaffolds. Values in (FIGS. 7A and 7B) represent mean and standard deviation (n=5).
  • FIG. 8 is a line graph showing controlled release of GM-CSF for DC recruitment and programming. Cumulative release of GM-CSF from Alginate-based cryogel matrices over a period of 2 weeks; (A) 3 μg GM-CSF, (B) 3 μg GM-CSF+100 μg CpG-ODN, (C) PLG microsphere containing 3 μg GM-CSF. Values represent mean and standard deviation (n=5).
  • FIG. 9A is a line graph showing cryogel-enhanced plasmid DNA transfection. Relative bioluminescence over time for cells transfected with a luciferase expression plasmid (150 μg/cryogel, 2 injections/animal). Cryogels assist in efficient delivery and cell transfection of polyethylenimine (PEI)/plasmid DNA (blue) when compared to naked PEI/DNA (red). Values represent mean and standard deviation (n=5). FIG. 9B is a photograph that shows a representative localized light emission in response to application of firefly luciferin after 29 d post injection in mice inoculated with PEI/DNA-containing cryogels.
  • FIG. 10 is a line graph showing 1H NMR for MA-alginate with its characteristic vinylic peaks (˜5.3-5.8 ppm). Deuterated chloroform (D2O) was used as solvent, and the polymer concentration was 1% wt/v. The efficiency of alginate methacrylation was calculated based on the ratio of the integrals for alginate protons to the methylene protons of methacrylate. MA-alginate macromonomer was found to have approximately a degree of methacrylation (DM) of 49%.
  • FIGS. 11A and 11B are a series of line graphs showing 1H NMR of uncross-linked (FIG. 11A) and cryopolymerized (FIG. 11B) 1% wt/v MA-alginate in D2O. Cryogelation is induced directly in an NMR tube. 1 mL of macromonomer solution containing the initiator system was transferred into the NMR tube before cryogenic treatment at −20° C. for 17 hr. The vinylic peaks (between 5.3-5.8 ppm) disappeared after cryo-crosslinking. The conversion was evaluated by comparing the relative peaks of uncross-linked and cross-linked methylene protons.
  • FIGS. 12A-12C are a series of photographs showing scanning electron microscopic images of free PLGA microspheres (FIG. 12A) and PLGA microspheres dispersed in a alginate square-shaped cryogel (FIGS. 12B and 12C).
  • FIGS. 13A and 13B are a series of photographs showing that cells injected via the cryogels have a low apoptosis and cell death. In this example, a RGD-containing peptide was chemically attached to the cryogels to improve cell adhesion to the 3D-structure alginate-based scaffolds. Cell viability, spreading, and actin cytoskeleton organization process was assessed by confocal microscopy. Cells colonize the porous structure of the alginate-based cryogel and were observed to be growing inside the pores. (FIG. 13A) live/dead cell viability assay of D1 mesenchymal stem cells (MSC, 1 d incubation post-injection) and (FIG. 13B) confocal image showing injected D1 MSC (6 d incubation post-injection) in RGD-modified MA-alginate cryogels.
  • DETAILED DESCRIPTION
  • A major drawback in today's surgical implantation of three dimensional scaffolds is the trauma created by physicians while administering the scaffolds/devices. The compositions and methods described herein reduce the cost and invasiveness of the tissue engineering approach. Prior to the invention described herein, tissue engineering used devices and polymer scaffolds that required surgical implantation. Implantation of polymer scaffolds at a surgical site requires anesthesia and incisions, each of which treatment methods have undesirable side effects. Described herein are compositions and methods that allow tissue engineers and surgeons to engage in tissue engineering applications in a less invasive manner, thereby removing the need for surgical implantation. As described in detail below, injectable scaffolds were developed to reduce the invasiveness of a tissue engineering system, thereby eliminating the need for, or reduce the size of, any incisions required to implant the material. For a system to be injectable, it must be capable of flowing through a hollow small-bore needle. Methods of implantation of a preformed scaffold or injection of a liquid for polymerization in situ presented a number of challenges including short response time, proper gelation conditions, appropriate mechanical strength and persistence time, biocompatibility, and the likelihood to protect protein drugs or cells in some adverse environments. In order to overcome these limitations, deformable fully-crosslinked and pre-shaped porous scaffold that is easily prepared, processed, and injected through the needle of a syringe was developed.
  • Earlier injectable hydrogels (e.g., U.S. Pat. No. 6,129,761) allowed for the formation of scaffolds in situ but had several major drawbacks. First, potential problems occur with in situ polymerization including heat generation and un-reacted toxic chemicals. Additionally, slow gelation kinetics and in vivo biofluid dynamics involve dispersion of pre-gel solution leading to poor cell entrapment and physical integrity of the gel. Finally, nanosized pore architecture of scaffolds impedes efficient oxygen delivery, nutrient exchange, cell-movement, and long-term survivability of tissue cells.
  • The invention described herein provides a minimally-invasive method of injecting preformed macroporous hydrogels that are loaded with cells and/or therapeutics. Cells are implanted and cultured onto the polymeric matrix before or after administration to a subject. FDA-approved polymer-based scaffolds that support the attachment and proliferation of cells, degradable and capable of releasing drugs (e.g., proteins) at a controlled rate in vivo are designed in any desirable size and shape, and injected in situ as a safe, preformed, fully characterized, and sterile controlled delivery device. Described in detail below are biologically active cell-seeded injectable scaffolds with structural integrity within the body that controllably deliver growth factors while providing cellular building blocks to enhance tissue formation. Seeding and organizing cells prior administration of macroscopic injectable matrices enhance in vivo cell engraftment and provide cell support and guidance in the initial tissue formation stage. This invention is useful for clinical applications including artificial extracellular matrix for tissue engineering, dermal filler in cosmetic surgery, controlled release reservoir for drug and cell delivery, and immune cell reprogramming for cancer vaccines. Additional benefits include less injection pain, less bleeding/bruising and higher levels of patient satisfaction.
  • The present invention describes a non invasive strategy to administer large-size macroporous biodegradable hydrogels as a 3-D scaffold and a drug delivery platform. Any biocompatible polymers or monomers undergoing cryopolymerization are utilized. Suitable polymers and monomers include naturally derived polymers (alginate, hyaluronic acid, heparin, gelatin, carob gum, collagen, etc.) and synthetic polymers (poly(ethylene glycol) (PEG), PEGylated glutaminase (PEG-PGA), PEG-poly(L-lactide; PLA), poly(2-hydroxyethyl methacrylate) (pHEMA), PAAm, poly(N-isopropylacrylamide) (PNIPAAm), etc.). This ability to use different materials is useful in different applications and adds a further degree of versatility to the compositions and methods described herein. The highly elastic macroscopic scaffolds with spongy-like morphology are prepared by cryogelation, a technique used to produce polymeric materials with large interconnected pores, high volume fraction porosity within soft, mechanically stable and high water absorbing capacity. As described below, the cryogels allow for the injectability of preformed large-size scaffolds through a needle without the need of an invasive implantation. Flowable material can fill any defect due to the sponginess of the network. Elastic deformation of cryogels by external forces (mechanical deformation) led to abrupt gel shrinkage with full shape recovery capability, which is useful in the design of injectable preformed scaffolds for cell delivery in a minimally-invasive fashion for tissue engineering and regenerative medicine.
  • The use of large-size preformed scaffolds (>1 mm) mimicking the extracellular matrix was evaluated. Described herein is the design of large biomaterials with various shapes and sizes ranging from 2 mm up to 8 mm that are employed as injectable cell-laden scaffold cryogels. Injectable macroscopic hydrogels are supplied in individual treatment syringes for single patient use and ready for injection (implantation). The gel, consisting of crosslinked alginate suspended in a physiologic buffer, is a sterile, biodegradable, non-pyrogenic, elastic, clear, colorless, homogenized scaffold implant. The injectable gels are packaged in proprietary luer-lock syringes that are injected via a 16-gauge or smaller diameter needle depending on the size of the gel.
  • The strategies described herein are for delivery of preformed biomaterials suitable for minimally invasive therapies. Injectable macroscopic biomaterials are useful as surgical tissue adhesives, space-filling injectable materials for hard and soft tissue repair, drug delivery, and tissue engineering. Described herein is an approach of pure alginate scaffolds fabrication, which resulted in the formation of, interconnected, superporous network (pore size in the range of 10 μm-600 μm). These spongy-like gels are highly flexible and squeezable, capable of releasing up to 70% of their water content without altering the gel microstructure. Optionally, the gel further includes a large range of purified polymers such as hyaluronic acid, heparin, carob gum, gelatin etc; or a cell adhesive molecule such as fibronectin, or integrin binding peptide. In addition, the hydrogel is used as a drug reservoir for the controlled delivery of one or more therapeutic agents. Alginate-based gels have excellent mechanical properties, elongation, and fast shape recovery by elasticity. The shape of the gels, which was deformed by an external force (e.g., shear stress), was recovered by swelling in a very short time (<1 s). This recovery had good persistence and repeatability. The superporous (e.g., greater than 75% porosity) scaffolds described herein offer significant advantages such as injectability and easy and efficient cell encapsulation post-polymerization. For example, the cryogels are characterized by porosities of 80-90% or more. Animal studies were performed to examine the integration of the spongy-like gels with the host tissue show that the alginate-based scaffolds are biocompatible and do not elicit an immune response or rejection when injected in mice.
  • Synthesis of Methacrylated-Alginate (MA-Alginate) and Other Modified Polymers
  • Methacrylated alginate (MA-alginate) was prepared by reacting high molecular weight alginate with aminoethyl methacrylate (AEMA). To synthesize methacrylated alginate with 100% theoretical methacrylation of uronic acid carboxylate groups, high molecular weight sodium alginate (1 g) was dissolved in a buffer solution (0.6% w/v, pH ˜6.5) of 100 mM MES containing 0.5 M NaCl. N-Hydroxysuccinimide (NHS, 1.3 g) and N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDC, 2.8 g) was added to the reaction mixture to activate the carboxylic acid groups of the alginate. After 5 min, AEMA (2.24 g, molar ratio of NHS:EDC:AEMA=1:1.3:1.1) was added to the product and the reaction was maintained at room temperature for 24 h. The mixture was precipitated with the addition of excess of acetone, filtered, and dried in a vacuum oven overnight at room temperature. 1H NMR was used to confirm the chemical modification of alginate and characterize the degree of functionalization of MA-alginate (FIG. 10).
  • Any biocompatible water-soluble polymer or monomer can be used to make injectable cryogels. Several monomers/polymers or a combination of polymers have been used to make the injectable cryogel devices described herein, e.g., hyaluronic acid, gelatin, heparin, dextran, carob gum, PEG, PEG derivatives including PEG-co-PGA and PEG-peptide conjugates. For example, the polymers may be a combination of degradable and non-degradable synthetic polymers and natural polymers (polysaccharides, peptides, proteins, DNA). Biocompatible synthetic polymers include Polyethylene glycol (PEG), Polyvinyl alcohol (PVA), Poly(2-hydroxyethyl methacrylate) (PHEMA), Poly(N-isopropylacrylamide) (PNIPAAm), Poly(acrylic acid) (PAAc), Polyesters (e.g. Polylactide, Polyglycolide, Polycaprolactone), and Polyanhydrides. Naturally-occurring polymers include Carbohydrates (e.g. Starch, Cellulose, Dextrose, Alginate, Hyaluronic Acid, Heparin, Dextran, Gellan Gum, etc), Proteins (e.g. Gelatin, Albumin, Collagen), Peptides, and DNA. All compositions are purified prior to fabrication of the hydrogels.
  • In addition to the free radical polymerization process to cross-link the polymers and make chemically cross-linked injectable cryogels (polymerization time is about 17 hr), gels are optionally polymerized using other processes. Injectable cryogels can be classified under two main groups according to the nature if their cross-linking mechanism, namely chemically and physically cross-linked gels. Covalent cross-linking processes include radical polymerization (vinyl-vinyl coupling), michael-type addition reaction (vinyl-thiol cross-linking), Condensation (carboxylic acid-alcholol and carboxylic acid-amine cross-linking), Oxidation (thiol-thiol cross-linking), Click chemistry (1,3-dipolar cycloaddition of organic azides and alkynes), Diels-Alder reaction (cycloaddition of dienes and dienophiles), Oxime, Imine and Hydrazone chemistries. Non-covalent cross-linking include Ionic cross-linking (e.g. calcium-crosslinked alginate), Self assembly (phase transition in response to external stimuli, such as Temperature, pH, ion concentration, hydrophobic interactions, light, metabolite, and electric current).
  • Cryogel Fabrication
  • Cryogel matrices were synthesised by redox-induced free radical polymerization of MA-alginate in water. Alginate cryogels are synthesized by mixing 10 mg (1% wt/v) of MA-alginate macromonomer in deionized water with TEMED (0.5% wt/v) and APS (0.25% wt/v). The mixture is immediately poured into a pre-cooled Teflon mold and frozen at −20° C. After cryo-crosslinking has finished, gels are heated to room temperature to remove ice crystals, and washed with distilled water. Cell-adhesive cryogels were synthesized using a RGD-containing peptide composition, e.g., ACRL-PEG-G4RGDASSKY (SEQ ID NO:2) as a comonomer (0.8% wt/v) during the polymerization. (Acryloyl is abbreviated ACRL.) By mixing the RGD-containing peptide composition (monomers) with the alginate, the RGD becomes chemically attached (covalently attached) to the polymer structure. RGD integrin-binding motif was used to promote cell-substrate interactions. NMR spectroscopy was used to characterize vinyl conversion of MA-alginate macromonomer after cryopolymerization. As shown in FIG. 2, full disappearance of methylene protons (between 5.3-5.8 ppm) for MA-alginate macromonomer (1% wt/v) was reached after the cryopolymerization process in the presence of the initiator system (APS/TEMED). This indicates that high vinyl conversions can be achieved for cryogels (see FIG. 11). Injectable cryogels can be prepared at different concentrations depending on the MW and the degree of chemical modification of the polymer itself (1% wt/v was chosen as a proof of concept).
  • As described above, RGD remains attached to the polymer structure by virtue of covalent bonding (co-polymerization). However, certain biomolecules are to be released following administration of the cryogel to the subject. In this case, the biomolecules are simply mixed with the polymer prior to the cryogelation process.
  • Cryogelation
  • Cryogels are a class of materials with a highly porous interconnected structure that are produced using a cryotropic gelation (or cryogelation) technique. Cryogelation is a technique in which the polymerization-crosslinking reactions are conducted in quasi-frozen reaction solution. During freezing of the macromoner (MA-alginate) solution, the macromonomers and initiator system (APS/TEMED) expelled from the ice concentrate within the channels between the ice crystals, so that the reactions only take place in these unfrozen liquid channels. After polymerization and, after melting of ice, a porous material is produced whose microstructure is a negative replica of the ice formed. Ice crystals act as porogens. Pore size is tuned by altering the temperature of the cryogelation process. For example, the cryogelation process is typically carried out by quickly freezing the solution at −20° C. Lowering the temperature to, e.g., −80° C., would result in more ice crystals and lead to smaller pores.
  • The advantage of these so-called “cryogels” compared to conventional macroporous hydrogels obtained by phase separation is their high mechanical stability. They are very tough, and can withstand high levels of deformations, such as elongation and torsion; they can also be squeezed under mechanical force to drain out their solvent content. The improved mechanical properties of alginate cryogels originate from the high crosslinking density (highly methacrylated alginate polymerizes into cross-linked polymer structures with a relatively high crosslink density) of the unfrozen liquid channels of the reaction system. Thus, after polymerization, the gel channels with high polymer content are perfect materials for building the pore walls.
  • Biomolecules, e.g., GM-CSF, CpG nucleic acids, are entrapped in the polymer structure but not chemically linked to it. Thus, these molecules are released from the cryogel by diffusion or gel degradation over time. For example, low molecular weight compositions (less than 10 kDa molecular mass), e.g., CpG oligonucleotides, are released by diffusion. Larger entrapped molecules (greater than about 10 kDa, e.g., 10-50 kDa in molecular mass), e.g., proteins, large DNAs, e.g., plasmid DNA, are released primarily by cryogel degradation. Human Recombinant GM-CSF (e.g., available from PeproTech, Catalog #300-03) is encoded by the following polypeptide sequence (SEQ ID NO:1):
  • MAPARSPSPS TQPWEHVNAI QEARRLLNLS RDTAAEMNET
    VEVISEMFDL QEPTCLQTRL ELYKQGLRGS LTKLKGPLTM
    MASHYKQHCP PTPETSCATQ IITFESFKEN LKDFLLVIPF
    DCWEPVQE
  • Injectable Hybrid Cryogels
  • Injectable delivery systems for therapeutic proteins (e.g., hydrogels and microspheres) have attracted wide attention. Conventional hydrogels, however, typically release their hydrophilic contents too rapidly in a large initial burst, and phagocytes may clear microspheres within a relatively short time period after administration.
  • Microsphere/cryogel combination systems achieve a controlled and sustained release of proteins as an injectable delivery system. PLGA microspheres (size ˜10-50 μm) containing a model protein (GM-CSF) were prepared and then mixed with a MA-alginate pre-gel solution prior cryopolymerization. The mixing ratio of the components was optimized to retain injectability and shape memory properties of pure alginate cryogels. As shown in FIG. 12, PLGA microspheres were physically entrapped within the cryogel network (polymeric walls) of cryogels. Also, hybrid cryogel have been created as a carrier for controlled delivery of hydrophobic and/or low molecule weight drugs. The results not only provide a strategy for delivery drugs from an injectable 3-D preformed macroporous scaffolds as a sustained-release drug carrier but also open an avenue for the design of the hybrid injectable hydrogels.
  • Other examples of hybrid polymer combinations include cryo-ferrogels and polydiacetylene-based cryogels. One class of injectable porous biomaterials for on-demand drug and cell delivery comprises cryo-ferrogels. The magnetic-sensitive scaffolds based on macroporous elastic alginate-based cryo-ferrogels, were fabricated with 3-D connected macropores and coupled with magnetic particles (Fe3O4 nano- and micro-particles) and cell-binding moieties. Under applied magnetic fields, the loaded macroporous ferrogel with biological agents lead to large and prompt deformation triggering release of drugs and cells in a controlled fashion. In another example, injectable color-changing biomaterials such as polydiacetylene-based cryogels, which change in response to external stimuli such as mechanical forces. The materials contain mechanophore-molecules (e.g., Polydiacetylene Liposome) that undergo a geometric distortion when a certain amount of force is exerted upon it, leading to a color transition. Smart polymers that change color when the material becomes overstressed are very useful to identify cell-substrate interactions and to accurately measure deformations.
  • Administration of Injectable Cryogels
  • Syringes and needles are typically used to introducing the cryogels into the body. The term “syringe” technically refers to the reservoir (that holds the liquid) and the plunger (which pushes the liquid out of the reservoir). The “needle” is the part that enters the body, e.g., into a vein, under the skin, or into muscle or other tissue. The word “syringe” is also sometimes used to refer to the entire reservoir/plunger/needle combination. They come in a variety of sizes, e.g., a common reservoir size is 1 cc (1 cubic centimeter (cc)=1 milliliter), with a 25 gauge needle size or smaller.
  • The needle gauge refers to the size of the bore or hole in the needle. The higher the gauge, the thinner the needle (and the smaller the hole). A 28 gauge needle (abbreviated 28G) is therefore thinner than a 25 gauge needle, which is in turn thinner than an 18 gauge needle. Insulin needles are typically ½ inch in length and tuberculin needles are typically ⅝ of an inch in length. As inscribed on packaging, needle length appears after the gauge number: “28G ½” refers to a 28 gauge needle that is ½ inch long.
  • Larger gauge (frequently 23G or 21G), longer needles are often used for intramuscular injections. Muscle syringes are typically 1 cc in volumes, but larger volumes are sometimes, e.g., 2 to 5 ccs syringes, depending on the application. Larger volumes and larger bores are appropriate for delivery of cryogels for larger scale muscle repair or regeneration, e.g., after extensive or traumatic laceration of tissue such as injuries incurred in battle or car/plane accidents. Intravenous injectors or needles are used for fine or delicate tissue therapy, e.g., cosmetic dermal filler administration. Such applications typically use shorter needles no larger than 25G.
  • Survivability of Cells after Injection
  • Reversible compatible behavior enables pre-formed cryogels with desired physical properties, as characterized ex-vivo, to be delivered in-vivo via application of a moderate non-destructive shear stress during injection through a syringe. Studies were carried out to evaluate whether the fluid velocity, dynamic pressure, and shear stress resulting from the injection affects cell viability.
  • The data indicated that, during the injection, cells integrated in the RGD-modified cryogel were protected by the scaffold from mechanical damage. Although adherent cells may experience some shear stress applied during the injection, cryogels are capable of absorbing most of the energy when the scaffolds are compressed, thereby, maintaining high cell viability (92%) and their proliferative potential as shown in FIG. 13.
  • Thus, the shear stress (or compression) applied to cells in the cryogel as they pass through the bore of a needle or other delivery apparatus such as a catheter does not measurably hurt or damage the cells within the cryogel. Following passage through a needle or other delivery apparatus, cell viability was routinely 90% or greater.
  • Example 1: Injectable Biodegradable Preformed Macroscopic Geometric Gels
  • The compositions and methods described herein provide hydrogels for minimally invasive delivery of shape memory scaffolds for in vivo applications. This method has demonstrated highly efficient and reproducible fabrication of injectable shape-defined macroporous scaffolds. Although only one type of covalently alginate-based crosslinked gel system was evaluated herein, the material performance is readily manipulated by altering its composition, formulation, and degradation profile. The formation of specific shapes and structural stability are desirable characteristics for shape-defined materials, and the most important requirement of these types of materials for minimally invasive therapies is the ability to collapse and faithfully reform the scaffold's structure in a stimulus-responsive manner. A combination of mechanical compression and dehydration is sufficient to compress the scaffolds developed in this work, allowing minimally invasive delivery through a conventional-gauge needle.
  • These results described herein demonstrated that shape-defined macroporous alginate-based scaffolds were prepared with different geometric sizes and shapes, and successfully passed through a surgical needle without mechanical fracture, and all scaffolds regained their three-dimensional shape immediately (<1 s) after rehydration (FIG. 1). The fabrication method is capable to manufacture biocompatible, biodegradable and complicated macroporous tissue scaffolds efficiently and economically. In addition to the application described herein, shape memory scaffolds are especially useful in applications in which large, structurally defined implants are required.
  • Example 2: Structural Integrity of Injectable Macroscopic Shape-Defined Gels
  • The deformation of conventional (nanoporous) and macroporous 1% MA-alginate gels under mechanical compression associated with shear forces was examined. Subject to mechanical compression, the gels experience a body of force, which results in a shape change. The influence of the macropores on the gel mechanical properties was also evaluated since the stiffness of the scaffold dictate the extent of the deformation under an applied shear force. Conventional gels give a Young's modulus (i.e., the slope of the initial part of the stress vs. strain curves in FIG. 2) of 42±4 kPa in compression test. However, macroporous gels led to a dramatic reduction in the modulus to 4±2 kPa. As shown in FIG. 2, cylindrical (4 mm diameter×8 mm height) nanoporous gels reduced their heights by ˜16% when subjected to a vertical load before mechanical fracture. In comparison, cylindrical macroporous gels give much larger deformation under lower mechanical stress, due to its lower modulus. Macroporous scaffolds attained 90% or more of compression strain without mechanical fracture, demonstrating their ability to maintain their structural integrity after compression, compaction, and minimally invasive delivery. Also, these results confirmed that the scaffolds displayed shape memory in vitro.
  • In the hydrogels described herein, the large volume change of the macroporous shape-defined gels was caused by reversible collapse of the interconnected pores. The collapsing pores force water contained in the macropores to flow out of the gel. Gel deformation and water convection enhances water transport in and out of the gel. Once the mechanical load is removed, the elastically deformed gel immediately returns to its original, unreformed shape-defined configuration in less than 1 s, as surrounding water was reabsorbed into the gel.
  • Example 3: Shape Memory Injectable Scaffolds as a Controlled Drug Delivery Carrier
  • Covalently crosslinked alginate scaffolds possessing shape memory properties were successfully used as a drug delivery system in vivo. The gels having a predefined size and structure were able to exceptionally maintain their structural features after minimally invasive subcutaneously insertion in mice. Suspended gels in PBS were spontaneously hydrated with full geometric restoration after one single injection per site on the lower back of mice. Injected animals did not demonstrate abnormalities in feeding, grooming, or behavior during the time frame of the experiment, nor did they exhibit signs of distress.
  • The hydrogels maintained their hydrogel shape integrity at the site of injection. Animal studies performed to examine the integration of the spongy-like gels with the host tissue showed that the alginate-based scaffolds were biocompatible and did not elicit an immune response or rejection when injected in mice. After 3 days post-injection, rhodamine-labeled scaffolds were surgically removed from mice and analyzed. As shown in FIG. 3B, the scaffold guided in vivo tissue formation around the scaffold indicating the scaffolds could support tissue growth and integration. Furthermore, fluorescent microscopy used to visualize the rhodamine-labeled scaffold, noticeably displayed the original geometry, structural integrity, square-defined shape retention of the gels in vivo (FIG. 3C).
  • Rhodamine-labeled BSA was also used as a drug delivery model. By providing a drug depot at the site of injection, such devices achieve high local drug concentrations without significant systemic administration. Sustained release of BSA was achieved from the injected square-defined scaffolds as shown in FIG. 3D. Targeted and controlled delivery of rhodamine-labeled BSA in mice was quantified via real-time non-invasive live imaging (FIG. 3A). Exemplary compound, BSA, was either physically entrapped or chemically grafted to the scaffold during the cryopolymerization process. As illustrated in FIG. 3E, sustained controlled release of BSA was achieved over of period of 4 months. Surprisingly, the release profiles for both types of BSA were similar indicating that the release is mainly mediated by matrix degradation over protein diffusion.
  • Example 4: Cryogel Compositions Enhance Survivability and Limit Migration of Injected Cells In Vivo
  • One application for the compositions and methods described herein is the non-invasive method of cell injection based on cell-scaffold integration. Cell transplantation is a therapeutic option for patients with impaired regional or global function due to cell death. However, the limited number of transplantation methods of cells is considered a major factor limiting the efficacy of cell therapies. As cell and bioactive molecule carriers, injectable preformed scaffolds offer the possibility of homogeneously distributing cells and molecular signals throughout the scaffold. Moreover, the scaffolds are injected directly into tissues or cavities, e.g., muscle, bone, skin, fat, organs, even of irregular shape and size, in a minimally invasive manner. The compositions and methods described herein offer significant advantages such as injectability and efficient cell encapsulation post-polymerization while allowing sufficient mechanical strength to withstand biomechanical loading and providing temporary support for the cells.
  • Square-shaped rhodamine-labeled RGD-containing alginate cryogels (4×4×1; units: mm) were prepared, purified, sterilized, and subsequently seeded with bioluminescent B16 cells, and maintained in culture for 6 hr in cell culture medium before animal subcutaneous injection to promote cell-scaffold integration (FIGS. 4A, 4B, and 4C). Large interconnected pores significantly enhanced cell seeding and distribution, while maintaining relatively high seeding efficiencies (>50%) and viability (>95%). To image bioluminescence of seeded B16 melanoma cells in vitro, 0.15 mg/g of luciferin was added on top of the gel, which freely diffused through the gel network, staining the cells and indicating homogeneous infiltration and depth viability of cells throughout the 3-D construct (FIG. 4B). This is due to the effective nutrient delivery into and waste removal from the inner regions of the scaffold. SEM images confirmed a homogeneous distribution and engraftment of cells within the scaffold (FIG. 4C).
  • A unique characteristic of these cell/scaffold constructs is that when an appropriate shear stress is applied, the deformable hydrogel is dramatically and reversibly compressed (up to 90% of its volume) resulting in injectable macroporous preformed scaffolds. This property allows gel/cell constructs to be delivered via syringe with high precision to target sites. Homogenous cellular distribution and cell viability are unaffected by the shear thinning process and gel/cell constructs stay fixed at the point of introduction, suggesting that these gels are useful for the delivery of cells to target biological sites in tissue regeneration efforts.
  • Subsequently, healthy C57BL/6 mice received a subcutaneous injection on their backs of 200×103 B16's integrated into alginate macroporous scaffolds. The resulting injected gels were delivered to a targeted site where they quickly recovered to their original mechanical rigidity with location permanency. As shown in FIG. 4D, cell-loaded rhodamine-labeled alginate scaffolds were syringe-delivered (1 cc, 16G) with high precision in the back of mice and visualized by in vivo optical live imaging. Integration of melanoma B16 cells to RGD-modified alginate cryogel scaffolds and their injections into healthy mice was investigated to demonstrate successful syringe-delivery and function of pre-cultured cells while promoting homing, survival, and engraftment of tumorigenic cells. The results presented herein demonstrate that the designed tissue-engineered scaffolds mimic the natural environment where cells normally reside, and as a result tumors are formed after every injection of tumorigenic cell-embedded matrix in healthy BALB/c mice. The inoculation of melanoma cells subcutaneously was monitored via real-time non-invasive live imaging (FIG. 4D). The incidence of tumor formation and tumor growth was examined over a period of 9 days. The success of the melanoma B16 tumor model is clearly evident as shown in FIGS. 4D-4E. As an in vivo model, the cell/scaffold construct has fulfilled several criteria: successful syringe-delivery with precision to a target site and cell survival in their current local environment resulting in tumor formation.
  • As described herein rhodamine-labeled (1) and rhodamine-labeled RGD-modified (2) cell-seeded alginate cryogels were administered in mice to study the effect of cell-engraftment in cell transplantation and homing. As a control, a bolus of free cells (B) was also injected. Rhodamine-labeled scaffolds were successfully injected subcutaneously as shown in FIG. 4D. Except for the bolus injection site, red-emitting rhodamine dyes show intense fluorescent red spots in each side of the mice's back indicating in vivo localization of cell-seeded scaffolds. After 2 days post-injection, bioluminescence of cell-seeded scaffolds was measured 30 min after intraperitoneal injection of luciferin. As shown in FIG. 4E, bioluminescence for injected RGD-modified cell-seeded gels was particularly brighter when compared to the plain scaffolds showing the necessity to incorporate RGD to the polymeric network to support cell-engraftment and thus efficient cell transplantation. For the injection of the cellular bolus, the absence of bioluminescence suggests minimal cell retention at the injection site, rapid cell migration, and likely limited cell transplants survival. Similarly, 9 days post-injection, bioluminescence of cell-seeded scaffolds was mainly apparent for RGD-modified scaffolds confirming the developed non-invasive method for cell injection based on cell-scaffold integration is crucial to decrease migration, promote homing, enhance survivability, and engraftment of cells in vivo (FIG. 4F).
  • Decreasing the rapid cell death that occurs within a few days after transplantation of graft cells is of great relevance for the success of cell transplantation therapies. The results presented herein confirm that the incorporation of the cell-adhesive peptide plays a key role in regulating interactions between cells and the scaffold and cell-fate. These gels are also suitable for use as a delivery system for the sustained delivery of proteins (e.g., growth factors) involved in cell differentiation and maturation (FIG. 3E). This technique is also a tool for enhancing stem cell survival in vivo.
  • Example 5: Injectable Biodegradable Cryogels for Immunotherapy Applications
  • A minimally invasive scaffold-based active vaccine containing host pathogens was developed for the therapeutic treatment of cancer. In the case of cancer, the immune system needs an external boost from immunotherapies to be able to become more effective in fighting cancer. The active immunotherapy system described herein was designed to stimulate the patient's immune system, with the objective of promoting an antigen-specific antitumor effect using the body's own immune cells. In addition, the cryogel-vaccine leads to a durable antitumor response that protects tumor recurrence. Dendritic cells (DCs) are antigen-presenting cells critically involved in regulating the immune system. The vaccine mediates in situ manipulation of dendritic cell recruitment, activation, and their dispersion to the lymph nodes. Cytosine-guanosine oligonucleotide (CpG-ODN) was used as an adjuvant further stimulate responses to the vaccine.
  • As shown in FIG. 5, both components (adjuvant and cytokine) can be easily incorporated into the cryogel matrix and released in a sustained fashion to recruit and host DCs, and subsequently present cancer antigens from the irradiated cells (or other cell-associated antigens) and danger signals to activate resident naïve DCs and promote their homing to the lymph nodes, which is necessary for a robust anti-cancer immune response. Specific and protective anti-tumor immunity was generated with our minimally invasive alginate-based active vaccine, as 80% survival was achieved in animals that otherwise die from cancer within a couple of months. The data using the cryogel-based prophylactic vaccine for melanoma was shown to induce a very strong immunologic memory, as 100% survival was achieved in the rechallenged animals following 100 days post vaccination.
  • Different tumor cell-associated antigens are used in the cellular cryogel-based vaccine platform, thereby permitting treatment or prophylaxis for a variety of cancers. Active specific immunotherapy involves the priming of the immune system in order to generate a T-cell response against tumor-associated antigens. One example of the active specific approach is adoptive T-cell therapy, which involves the ex vivo cultivation of T cells with demonstrated activity against a specific target cancer antigen. Cells are obtained from the subject, purified, and cultured. Such ex vivo cultivation increases the frequency of these T cells to achieve therapeutic levels. The cells are then infused back into the patient via injectable alginate-based cryogel.
  • Creating an infection-mimicking microenvironment by appropriately presenting exogenous cytokines (e.g., GM-CSF) and danger signals (e.g., CpG-ODN), in concert with cancer antigen provides a means to precisely control the number and timing of DC tr afficking and activation, in situ. At different time points post scaffold-based vaccine injection (vax C), cells were isolated from the cryogels and surrounding tissues, spleen, and lymph nodes (LN) for cell counting and fluorescence-activated cell sorting (FACS) analysis to determine the overall number of cells and percentage of DCs (CD11c+cells) and T cells (CD3+cells). Cells infiltrating the vaccine site and the enlargement of spleen and LN after vaccination revealed a significant immunologic response to cancer. The increased numbers of immune system cells fighting cancer antigens made the two organs expand and become “swollen.” As shown in FIG. 3A, the total numbers of cells increased dramatically for the vaccinated (V) and vaccinated/challenged (VC) mice when compared to the control groups (C) for the spleen, LN, and cryogels. The increase number of cells remained relatively high within the first 2 weeks post vaccination and started to noticeably drop by day 13 impaired with a reduction of immunologic and inflammatory responses.
  • Macroporous cryogel matrices were fabricated for controlled release of GM-CSF to recruit and house host DCs, and with an interconnected porous structure that allows for cell infiltration and subsequently present cancer antigens (irradiated B16F10 melanoma cells) and danger signals (CpG-ODN) to activate the resident DCs and dramatically enhance their homing to lymph nodes and proliferation. Matrices were loaded with 3 mg of GM-CSF and injected into the subcutaneous pockets of C57BL/6J mice. FIG. 3B indicates that the cryogel vaccine controls or therapeutically alters immune cell trafficking and activation in the body. Within the first 10 d post vaccination, a large number of DCs are recruited to the vaccine site. As these activated DCs may home to the inguinal lymph nodes and spleen, present antigens to naive T cells, and stimulate and expand specific T-cell populations that elicit anti-tumor responses, the total number of CD11c(+) DCs is inversely proportional to the total number of CD3(+) T cells. FACS analysis of cells infiltrating the vaccine site revealed a significant CD3(+) T cell response peaking at day 13. Local CD3(+) T cell numbers dropped sharply by day 24 and were negligible at day 30.
  • These cryogel matrices released approximately 20% of their bioactive GM-CSF load within the first 5 days, followed by slow and sustained release of bioactive GM-CSF over the next 10 days (FIG. 8, cryogel A); this release profile was chosen to allow diffusion of the factor through the surrounding tissue to effectively recruit resident DCs. Cryogels can be successfully used for specific spatiotemporal delivery of several drugs, as the incorporation of a second biomolecule (CpG-ODN) did not alter the release profile of GM-CSF over time (FIG. 8, cryogel B). However, slowly degrading PLG microspheres integrated in the scaffolds seem to release GM-CSF much more slowly than pure cryogels (5% vs 24% release at day 14). Hybrid cryogel have been created as a potential carrier for controlled delivery of hydrophobic and/or low molecule weight drugs. Our results not only provide a new strategy for delivery drugs from an injectable 3-D preformed macroporous scaffolds as a sustained-release drug carrier but also open an avenue for the design of new hybrid injectable hydrogels.
  • Example 6: Injectable Biodegradable Cryogels as a Gene Delivery System
  • Nonviral gene delivery systems based upon polycation/plasmid DNA complexes are gaining recognition as an alternative to viral gene vectors for their potential in avoiding immunogenicity and toxicity problems inherent in viral systems. Studies were carried out to determine the feasibility of using a controlled release system based on encapsulated condensed plasmid DNA in injectable cryogels to achieve gene transfer in the surrounding tissues after injection. A unique feature of the cryogel-based gene delivery system is the biodegradability of the polymeric system, which can provide a sustained release of DNA at different rates depending on the polymer, cross-link density, mass fraction, and porosity created during the cryogelation process. Encapsulated DNA complexed with polyethylenimine (PEI), a nondegradable cationic polymer known to be an effective gene carrier, and naked PEI/DNA complexes, which were prepared at a ratio of 7:1 (PEI:DNA) were injected subcutaneously on the lower back of naïve mice using luciferase as a reporter gene (FIG. 9). At 1 day after injection, encapsulated PEI/DNA displayed strong bioluminescence providing the highest transgene expression at ˜10 photons/s, about two-order of magnitude higher than that produced by naked PEI/DNA. After 10 days, the expression levels for naked PEI/DNA were about the same as day 1 but increased by 1 order of magnitude when released in a controllable fashion from the cryogels. Till 29 days, encapsulated PEI/DNA still provided a level of transgene expression at ˜107 photons/s, similar to that observed at previous time points. This level was significantly higher than those offered by naked PEI/DNA.
  • In this study, subcutaneous gene delivery allowed gene expression on the lower back of naïve mice, although the distribution pattern and intensity was vehicle-dependent. Naked PEI/DNA complexes produced limited bioluminescence (signal nearly above background), probably because of its vulnerability to DNAses. However, encapsulated PEI/DNA complexes in cryogels used in this study provided a targeted and sustained high level of gene expression around the injection site for at least 3 weeks. These findings indicate that a 3-D macroporous scaffold may facilitate sustained release and efficient cell transfection of polymer/DNA complexes.
  • In summary, the present approach has demonstrated that cryogels promote gene transfection to surrounding cells in the subcutis of mice, with an efficiency superior in terms of prolonged gene expression to naked DNA. The results establish an injectable delivery system as an effective gene carrier applicable to program or treat targeted cells.
  • OTHER EMBODIMENTS
  • While the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
  • The patent and scientific literature referred to herein establishes the knowledge that is available to those with skill in the art. All United States patents and published or unpublished United States patent applications cited herein are incorporated by reference. All published foreign patents and patent applications cited herein are hereby incorporated by reference. Genbank and NCBI submissions indicated by accession number cited herein are hereby incorporated by reference. All other published references, documents, manuscripts and scientific literature cited herein are hereby incorporated by reference.
  • While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (52)

1-28. (canceled)
29. An injectable cell-compatible highly crosslinked cryogel polymer composition comprising open interconnected pores,
wherein the cryogel comprises at least 75% pores;
wherein the cryogel polymer composition is characterized by shape memory following deformation by compression or dehydration; and
wherein the cryogel composition comprises a crosslinked gelatin polymer or a crosslinked alginate polymer.
30. The cryogel polymer composition of claim 29, wherein the cryogel composition is characterized by shape memory following deformation by compression through a needle.
31. The cryogel polymer composition of claim 29, further comprising a biomolecule.
32. The cryogel polymer composition of claim 31, wherein the biomolecule comprises a small molecule, a nucleic acid, or a protein.
33. The cryogel polymer composition of claim 32, wherein the biomolecule is a protein that recruits a cell into the cryogel composition upon injection into a subject.
34. The cryogel polymer composition of claim 33, wherein the protein is GM-CSF.
35. The cryogel polymer composition of claim 33, wherein the cell is an immune cell.
36. The cryogel polymer composition of claim 35, wherein the immune cell is a dendritic cell.
37. The cryogel polymer composition of claim 29, further comprising an adjuvant.
38. The cryogel polymer composition of claim 37, wherein the adjuvant is cytosine-guanosine oligonucleotide (CpG-ODN).
39. The cryogel polymer composition of claim 29, wherein the gelatin or alginate is acrylated or methacrylated.
40. The cryogel polymer composition of claim 39, wherein the alginate is a methacrylated alginate macromonomer with a concentration of about 1% (w/v).
41. The cryogel polymer composition of claim 29, wherein the cryogel composition is characterized by at least 50% polymer crosslinking.
42. The cryogel polymer composition of claim 41, wherein the cryogel composition is characterized by 50-98% polymer crosslinking.
43. The cryogel polymer composition of claim 29, wherein the cryogel comprises:
(a) at least 90% water in a hydrated state; or
(b) less than 25% water in a compressed state.
44. The cryogel polymer composition of claim 29, further comprising an antigen.
45. The cryogel polymer composition of claim 44, wherein the antigen comprises a cancer antigen.
46. The cryogel polymer composition of claim 45, wherein the cancer antigen comprises a protein, a peptide, or a living attenuated cancer cell.
47. The cryogel polymer composition of claim 45, wherein the cancer antigen comprises a melanoma antigen, or an attenuated melanoma cell.
48. The cryogel polymer composition of claim 47, wherein the attenuated melanoma cell comprises an irradiated melanoma cell.
49. The cryogel polymer composition of claim 29, comprising a eukaryotic cell in one or more of the open interconnected pores.
50. The cryogel polymer composition of claim 49, wherein the eukaryotic cell comprises a live attenuated cancer cell.
51. The cryogel polymer composition of claim 29, wherein the cryogel composition is between 100 μm3 to 100 mm3 in size.
52. The cryogel polymer composition of claim 29, further comprising a microsphere.
53. The cryogel polymer composition of claim 52, wherein the microsphere comprises PLGA.
54. The cryogel polymer composition of claim 52, wherein the microsphere comprises GM-CSF.
55. The cryogel polymer composition of claim 52, wherein the microsphere comprises a drug.
56. The cryogel polymer composition of claim 29, comprising GM-CSF, a pathogen-associated molecular pattern (PAMP), and a tumor antigen.
57. The cryogel polymer composition of claim 29, further comprising a magnetic particle.
58. The cryogel polymer composition of claim 57, wherein the magnetic particle comprises a Fe3O4 nanoparticle or a Fe3O4 micro-particle.
59. The cryogel polymer composition of claim 29, further comprising a mechanophore molecule.
60. The cryogel polymer composition of claim 59, wherein the mechanophore molecule comprises a polydiacetylene liposome.
61. The cryogel polymer composition of claim 29, wherein the cryogel composition comprises macropores having a diameter of 10 μm to 600 μm.
62. The cryogel polymer composition of claim 29, further comprising a polymer selected from the group consisting of heparin, dextran, carob gum, PEG, a PEG derivative, collagen, chitosan, carboxymethylcellulose, pullulan, PVA, PHEMA, PNIPAAm, and PAAc.
63. The cryogel polymer composition of claim 29, wherein the composition is in the shape of a disc, a cylinder, a square, a rectangle, or a string.
64. The cryogel polymer composition of claim 29, wherein the composition has a Young's modulus of 4±2 kPa.
65. A method of minimally invasive administration of a polymer scaffold for agent delivery to a subject, comprising:
administering a cryogel polymer composition of claim 29 to the subject.
66. The method of claim 65, wherein the administration of the cryogel polymer composition is through injecting.
67. The method of claim 65, wherein the method elicits an immune response the subject.
68. The method of claim 67, wherein the cryogel composition comprises a biomolecule that recruits an immune cell to the cryogel composition.
69. The method of claim 67, wherein the cryogel composition further comprises an adjuvant.
70. The method of claim 67, wherein the cryogel composition further comprises an antigen.
71. The method of claim 70, wherein the antigen comprises a cancer antigen.
72. The method of claim 71, wherein the immune response is an anti-cancer immune response.
73. The method of claim 71, wherein the cancer antigen comprises a live attenuated cancer cell.
74. The method of claim 73, wherein cell viability is 90% or more following passage of the cryogel composition through a needle or other delivery apparatus.
75. A syringe comprising:
(i) a needle;
(ii) a reservoir comprising the cryogel composition of claim 29; and
(iii) a plunger.
76. The syringe of claim 75, comprising a 16-gauge, an 18-gauge, a 22-gauge, a 24-gauge, a 26-gauge, a 28-gauge, a 30-gauge, a 32-gauge, or a 34-gauge needle.
77. The syringe of claim 75, comprising an 18 to 30-gauge needle.
78. The syringe of claim 75, wherein the cryogel composition further comprises cells, wherein 90% or more of the cells survive passage of the cryogel composition through a needle.
79. The syringe of claim 78, wherein the cells comprise live attenuated cancer cells.
US16/033,025 2011-04-28 2018-07-11 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration Abandoned US20190076373A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/033,025 US20190076373A1 (en) 2011-04-28 2018-07-11 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US18/095,488 US20230404936A1 (en) 2011-04-28 2023-01-10 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161480237P 2011-04-28 2011-04-28
PCT/US2012/035505 WO2012149358A1 (en) 2011-04-28 2012-04-27 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US201314112096A 2013-12-27 2013-12-27
US16/033,025 US20190076373A1 (en) 2011-04-28 2018-07-11 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US14/112,096 Continuation US10045947B2 (en) 2011-04-28 2012-04-27 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
PCT/US2012/035505 Continuation WO2012149358A1 (en) 2011-04-28 2012-04-27 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/095,488 Continuation US20230404936A1 (en) 2011-04-28 2023-01-10 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Publications (1)

Publication Number Publication Date
US20190076373A1 true US20190076373A1 (en) 2019-03-14

Family

ID=47072783

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/112,096 Active US10045947B2 (en) 2011-04-28 2012-04-27 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US16/033,025 Abandoned US20190076373A1 (en) 2011-04-28 2018-07-11 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US18/095,488 Pending US20230404936A1 (en) 2011-04-28 2023-01-10 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/112,096 Active US10045947B2 (en) 2011-04-28 2012-04-27 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/095,488 Pending US20230404936A1 (en) 2011-04-28 2023-01-10 Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration

Country Status (7)

Country Link
US (3) US10045947B2 (en)
EP (2) EP2701745B1 (en)
JP (4) JP6359966B2 (en)
AU (3) AU2012249456A1 (en)
CA (1) CA2833385C (en)
ES (2) ES2878089T3 (en)
WO (1) WO2012149358A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US11096997B2 (en) 2005-12-13 2021-08-24 President And Fellows Of Harvard College Scaffolds for cell transplantation
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
US11278604B2 (en) 2012-04-16 2022-03-22 President And Fellows Of Harvard College Mesoporous silica compositions comprising inflammatory cytokines comprising inflammatory cytokines for modulating immune responses
WO2022133201A1 (en) * 2020-12-18 2022-06-23 Drexel University Injectable, cross-linkable and subcellular size microfibers for soft tissue repair
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
US10328133B2 (en) 2008-02-13 2019-06-25 President And Fellows Of Harvard College Continuous cell programming devices
WO2009146456A1 (en) 2008-05-30 2009-12-03 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
US9297005B2 (en) 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
EP2461828B1 (en) 2009-07-31 2017-06-21 President and Fellows of Harvard College Programming of cells for tolerogenic therapies
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
WO2012064697A2 (en) 2010-11-08 2012-05-18 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
WO2012148684A1 (en) 2011-04-27 2012-11-01 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
EP2701745B1 (en) 2011-04-28 2018-07-11 President and Fellows of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
US8753309B2 (en) 2011-06-24 2014-06-17 The Invention Science Fund I, Llc Device, system, and method including micro-patterned cell treatment array
US10918765B2 (en) 2013-04-11 2021-02-16 University Of Vermont And State Agricultural College Decellularization and recellularization of whole organs
US20170119892A1 (en) 2014-04-04 2017-05-04 President And Fellows Of Harvard College Refillable drug delivery devices and methods of use thereof
JP6338666B2 (en) * 2014-07-24 2018-06-06 国立研究開発法人物質・材料研究機構 Medical bioabsorbable member and method for producing the same
CA2957970A1 (en) * 2014-08-15 2016-02-18 The Johns Hopkins University Technology Ventures Composite material for tissue restoration
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
AU2015333687B2 (en) 2014-10-14 2021-03-18 Dana-Farber Cancer Institute, Inc. Antibody molecules to PD-L1 and uses thereof
DK3317301T3 (en) 2015-07-29 2021-06-28 Immutep Sas COMBINATION THERAPIES INCLUDING ANTIBODY MOLECULES AGAINST LAYER-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
DE102016203287A1 (en) * 2015-08-07 2017-02-09 Aesculap Ag Mechanophores medical device
EP3337426A4 (en) * 2015-08-17 2019-04-24 The Johns Hopkins University Mesenchymal cell-binding composite material for tissue restoration
GB201521784D0 (en) 2015-12-10 2016-01-27 Univ Birmingham Cell purification
AU2016369537B2 (en) 2015-12-17 2024-03-14 Novartis Ag Antibody molecules to PD-1 and uses thereof
MA44252A (en) 2016-02-16 2018-12-26 Harvard College VACCINES AGAINST PATHOGENIC AGENTS AND THEIR PRODUCTION AND USE PROCESSES
CN110891611B (en) 2017-03-22 2024-03-29 阿森迪斯制药公司 Hydrogel crosslinked hyaluronic acid prodrug compositions and methods
JP2021522938A (en) 2018-05-09 2021-09-02 ザ ジョンズ ホプキンス ユニバーシティ Nanofiber-hydrogel complex for cell and tissue delivery
WO2020116989A1 (en) * 2018-12-07 2020-06-11 한국생명공학연구원 Anti-cancer composition comprising in vivo cell injection chip
WO2021076837A1 (en) * 2019-10-17 2021-04-22 The Texas A&M University System Drug eluting shape memory foam
US11850325B2 (en) 2019-10-24 2023-12-26 Northeastern University Injectable, bioadhesive cryogel scaffolds for biomedical uses
EP4164706A1 (en) 2019-12-04 2023-04-19 Centre Hospitalier Universitaire Vaudois (CHUV) Device and process for tissue-engineering and regenerative medicine
CN112245658A (en) * 2020-10-09 2021-01-22 北京大学 Injectable crystal gel microsphere cell amplification carrier and preparation method thereof
EP4147730A1 (en) 2021-09-09 2023-03-15 ETH Zurich Method of preparing a functional hydrogel material that can be injected
CN114957810B (en) * 2022-05-05 2023-04-18 中国科学院天津工业生物技术研究所 Preparation method of pullulan shape memory material, 4D printing method of pullulan shape memory material and product
CN115804870B (en) * 2022-12-02 2023-12-19 浙江大学 Minimally invasive injection biological stent and manufacturing method and application thereof

Family Cites Families (168)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
GB9206504D0 (en) 1992-03-25 1992-05-06 Jevco Ltd Heteromorphic sponges as wound implants
EP0741580A4 (en) 1993-12-14 2001-07-11 Univ Johns Hopkins Med Controlled release of pharmaceutically active substances for immunotherapy
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US5625048A (en) 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US6329499B1 (en) 1994-11-18 2001-12-11 Neurocrine Biosciences, Inc. Methods for treatment of multiple sclerosis using peptide analogues of human myelin basic protein
US6251396B1 (en) 1994-11-18 2001-06-26 Neurocrine Biosciences, Inc. Methods for treatment of multiple sclerosis using peptide analogs of human myelin basic protein
US6129761A (en) 1995-06-07 2000-10-10 Reprogenesis, Inc. Injectable hydrogel compositions
IL118376A0 (en) 1996-05-22 1996-09-12 Univ Ben Gurion Polysaccharide sponges for cell culture and transplantation
AU3214797A (en) 1996-05-28 1998-01-05 Regents Of The University Of Michigan, The Engineering oral tissues
US6124128A (en) 1996-08-16 2000-09-26 The Regents Of The University Of California Long wavelength engineered fluorescent proteins
US6642363B1 (en) 1996-09-19 2003-11-04 The Regents Of The University Of Michigan Polymers containing polysaccharides such as alginates or modified alginates
US5863551A (en) 1996-10-16 1999-01-26 Organogel Canada Ltee Implantable polymer hydrogel for therapeutic uses
GB2318577B (en) 1996-10-28 2000-02-02 Johnson & Johnson Medical Solvent dried polysaccharide sponges
GB2323282B (en) 1997-03-17 2001-03-07 Bristol Myers Squibb Co Improvements relating to hygiene and medical products
DK0991705T3 (en) 1997-03-31 2004-01-12 Univ Michigan Technology Man W Open-traced, biodegradable matrices
JP4132089B2 (en) 1997-05-30 2008-08-13 オステオバイオロジックス,インコーポレイテッド Fiber reinforced porous biodegradable implantation device
EP1067956B1 (en) 1998-04-03 2007-03-14 University Of Iowa Research Foundation Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
US7427602B1 (en) 1998-05-13 2008-09-23 The Regents Of The University Of Michigan Sustained DNA delivery from structural matrices
FR2780730B1 (en) 1998-07-01 2000-10-13 Corneal Ind INJECTABLE BIPHASIC COMPOSITIONS, ESPECIALLY USEFUL IN RESTORATIVE AND AESTHETIC SURGERIES
EP2311485A1 (en) 1998-07-30 2011-04-20 The Government of the United States of America, as represented by the Secretary of Health and Human Services Thymosin beta 4 promotes wound repair
EP1117444A2 (en) 1998-10-09 2001-07-25 The University Of Michigan Hydrogels and water soluble polymeric carriers for drug delivery
EP1574210B1 (en) 1999-02-26 2016-04-06 Novartis Vaccines and Diagnostics, Inc. Microemulsions with adsorbed macromolecules
WO2000056375A2 (en) 1999-03-19 2000-09-28 The Regents Of The University Of Michigan Mineralization and cellular patterning on biomaterial surfaces
US6767928B1 (en) 1999-03-19 2004-07-27 The Regents Of The University Of Michigan Mineralization and biological modification of biomaterial surfaces
DE60036863T2 (en) 1999-03-25 2008-07-31 Metabolix, Inc., Cambridge Medical devices and uses of polyhydroxyalkanoate polymers
EP1169378A4 (en) 1999-04-09 2004-06-02 Univ Michigan Preparing porous hydrogel products
AU782297B2 (en) 1999-06-30 2005-07-14 Ethicon Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US8084258B2 (en) 1999-07-12 2011-12-27 University Of Basel Manipulation of tissue of organ type using the notch pathway
WO2001010421A1 (en) 1999-08-06 2001-02-15 Board Of Regents, The University Of Texas System Drug releasing biodegradable fiber implant
US7015205B1 (en) 1999-10-18 2006-03-21 St. Vincent's Hospital And Medical Center Of New York Melanoma vaccine and methods of making and using same
WO2001035932A2 (en) 1999-11-18 2001-05-25 The Regents Of The University Of Michigan Sustained drug delivery from structural matrices
US6790840B1 (en) 1999-11-26 2004-09-14 The Regents Of The University Of Michigan Reversibly cross-linked hydrogels
US20020061587A1 (en) 2000-07-31 2002-05-23 Piero Anversa Methods and compositions for the repair and/or regeneration of damaged myocardium
EP1311657A2 (en) 2000-08-21 2003-05-21 Rice University Tissue engineering scaffolds promoting matrix protein production
ATE475887T1 (en) 2000-09-09 2010-08-15 Univ New York State Res Found METHOD AND COMPOSITIONS FOR ISOLATION OF METASTATIC CANCER CELLS AND APPLICATION FOR MEASURING THE METASTATIC POTENTIAL OF A CANCER
US6748954B2 (en) 2000-10-27 2004-06-15 The Regents Of The University Of Michigan Drug release from polymer matrices through mechanical stimulation
JPWO2002040071A1 (en) 2000-11-14 2004-03-18 上田 実 Composition for forming bone or periodontal tissue, and injection for forming bone or periodontal tissue
JP2004520043A (en) 2001-01-24 2004-07-08 シェーリング コーポレイション Chemokines as adjuvants of the immune response
WO2002068605A2 (en) 2001-02-26 2002-09-06 The Regents Of The University Of California Non-oligomerizing tandem fluorescent proteins
JP2002263981A (en) 2001-03-14 2002-09-17 Murata Mach Ltd Suction control device for plate suction-lifting device
US6656488B2 (en) 2001-04-11 2003-12-02 Ethicon Endo-Surgery, Inc. Bioabsorbable bag containing bioabsorbable materials of different bioabsorption rates for tissue engineering
US20030082806A1 (en) 2001-04-27 2003-05-01 Xcyte Therapies, Inc. Maturation of antigen-presenting cells using activated T cells
AU2002345691C1 (en) 2001-06-13 2008-07-24 Massachusetts Institute Of Technology In vivo bioreactors
US7297343B2 (en) 2001-07-31 2007-11-20 Biosurface Engineering Technologies, Inc. Bioactive medical films
AU2002361468A1 (en) 2001-08-14 2003-03-18 The Government Of The United States Of America As Represented By The Secretary Of Health And Human S Method for rapid generation of mature dendritic cells
SE0103404D0 (en) * 2001-10-12 2001-10-12 Protista Internat Ab Macroporous gel, ITS preparation and its use
CA2412012C (en) 2001-11-20 2011-08-02 Ed. Geistlich Soehne Ag Fuer Chemische Industrie Resorbable extracellular matrix containing collagen i and collagen ii for reconstruction of cartilage
AU2002361902A1 (en) 2001-12-31 2003-07-24 Ares Medical, Inc. Hemostatic compositions and methods for controlling bleeding
US7575759B2 (en) 2002-01-02 2009-08-18 The Regents Of The University Of Michigan Tissue engineering scaffolds
US20070026518A1 (en) 2005-03-29 2007-02-01 The Regents Of The University Of California Controlling stem cell destiny with tunable matrices
US6811777B2 (en) 2002-04-13 2004-11-02 Allan Mishra Compositions and minimally invasive methods for treating incomplete connective tissue repair
WO2003089506A1 (en) 2002-04-22 2003-10-30 Purdue Research Foundation Hydrogels having enhanced elasticity and mechanical strength properties
US7371734B2 (en) 2002-04-22 2008-05-13 Bioniche Life Sciences Inc. Oligonucleotide compositions and their use for the modulation of immune responses
WO2003096972A2 (en) 2002-05-13 2003-11-27 Massachusetts Institute Of Technology Angiogenesis and cardiac tissue engineering with peptide hydrogels and related compositions and methods of use thereof
AUPS312602A0 (en) 2002-06-21 2002-07-18 James Cook University Organ arrest, protection, preservation and recovery
US7332160B2 (en) 2002-07-12 2008-02-19 Boston Scientific Scimed, Inc. Medical device and method for tissue removal and repair
US20040136968A1 (en) 2002-09-27 2004-07-15 Verigen Ag Autologous cells on a support matrix for tissue repair
US20040063206A1 (en) 2002-09-30 2004-04-01 Rowley Jon A. Programmable scaffold and method for making and using the same
WO2004030706A2 (en) 2002-10-01 2004-04-15 Law Peter K Bioactive implants
US8940292B2 (en) 2003-01-28 2015-01-27 Wake Forest University Health Sciences Enhancement of angiogenesis to grafts using cells engineered to produce growth factors
SE0301109D0 (en) 2003-04-14 2003-04-14 Mallen Huang Nucleotide vaccine composition
EP1620140B1 (en) 2003-05-05 2013-10-09 Ben-Gurion University Of The Negev Research And Development Authority Injectable cross-linked polymeric preparations and uses thereof
EP1475434A1 (en) 2003-05-09 2004-11-10 Oncoscience AG Method for storing tumor cells
EP1641424A4 (en) 2003-07-09 2007-05-02 Vaxdesign Corp Programmed immune responses using a vaccination node
US20060264380A1 (en) 2003-07-21 2006-11-23 Mats Hellstrom Compounds and Methods for Promoting Angiogenesis
JP2007500721A (en) 2003-07-31 2007-01-18 エンダセア,インク. Methods and compositions for generating antigenic responses
GB0317999D0 (en) 2003-07-31 2003-09-03 Univ Liege Improvements in or relating to drug delivery systems
EP1664168B1 (en) 2003-08-29 2008-12-03 Mayo Foundation for Medical Education and Research Hydrogel porogens for fabricating biodegradable scaffolds
EP2481422A3 (en) 2003-09-03 2013-04-03 Dendritherapeutics, Inc. Multiplex vaccines
EP1682159A4 (en) 2003-10-16 2010-07-21 Stephen John Ralph Immunomodulating compositions and uses therefor
US8162925B2 (en) 2003-11-07 2012-04-24 Carnegie Mellon University Robot for minimally invasive interventions
CA2448995A1 (en) 2003-11-12 2005-05-12 James Keenan Device and method for attracting diseased cells and foreign substances
JP2005160669A (en) 2003-12-02 2005-06-23 Olympus Corp Manufacturing method of biological tissue prosthesis
WO2005072088A2 (en) 2003-12-11 2005-08-11 Sciperio, Inc. Immunotherapy compositions, method of making and method of use thereof
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
US7192693B2 (en) 2004-02-24 2007-03-20 University Of Washington Methods for photopatterning hydrogels
US7235592B2 (en) 2004-10-12 2007-06-26 Zimmer Gmbh PVA hydrogel
US7790699B2 (en) 2004-10-12 2010-09-07 Fmc Biopolymer As Self-gelling alginate systems and uses thereof
US7999161B2 (en) 2005-01-22 2011-08-16 Alexander Oraevsky Laser-activated nanothermolysis of cells
US8343527B2 (en) 2005-03-23 2013-01-01 Mahrokh Dadsetan Photocrosslinkable oligo(poly (ethylene glycol) fumarate) hydrogels for cell and drug delivery
US20070003595A1 (en) 2005-04-19 2007-01-04 Shaopeng Wang Three dimensional micro-environments and methods of making and using same
US8828433B2 (en) * 2005-04-19 2014-09-09 Advanced Cardiovascular Systems, Inc. Hydrogel bioscaffoldings and biomedical device coatings
CN103285392A (en) 2005-04-26 2013-09-11 卫材R&D管理株式会社 Compositions for cancer immunotherapy and use thereof
WO2006119619A1 (en) 2005-05-06 2006-11-16 Replicor Inc. Oligonucleotides inhibiting cell proliferation
ITPI20050071A1 (en) 2005-06-20 2006-12-21 Giuseppe Calvosa COMPOUND COMPOSITION FOR FABRIC REPLACEMENT / REGENERATION
US7645742B2 (en) 2005-06-22 2010-01-12 Advocare International, L.P. Composition for enhancing cellular energy
CN1302050C (en) 2005-07-07 2007-02-28 复旦大学 Interpenetrating network polymer type super porous aquogel, its prepn. method and application
US20090017096A1 (en) 2005-08-15 2009-01-15 Anthony Lowman Porous non-biodegradable hydrogel admixed with a chemoattractant for tissue replacement
BRPI0503817A (en) 2005-09-12 2007-05-15 Cristalia Prod Quimicos Farm immunogenic complex formed by nanostructured silica mesoporous encapsulated vaccine antigens
US20070081972A1 (en) 2005-09-30 2007-04-12 The University Of Iowa Research Foundation Polymer-based delivery system for immunotherapy of cancer
US20070116680A1 (en) 2005-11-18 2007-05-24 Rensselaer Polytechnic Institute Stem cells within gel microenvironments
DK2119450T3 (en) 2005-11-29 2013-05-06 Actogenix Nv Induction of mucosal tolerance to pancreatic islet beta cell autoantigens
KR100687281B1 (en) 2005-11-30 2007-02-27 한국과학기술연구원 Injectable thermosensitive pluronic hydrogels coupled with bioactive materials for tissue regeneration and preparation method thereof
ES2804472T3 (en) 2005-12-13 2021-02-08 Harvard College Structures for cell transplantation
EP1806395A1 (en) 2006-01-06 2007-07-11 Stichting Sanquin Bloedvoorziening Maturation of dendritic cells
US20070178159A1 (en) 2006-01-30 2007-08-02 Alza Corporation In-Situ Forming Porous Scaffold
US20100015709A1 (en) 2006-02-10 2010-01-21 Trustees Of The University Of Pennsylvania Regulating Stem Cell Differentiation By Controlling 2D and 3D Matrix Elasticity
US20070190646A1 (en) 2006-02-10 2007-08-16 The Trustees Of The University Of Pennsylvania Regulating stem cell differentiation by controlling matrix elasticity
WO2007097710A1 (en) 2006-02-27 2007-08-30 Agency For Science, Technology And Research Curable bone cement
US9456860B2 (en) 2006-03-14 2016-10-04 Kci Licensing, Inc. Bioresorbable foaming tissue dressing
GB0605521D0 (en) 2006-03-18 2006-04-26 Isis Innovation Adjuvant
US20090297579A1 (en) 2006-06-01 2009-12-03 Massachusetts Institute Of Technology Control of Cells and Cell Multipotentiality in Three Dimensional Matrices
WO2007146319A2 (en) 2006-06-13 2007-12-21 Symphony Medical, Inc. Methods and apparatus for using polymer-based beads and hydrogels for cardiac applications
US20070298067A1 (en) 2006-06-22 2007-12-27 Boston Scientific Scimed, Inc. Control release drug coating for medical devices
EP2041175A4 (en) 2006-06-23 2011-03-16 Augmenta Biolog Llc Targeted immune conjugates
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
KR100802139B1 (en) 2006-08-08 2008-02-11 한국생명공학연구원 Gold nanocages containing magnetic nanoparticles
KR100818708B1 (en) 2006-08-18 2008-04-01 주식회사 하이닉스반도체 Semiconductor device manufacturing method including cleaning surface layer
ITMI20061726A1 (en) * 2006-09-11 2008-03-12 Fidia Farmaceutici CROSSLINKATI DERIVATIVES BASED ON HYALURONIC ACID RETICULATED VIA CLICK CHEMISTRY
WO2008036393A1 (en) 2006-09-21 2008-03-27 Purdue Research Foundation Collagen preparation and method of isolation
AU2007317809B2 (en) 2006-11-09 2013-09-12 Kci Licensing Inc. Porous bioresorbable dressing conformable to a wound and methods of making same
WO2008086486A1 (en) 2007-01-10 2008-07-17 The Regents Of The University Of Michigan Porous objects having immobilized encapsulated biomolecules
US8354110B2 (en) 2007-03-07 2013-01-15 Uti Limited Partnership Compositions and methods for the prevention and treatment of autoimmune conditions
CA2684578A1 (en) 2007-03-21 2008-09-25 Id Biomedical Corporation Of Quebec Chimeric antigens
US8501905B2 (en) 2007-03-22 2013-08-06 The Regents Of The University Of California Synthetic cell platforms and methods of use thereof
EP1975230A1 (en) * 2007-03-30 2008-10-01 Capsulution Nanoscience AG Method for forming glucose sensing micro-particles, use of micro-particles, and kit
WO2008131074A1 (en) 2007-04-19 2008-10-30 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of toll-like receptor-4-associated disorders
KR20100035643A (en) 2007-06-05 2010-04-05 노파르티스 아게 Induction of tolerogenic phenotype in mature dendritic cells
JP2010531138A (en) 2007-06-13 2010-09-24 ラ ホヤ インスティテュート フォア アラージー アンド イムノロジー Regulatory T cells and methods of production and use thereof
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
CN101855337B (en) 2007-06-29 2017-10-31 船木真理 Soft gel systems for carrying out stem cell regulation
US20090123467A1 (en) 2007-07-31 2009-05-14 The Johns Hopkins University Polypeptide-Nucleic Acid Conjugate for Immunoprophylaxis or Immunotherapy for Neoplastic or Infectious Disorders
US20090192079A1 (en) 2007-10-09 2009-07-30 Genzyme Corporation Prolonged delivery of heparin-binding growth factors from heparin-derivatized collagen
KR100900837B1 (en) 2007-12-07 2009-06-04 (주)두비엘 A powerful vaccine composition comprising lipopeptide and poly(i:c) as an adjuvant
EP2072617A1 (en) 2007-12-12 2009-06-24 Trimed Biotech GmbH Method for producing dendritic cells
DE102008008522A1 (en) 2008-02-11 2009-08-13 Magforce Nanotechnologies Ag Implantable nanoparticle-containing products
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
WO2011063336A2 (en) 2009-11-20 2011-05-26 President And Fellows Of Harvard College Secondary site of antigen stimulation for therapeutic vaccination
US10328133B2 (en) 2008-02-13 2019-06-25 President And Fellows Of Harvard College Continuous cell programming devices
US20090238853A1 (en) 2008-03-21 2009-09-24 3D Biotek, Llc Hybrid Biomedical Device Fabricated From Biomaterials and Coated With a Natural Extra Cellular Matrix (ECM) Coating
WO2009146456A1 (en) 2008-05-30 2009-12-03 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
WO2009155583A1 (en) 2008-06-19 2009-12-23 The Trustees Of The University Of Pennsylvania Biomaterials for tissue replacement
AU2009290442A1 (en) * 2008-09-15 2010-03-18 Albert Mihranyan Vinyl alcohol co-polymer cryogels, vinyl alcohol co-polymers, and methods and products thereof
US8889124B2 (en) 2008-09-25 2014-11-18 The Board Of Trustees Of The Leland Stanford Junior University Tolerogenic populations of dendritic cells
KR101132732B1 (en) 2008-11-26 2012-04-06 한국과학기술연구원 Intelligent porous biodegradable polymer scaffolds for in situ tissue regeneration and method for the preparation thereof
US8273373B2 (en) 2008-12-30 2012-09-25 Case Western Reserve University Photocrosslinked biodegradable hydrogel
US9297005B2 (en) 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
US8551749B2 (en) 2009-04-23 2013-10-08 The Invention Science Fund I, Llc Device including bone cage and method for treatment of disease in a subject
EP2461828B1 (en) 2009-07-31 2017-06-21 President and Fellows of Harvard College Programming of cells for tolerogenic therapies
CN101655611B (en) 2009-09-11 2011-06-08 中国科学院长春应用化学研究所 Preparation method of inverse opal hydrogel photonic crystal with double layer hybridized structure
CN102656119B (en) 2009-12-18 2015-11-25 花王株式会社 The manufacture method of mesoporous silica particles
JP5603063B2 (en) 2009-12-21 2014-10-08 花王株式会社 Method for producing composite silica particles
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
JP2013522246A (en) 2010-03-15 2013-06-13 フェロサン メディカル デバイシーズ エイ/エス Method for promoting hemostasis and / or wound healing
US20110300186A1 (en) 2010-04-14 2011-12-08 Battelle Memorial Institute Functionalized Nano- and Micro-materials for Medical Therapies
WO2011150258A1 (en) 2010-05-26 2011-12-01 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
GB201009273D0 (en) 2010-06-03 2010-07-21 Glaxosmithkline Biolog Sa Novel vaccine
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
CN103118678A (en) 2010-07-16 2013-05-22 约翰斯·霍普金斯大学 Methods and compositions for cancer immunotherapy
CA2807483C (en) 2010-08-04 2019-02-26 Georgia Tech Research Corporation Devices, systems, and methods for excavating cancer cells
KR102155383B1 (en) 2010-10-06 2020-09-11 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Injectable, pore-forming hydrogels for materials-based cell therapies
WO2012064697A2 (en) 2010-11-08 2012-05-18 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
WO2012148684A1 (en) 2011-04-27 2012-11-01 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
EP2701745B1 (en) 2011-04-28 2018-07-11 President and Fellows of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
EP2723741B8 (en) 2011-06-24 2016-09-21 ArQule, Inc. Substituted imidazopyridinyl-aminopyridine compounds
CN109125718A (en) 2012-01-13 2019-01-04 哈佛学院董事会 The control transmitting of TLR agonist in topology convergence device
CA2870309C (en) 2012-04-16 2024-02-20 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
EP3137105A4 (en) 2014-04-30 2017-12-27 President and Fellows of Harvard College Combination vaccine devices and methods of killing cancer cells
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
US20180117171A1 (en) 2015-04-01 2018-05-03 President And Fellows Of Harvard College Immunoconjugates for programming or reprogramming of cells

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11096997B2 (en) 2005-12-13 2021-08-24 President And Fellows Of Harvard College Scaffolds for cell transplantation
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
US11278604B2 (en) 2012-04-16 2022-03-22 President And Fellows Of Harvard College Mesoporous silica compositions comprising inflammatory cytokines comprising inflammatory cytokines for modulating immune responses
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
WO2022133201A1 (en) * 2020-12-18 2022-06-23 Drexel University Injectable, cross-linkable and subcellular size microfibers for soft tissue repair

Also Published As

Publication number Publication date
JP6741723B2 (en) 2020-08-19
JP7097629B2 (en) 2022-07-08
US20140112990A1 (en) 2014-04-24
EP2701745A4 (en) 2014-12-03
CA2833385C (en) 2020-10-27
EP2701745B1 (en) 2018-07-11
EP2701745A1 (en) 2014-03-05
JP2020143147A (en) 2020-09-10
ES2685327T3 (en) 2018-10-08
AU2017204078A1 (en) 2017-07-06
AU2019201669A1 (en) 2019-04-04
CA2833385A1 (en) 2012-11-01
ES2878089T3 (en) 2021-11-18
US20230404936A1 (en) 2023-12-21
US10045947B2 (en) 2018-08-14
JP2014514336A (en) 2014-06-19
JP2018162285A (en) 2018-10-18
AU2012249456A1 (en) 2013-10-31
WO2012149358A1 (en) 2012-11-01
EP3417876B1 (en) 2021-03-31
AU2017204078B2 (en) 2019-04-04
EP3417876A1 (en) 2018-12-26
JP2022092026A (en) 2022-06-21
AU2019201669B2 (en) 2021-04-01
JP6359966B2 (en) 2018-07-18

Similar Documents

Publication Publication Date Title
US20230404936A1 (en) Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US20210205233A1 (en) Injectable cryogel vaccine devices and methods of use thereof
JP7065806B2 (en) Scaffolding for cell transplantation
Andorko et al. Designing biomaterials with immunomodulatory properties for tissue engineering and regenerative medicine
JP5690143B2 (en) Continuous cell programming device
Conova et al. A pilot study of poly (N-isopropylacrylamide)-g-polyethylene glycol and poly (N-isopropylacrylamide)-g-methylcellulose branched copolymers as injectable scaffolds for local delivery of neurotrophins and cellular transplants into the injured spinal cord
EP3691738A1 (en) Implantable bioreactor and methods for making and using same
KR102194155B1 (en) Thermo-responsive biomaterial comprising thermo-responsive protein conjugated-mussel adhesive protein
Bhatt et al. Polymeric scaffolds for antitumor immune cell priming
Bhatta et al. Polymeric scaffolds for antitumor immune cell priming
Vanderburgh et al. A Novel Platform Technology for Cytosolic Peptide Delivery with Endosomolytic Nano-Polyplexes Applied to Vascular Graft Intimal Hyperplasia Evans BC, Hocking KM, Osgood MJ, Voskresensky I, Dmowska J, Kilchrist KV, Brophy CM, Duvall CL Autologous vein grafts are commonly used for coronary and peripheral artery bypass but have a high incidence of intimal hyperplasia (IH) and failure. Here, a nano-polyplex (NP) approach is presented that

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: PRESIDENT AND FELLOWS OF HARVARD COLLEGE, MASSACHU

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BENCHERIF, SIDI A.;EDWARDS, DAVID A.;MOONEY, DAVID J.;AND OTHERS;SIGNING DATES FROM 20130207 TO 20130212;REEL/FRAME:047963/0792

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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