WO2015187098A1 - Matière et structures destinées à l'adminisatration de cellules - Google Patents

Matière et structures destinées à l'adminisatration de cellules Download PDF

Info

Publication number
WO2015187098A1
WO2015187098A1 PCT/SG2015/050142 SG2015050142W WO2015187098A1 WO 2015187098 A1 WO2015187098 A1 WO 2015187098A1 SG 2015050142 W SG2015050142 W SG 2015050142W WO 2015187098 A1 WO2015187098 A1 WO 2015187098A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
microcapsules
biologically active
collagen
negatively charged
Prior art date
Application number
PCT/SG2015/050142
Other languages
English (en)
Inventor
Anna Maria BLOCKI
Kishore Kumar Bhakoo
Sebastian Beyer
Original Assignee
Agency For Science, Technology And Research
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 Agency For Science, Technology And Research filed Critical Agency For Science, Technology And Research
Priority to SG11201602828YA priority Critical patent/SG11201602828YA/en
Publication of WO2015187098A1 publication Critical patent/WO2015187098A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/58Materials at least partially resorbable by the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5036Polysaccharides, e.g. gums, alginate; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5021Organic macromolecular compounds
    • A61K9/5052Proteins, e.g. albumin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5089Processes
    • 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
    • A61L27/14Macromolecular materials
    • A61L27/26Mixtures of macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • A61L27/3834Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem 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
    • 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
    • A61L2300/62Encapsulated active agents, e.g. emulsified droplets
    • A61L2300/622Microcapsules
    • 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
    • A61L2430/00Materials or treatment for tissue regeneration
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material

Definitions

  • the present invention relates to biochemistry.
  • the present invention relates to micro- or macro-structures made of polymers, methods of using such structures, uses of such structures and methods of manufacturing thereof.
  • the issue of cell delivery into tissue can be aided with the use of a suitable encapsulation biomaterial.
  • the microcapsules show superior mechanical stability, allowing for a better protection of injected cells, and hence, better long-term retention of cells within the host tissue.
  • microcapsules were employed to deliver allogeneic or xenogeneic cells. Designed to be non-degradable and semipermeable, these microcapsules were meant to protect encapsulated cells from the host immune system.
  • alginate is known to be immunogenic due to presence of protein impurities in the polysaccharide hydrogel, and has been shown to induce a foreign body response (FBR), which can result in collagenous encapsulation and therefore isolation of encapsulated cells from the surrounding tissues. This immunogenic response is not restricted to alginates alone. In general, microcapsules for such applications fail in the long-term, due to rejection.
  • microstructure suitable for encapsulating biologically active materials.
  • the microstructure comprises a negatively charged polymer.
  • a macrostructure suitable for encapsulating biologically active materials comprises a negatively charged polymer.
  • the method comprises emulsifying methods, polymerization methods, forming methods, moulding methods, casting methods and coating methods.
  • this other method comprises the steps of: (i) mixing the biologically active material with a biocompatible, biodegradable polymer, at least one structural component capable of emulating ECM and a negatively charged polymer, resulting in a biomaterial mix; (ii) adding said biomaterial mix to an emulsifier to form an emulsion; (iii) isolating the formed structures from the emulsion; and optionally (iv) incubating the formed microstructures with a polymerization agent.
  • a method of manufacturing microcapsules/macrocapsules as described herein using an emulsion based method comprises the steps of (i) mixing an aqueous phase, wherein the aqueous phase comprises a solution of biologically active material and/or cellular cargo with an oil or non-aqueous phase containing a emulsifier; (ii) forming microcapsules/macrocapsules by shaking, membrane emulsification, droplet generator, microfluidics or any other method that is essentially emulsion based or forming the aqueous phase containing biomaterial components and cellular cargo into droplets that subsequently solidify in liquid phase, air phase or at the solid/air or solid liquid interface; (iii) generating desired shapes and sizes using an extrusion or mould-based method; (iv) allowing the aqueous phase containing biologically active material to solidify prior to a process that leads to smaller units; (v) isol
  • Fig. 1 shows the extent of cellular support provided by the various microcapsules.
  • live-dead cell staining of encapsulated cells is shown in a) and c), which show encapsulation at day 1 and day 3 post-encapsulation, respectively.
  • Quantification of live-to-dead cell staining area ratio at the respective days are shown as bar graphs in b) and d), which show results from day 1 and day 3 post-encapsulation, respectively.
  • Aga short abbreviation for agarose. *p ⁇ 0.05 indicates significant difference from all other conditions.
  • Fig. 1 shows addition of extracellular matrix (ECM) proteins and/or dextran sulfate (DxS) within microcapsules support cell survival.
  • ECM extracellular matrix
  • DxS dextran sulfate
  • Fig. 2 shows results on the studies of the fibrillogenesis caused by dextran sulfate (DxS).
  • a) shows a line graph of the kinetics of collagen fibrillogenesis in the presence of various concentrations of dextran sulfated (DxS), as followed by turbidimetry
  • b) shows a bar graph showing the nucleation rate and elongation rate of collagen as calculated from time-resolved spectrophotometry data
  • c) shows images of collagen hydrogels assembly in the presence of dextran sulfate (DxS) as taken by 3D confocal reflexion microscopy.
  • Fig. 2 shows the presence of dextran sulfate (DxS) influences the architecture of gels.
  • Fig. 3 shows the enhancement of cell-microenvironment interactions by dextran sulfate (DxS) and collagen I.
  • DxS dextran sulfate
  • a) shows a phase contrast and fluorescent microscopy of microcapsules containing FITC-labelled dextran sulfate (DxS).
  • FITC-labelled dextran sulfate (DxS) appears as bright spots in black background;
  • b) shows phase contrast and fluorescent microscopy of microcapsules containing phalloidin-stained fibroblasts. Stained cells appear as bright spots in black background.
  • Fig. 3 shows interactions of dextran sulfate (DxS) with collagen I within the microcapsules resulting in a stronger cell spreading.
  • Fig. 4 shows the investigation into the properties and cell functionalities within microcapsules.
  • a) shows the result of cell counting kit-8 (CCk-8) colorimetric assay (a metabolic assay) looking into the long-term cell survival and cell proliferation of mesenchymal stem cells over three weeks of culture.
  • Fig. 4a shows mesenchymal stem cells can proliferate and have long-term survival in the Col-Fb-DxSlOO microcapsules (i.e. microcapsules composed of collagen I, fibrin and 100 ⁇ g/ml dextran sulfate);
  • b) shows phase contrast pictures at various magnifications of microcapsules showing out-migrating cells;
  • c) shows microcapsules size distribution as diameter measurements.
  • Fig. 4 shows the various microcapsules size and pore-size of microcapsules that are composed of collagen I, fibrin and 100 ⁇ g/ml dextran sulfate (i.e. Col-Fb-DxS
  • Fig. 5 shows the investigation of the functionality of cells encapsulated in microcapsules.
  • a) shows the result of an enzyme-linked immunosorbent assay (ELISA) study on the amount of vascular endothelial growth factor (VEGF) in the culture medium of encapsulated mesenchymal stem cells, which were either non-treated or treated with 10 ⁇ ciclopirox olamine (CPX);
  • a) shows the microscope images of differentiation of out-migrating mesenchymal stem cells into adipocytes and osteoblasts;
  • c) confocal microscopy images of encapsulated mesenchymal stem cells four days in culture in the presence of ascorbic acid.
  • FIG. 5 shows encapsulation of mesenchymal stem cells (MSCs) in the microcapsule as described in the Example section allows the mesenchymal stem cells to remain functional and are capable of modifying their microenvironment within the microcapsules.
  • Fig. 6 shows studies on the in vivo functionality of microcapsules Col-Fb-DxS lOO.
  • a) shows histological hematoxylin and eosin stain (H&E) staining of microcapsules immediately after injection into rat thigh muscle;
  • b) shows phase contrast and fluorescent microscopy images of TRITC-labelled agarose containing microcapsules as well as H&E staining four weeks post injection. Fluorescent positive cells are seen as bright spots in panels labelled TRITC-agarose or darkened area in phase-contrast panels (bottom three panels).
  • FIG. 6 shows Col-Fb-DxS lOO microcapsules exhibit sufficient mechanical stability for intramuscular injection; c) shows immunohistochemistry images of rat calf muscles injected with TRITC-labelled agarose containing microcapsules. Grey spots: DAPI (nuclei staining), Bright spots: immunostaining against CD l ib, iNOS, CD206 and collagen I (Col I), collagen III (Col III) and rat endothelial cell antigen 1 (Reca-1). White arrows: phagocytized agarose. Immunohistochemistry demonstrated that many of the infiltrated cells expressed CDl lb, confirming their monocytic origin (first four images labelled "CDl lb").
  • FIG. 6 shows Col- Fb-DxSlOO microcapsules degraded slowly in vivo, and does not induce a fibrotic foreign body response but rather induced a 'healing' environment around the implant.
  • Fig. 7 shows the results of in vivo transplantation test.
  • Fig. 7 shows a magnetic resonance imaging (MRI) image of Tl-weighted short-axis analysis (T1W1, "spin-lattice” relaxation time, a basic pulse sequence in MRI) of rat left ventricle after implantation of microcapsules.
  • T1W1 Tl-weighted short-axis analysis
  • T1W1 spin-lattice” relaxation time, a basic pulse sequence in MRI
  • Fig. 8 shows a net negatively charged polycarbohydrate, such as sulfated dextran (branched glycan) polymer.
  • One of the methods known in the art of transplantation includes the use of a suitable encapsulation biomaterial.
  • encapsulation technologies known in the art can often trigger host immune response or unable to provide suitable microenvironment for the encapsulated cells to thrive.
  • microstructure suitable for encapsulating biologically active materials.
  • the microstructure comprises a negatively charged polymer.
  • the term "microstructure” refers to a construct, structure or arrangement of the present disclosure that may have a diameter of about ⁇ to about 2mm.
  • the microstructure may be a microcapsule, film, patch, bead, capsule, hydrogel, gel, microbead, injectable liquid, and moulded microstructure.
  • the microstructure may be a microcapsule that may have a diameter of between about 1 ⁇ to about 2000 ⁇ , for example, the microcapsule may be between about 1 ⁇ to about 50 ⁇ , or between about 50 ⁇ to about 100 ⁇ , or between about 100 ⁇ to about 200 ⁇ , or between about 200 ⁇ to about 400 ⁇ , or between about 400 ⁇ to about 800 ⁇ , or between about 800 ⁇ to about 1000 ⁇ , or between about 1000 ⁇ to about 1500 ⁇ , or between about 1500 ⁇ to about 2000 ⁇ .
  • the microcapsule 4c may have a diameter of about 60 ⁇ to about 80 ⁇ , or about 80 ⁇ to about 100 ⁇ , or about 100 ⁇ to about 120 ⁇ , or about 120 ⁇ to about 140 ⁇ , or about 140 ⁇ to about 160 ⁇ .
  • the microcapsule may have a diameter of about 100 ⁇ .
  • the structure as described herein may be provided in large scale that is not bound by the actual dimension of the structure.
  • a macrostructure suitable for encapsulating biologically active materials comprises a negatively charged polymer.
  • the term "macrostructure” refers to a construct, structure or arrangement of the present disclosure that may have a diameter of about 2mm to about 100mm.
  • the macrostructure may be a macrocapsule, film, patch, bead, capsule, hydrogel, gel, beads such as macrobead, injectable liquid and moulded microstructure.
  • the macrostructure may be a macrocapsule that may have a diameter of between about 2 mm to about 20mm, or between about 20 mm to 1000 mm.
  • the macrocapsule may have between about 2 mm to about 4mm, or between about 4 mm to about 8 mm, or between about 8 mm to about 12 mm, or between about 12 mm to about 16 mm, or between about 16 mm to about 20 mm or between about 20 mm to about 100 mm or between about 100 mm to about 1000 mm.
  • the term "encapsulating” refers to a process for coating biologically active materials or placing the biologically active materials into an encasement (casing or capsules) so that the coating (encasement, casing or capsule) will encase the biologically active materials, at least to some extent, in such a manner that the biologically active materials does not have direct cellular contact with the cellular environment the microstructure has been placed into.
  • the structure would prevent the encapsulated biologically active material from having direct cellular contacts with the cells of the tissue in which the encapsulated biologically active material have been placed (grafted or injected) into.
  • the encapsulation does not prevent extracellular fluid to come in contact with the biologically active material.
  • the encapsulated biologically active material is still in fluid contact with the extracellular fluid.
  • the phrase "negatively charged polymer” refers to a polymer that has a negative charge when located in an aqueous solution or an aqueous system such as blood or an extracellular fluid.
  • the term “polymer” refers to a material that is a homopolymer, copolymer, terpolymer or the like.
  • the negatively charged polymer may be capable of emulating glycosaminoglycan. That is, the negatively charged polymer may be mimicking the properties of glycosaminoglycan in an extracellular matrix.
  • negatively charged polymer such as glycosaminoglycan (GAG)
  • GAG glycosaminoglycan
  • the negatively charged polymer may not be capable of providing mechanical stability to the structure.
  • the negatively charged polymer may not have gelatinous properties. That is, in one example, the negatively charged polymer may not be able to form gelatinous structure.
  • negatively charged polymer binds to extracellular matrix proteins such as collagen I and affects how these proteins are presented to the cells.
  • the negatively charged polymer may include, but is not limited to, glycosaminoglycan-like (GAG) polysaccharides, non-glycosaminoglycan (non-GAG) polysaccharides, non-polysaccharides, amino acids, poly amino acids, macromolecules (such as glycosaminoglycan) and the like.
  • macromolecule refers to a molecule of high relative molecular mass, the structure of which essentially comprises the multiple repetitions of units derived, actually or conceptually, from molecules of low relative molecular mass.
  • the lower size limit for a macromolecule can be further defined as to have sufficient monomeric units to interact or bind with at least one component of the material by hydrogen bonding, electrostatic or steric interaction.
  • the minimum size of heparan sulfate fragments to mediate interaction of vascular endothelial growth factor (VEGF) with its receptor is about an octa-saccharide for stable interaction, which is larger than the oligomere size to provide stable electrostatic interaction— tri or tetra saccharide).
  • the upper size limit of a macromolecule can be derived from functional parameters - solubility in aqueous phase and viscosity. Typically, macromolecules are smaller than 1000 kDa. In exceptional cases, mainly synthetic polymers, the molecular weight may reach a maximum of 2500 kDa.
  • macromolecules with a molecular mass greater than 5000 kDa may not be suitable for use in the structure as described herein.
  • poly amino acids refers to proteins generally with a molecular mass between 5 kDa and 500 kDa, with or without a negative charge. These could be further defined as natural proteins and peptides, synthetically modified natural proteins and peptides or fully synthetic proteins, peptides, and poly amino acids whose isoelectric point indicates a negative net-charge at a pH range suitable for the intended application.
  • negatively charged poly amino acids are poly glutamic acids, poly aspartic acids and poly amino acids that have a significant proportion of these amino acids to render the net charge negative at pH range between 3-9.
  • Negatively charged macromolecules, polymers and macromolecules may also fall under this definition of poly amino acids, resulting in that at least a fraction of the monomeric component is derived from synthetic materials (e.g. non-amino acids). Accordingly, in one example, the negatively charged polymer may be a partially synthetic or fully synthetic polymer.
  • the macromolecules may include, but are not limited to, oligomers and peptides with a molecular mass of less than 5 kDa, oligosaccharides, di-,tri- and tetrasaccharides, tetra-, penta-, hexa-, hepta-, octa- to icosa-polymers and polymers greater in monomer units, low molecular weight heparin fractions, peptides, sulfonated carbohydrates, carbohydrate phosphates, nucleic acids, charged semi-synthetic or synthetic oligomers, peptide hormones, growth factors, physiological proteins, glycosaminoglycan (GAGs), structural proteins and combinations thereof.
  • peptide hormone refers to proteins with endocrine functions, for example insulin.
  • the macromolecule may have negatively charged side groups.
  • the negatively charged side groups may be selected from a group consisting of phosphate, carboxylate, sulfate and derivatives thereof.
  • the macromolecules may include, but are not limited to, polystyrene sulphate, vinyl sulphate and the like.
  • the glycosaminoglycan-like (GAG) polysaccharides may include, but are not limited to, chondroitin sulfate, dermatan sulfate, heparan sulfate, keratan sulfate, dextran sulfate, hyaluronan and the like.
  • non-glycosaminoglycan (non-GAG) polysaccharides may include, but are not limited to, dextran sulfate, cellulose sulphate, pullolan sulphate and the like.
  • the non-polysaccharides may include, but are not limited to, polystyrene sulfate, polyvinyl sulfate, polyvinyl phosphate, negatively charged polyaminoacids (e.g. polyglutamic acid), derivatives and the like.
  • the negatively charged polymer may be a natural, semi-synthetic or fully synthetic polymer.
  • the negatively charged polymer may include, but is not limited to, dextran sulfate, cellulose sulphate, pullolan sulphate, chondroitin sulfate, dermatan sulfate, heparin sulphate and the like.
  • the negatively charged polymer may not have a linear backbone of an ⁇ (1->4) linked repeating units (polysaccharide) and/or may have a linear backbone of an a(l->6) linked repeating units (such as polysaccharide).
  • the negatively charged polymer as exemplified in the Example section of the present disclosure is sulfated dextran (DxS) or dextran sulfate polymer, which is a negatively charged polycarbohydrate.
  • the negatively charged polymer may be dextran sulfate, which is a sulfated dextran polymer.
  • the structure as described herein may contain between about 1 g to about 8000 g of the negatively charged polymer per ml of structure. That is, the structure as described herein may have between about 1 to about 500 g, or between about 1 to about 1000 g, or between about 1000 to about 5000 ⁇ g, or between about 5000 to about 10000 ⁇ g, or between about 250 to about 750 ⁇ g, or between about 2000 to about 4000 ⁇ g, or between about 4000 to about 8000 ⁇ g of the negatively charged polymer per ml of structure.
  • biologically active material refers to an agent, substance or cell, which when provided into a subject, is capable of modulating the subject's physiology.
  • the "biologically active material” may be an agent, substance or cell intended for use in the treatment or prevention of disease in a subject, such as a mammal, such as human, dog, cat, rodent, horse, non-human primates, and the like.
  • biologically active material may include, but is not limited to, liver-derived cells, pancreas-derived cells (such as pancreatic islets), umbilical cord-derived cells, umbilical cord blood derived cells, brain-derived cells, spleen-derived cells, bone marrow derived cells, adipose derived cells, cells derived from IPS technology, cells derived from embryonic stem cells, genetically engineered cells, pluripotent cells, multipotent cells, neural cells, astrocytes, hepatocytes, fibroblasts, mesenchymal cells, pericytes, cardiomyocytes, cardiomyocyte progenitor cells, hematopoietic cells, endothelial cells, endothelial progenitors, smooth muscle cells, keratinocytes, stem cells and progenitors cells, cell mixtures, medicaments, pharmaceutical compositions, growth factors, differentiation factors, transcription factors, nucleic acids, amino acids, proteins, protein fragments and combinations thereof.
  • pancreas-derived cells such as
  • the biologically active material may be an adherent or non-adherent cell.
  • the biologically active material may be a stem cell, including but not limited to, mesenchymal stem cells, neural stem cells, hematopoietic stem cells, endothelial progenitor cells and adipose-derived stem cells.
  • the biologically active material is a mesenchymal stem cell.
  • the encapsulated mesenchymal stem cell may be provided into a subject requiring heart tissue regeneration.
  • the biologically active material may be a pancreatic islet.
  • the encapsulated islet cells may be provided into a subject having diabetes such as Type 1 or Type 2 diabetes.
  • the structure of the present disclosure may further comprise supplementation with components of extracellular matrix.
  • the structure as described herein may further comprise a biocompatible, biodegradable polymer and at least one structural component capable of emulating extracellular matrix (ECM).
  • ECM extracellular matrix
  • biocompatible refers to the property of being biologically compatible with living tissue of a host organism by not producing a toxic, injurious, foreign body response or immunological response.
  • biocompatible may also refer to a material which performs its desired function when introduced into an organism without inducing significant inflammatory response, immunogenicity, or cytotoxicity to native cells, tissues or organs.
  • the phrase "foreign body response” refers to an immunological response of a biological tissue to the presence of any foreign material in the tissue, which can include protein adsorption, chronic inflammation, multinucleated foreign body giant cells and fibrosis.
  • biodegradable polymer refers to a polymer or polymers which degrade in vivo, and wherein erosion of the polymer or polymers over time occur concurrent with or subsequent to release of the biologically active material.
  • the slow degradation of the biocompatible, biodegradable polymer results in the slow release and/or time release of the biologically active material that is encapsulated in the microstructure (such as for example conjugated into the biocompatible, biodegradable polymer).
  • the biologically active material may be conjugated to the biocompatible, biodegradable polymer for slow release and/or time release.
  • the biodegradable polymer as described herein may not be degraded by proteolytic degradation that may occur in view of the host immune system's reaction to the microstructure.
  • the biocompatible, biodegradable polymer may not be proteolytically degradable or essentially not proteolytically degradable.
  • the biocompatible, biodegradable polymer may be capable of providing mechanical stability (for example through gelation of the biocompatible, biodegradable polymer).
  • the biocompatible, biodegradable polymer may be soluble in aqueous solution.
  • the biocompatible, biodegradable polymer may be natural, semi- synthetic or fully synthetic.
  • the biocompatible, biodegradable polymer may include, but is not limited to, a polysaccharide, polyester, protein, polyhydroxybutyrate, poly glycolic acids, polyglycolide, polylactide, chitosan, hyaluronic acid, poly-(D,L)-lactic acid, ethylhydroxyethyl cellulose, polycaprolactone, polycaprolactone diol, polylysine, polyglycolic acid, polybenzyl-L- glutamic acid, polymaleic acid, hydrogels, polyethylene glycol (PEG), their derivatives and combinations thereof.
  • a polysaccharide polyester, protein, polyhydroxybutyrate, poly glycolic acids, polyglycolide, polylactide, chitosan, hyaluronic acid, poly-(D,L)-lactic acid, ethylhydroxyethyl cellulose, polycaprolactone, polycaprolactone diol, polylysine, polyglycolic acid,
  • the biocompatible, biodegradable polymer may have a linear backbone of an a(l->4) linked linear repeating units (polysaccharide).
  • the biocompatible, biodegradable polymer may be a polysaccharide such as agarose or alginate.
  • alginate refers to a class of linear polysaccharide copolymers formed from 1-4-flycosidically linked ⁇ -D-mannuronate (M) and its C-5 epimer a- L-guluronate (G).
  • Alginates are naturally occurring biopolymers produced by a variety of organisms, including marine brown algae and at least two genera of bacteria (Pseudomonas and Azotobacte). Typically, commercial alginates are isolated from marine algae, including Macrocystis pyrifera, Ascoph Hum nodosum, and various types of Laminaria.
  • Agarose refers to a biomaterial that is not degradable enzymatically within tissue, but has been shown to be phagocytized by macrophages. In contrast to other enzymatically degradable materials, it remains longer within the host tissue, thus allowing for a slow and gradual release of encapsulated biologically active material into the surrounding tissues of transplant (or graft) recipient.
  • agarose is a polysaccharide derived from seaweed, but is neutral in charge. It has previously been reported that neutrally charged polymers induce lower foreign body response (FBR) than charged ones.
  • agarose has been reported to induce a foreign body response at higher concentrations; therefore it was found that a specific range of percentages (w/v of total microstructure) of biocompatible, biodegradable polymer, such as agarose, is beneficial in avoiding a foreign body response (FBR).
  • FBR foreign body response
  • agarose was found to be insufficiently immunoprotective and allows extrusion of cells, it was previously dismissed for uses in allogeneic or xenogeneic cell transplantation.
  • the inventors of the present disclosure surprisingly found these properties enables agarose to be a suitable candidate for delivery of a biologically active material into a subject.
  • the inventors of the present disclosure found a specific range of percentages of biocompatible, biodegradable polymer (w/v of total structure) is ideal in providing such properties.
  • the structure as described herein may contain between about 0.1 to about 0.5% w/v (of the total w/v of the structure) of the biocompatible, biodegradable polymer.
  • the structure as described herein may contain between about 0.1 to about 0.5%, or between about 0.5 to about 1 %, or between about 0.25 to about 0.6%, or between about 0.3 to about 0.5%, or between about 1 to about 2%, or between about 1.5 to about 2.5%, or between about 2.5 to about 5%, or between about 5 to about 10%, or between about 10 to about 15%, or between about 15 to about 20%, or between about 20 to about 25%, or between about 7 to about 11%, or between about 13 to about 17%, or between about 17 to about 23% w/v of the biocompatible, biodegradable polymer.
  • the structure may contain between about 0.3% to about 0.4% w/v of the biocompatible, biodegradable polymer.
  • the structure as described herein further comprises at least one structural component.
  • the phrase “structural component” refers to scaffolding polymers or "polymer scaffold”, which refers to material that mimics, resembles or simulates the structure and function of the extracellular matrix.
  • extracellular matrix refers to the extracellular part of cellular structure (for example, organisms, tissues, biofilms) that typically provides structural and biological support to the surrounding cells.
  • the structural component may be natural, semi-synthetic or fully synthetic.
  • the structure as described herein may have at least one structural component.
  • the structure as described herein may have at least two, at least three, at least four, at least five, at least six, at least seven, at least eight or more structural components.
  • the structural components may include, but are not limited to, proteins, non-proteins, synthetic materials emulating structural components and mixtures thereof.
  • the structural component may be a non-protein, which may be synthetic materials containing amino acid sequences allowing cell interaction.
  • the structural component may be proteins including, but are not limited to, serum-derived proteins, extracellular matrix proteins, synthetically modified structural proteins, aggrecan, biglycan, collagens, gelatine, decorin, elastin, fibrinogen, fibrin, fibulins, fibrillins, tenascins, fibronectin, heparan sulphate proteoglycans, keratins, laminins, vimentin, vitronectin and combinations thereof.
  • the collagens that may include, but are not limited to, collagen I, collagen II, collagen III, collagen IV, collagen V, and the like.
  • the structural component may be a protein, which may be collagen and/or fibrinogen or derivatives thereof.
  • the structure may comprise at least two structural components namely collagen I and fibrinogen or derivatives thereof.
  • the structure may have at least two structural components such as collagen and fibrinogen.
  • the structure may contain between about 0.01 to about 40% w/v, or about 40% (w/v) to about 99% (w/v) of at least one structural component capable of emulating extracellular matrix. That is, the structure may contain between about 0.01 to about 1%, or between about 1 to about 5%, or between about 5 to about 15%, or between about 15 to about 30%, or between about 30 to about 50%, or between about 0.05 to about 2%, or between about 2 to about 10 %, or between about 10 to about 20%, or between about 20 to about 40% w/v, or between about 40 to about 50% w/v, or between about 50 to about 60% w/v, or between about 60 to about 70% w/v, or between about 70 to about 80% w/v, or between about 80 to about 90% w/v, or between about 90 to about 99% w/v of at least one structural component capable of emulating extracellular matrix (ECM).
  • ECM extracellular matrix
  • the components of the structure as illustrated in the Example section may be substituted with suitable materials as exemplified above.
  • the components of the structure as described herein may be substituted with a naturally occurring polymer known in the art, synthetic materials known in the art or a semi- synthetic material known in the art.
  • the biocompatible, biodegradable polymer, the at least one structural component and the negatively charged polymer of the structure as described herein may be natural, semi-synthetic or fully synthetic.
  • the structure may be a microcapsule comprising agarose and at least two structural components capable of emulating ECM and a negatively charged polymer.
  • the structure may be a microcapsule/macrocapsule comprising agarose, collagen, at least one further structural component capable of emulating ECM and a negatively charged polymer.
  • the structure may be a microcapsule and/or macrocapsule comprising agarose, collagen, fibrinogen and a negatively charged polymer.
  • the structure may be a microcapsule/macrocapsule comprising or consisting of agarose, collagen, fibrinogen and dextran sulfate.
  • the microcapsules/macrocapsules as described herein may comprise of a composition of the following materials, with the biocompatible, biodegradable polymer being agarose or a material with similar physical characteristics; structural components, including proteins such as serum-derived proteins, which may be selected from extracellular matrix proteins, or derivatives thereof, such as gelatin, or synthetic materials emulating structural proteins, as well as all synthetically modified structural proteins (such as alkylated collagen and the like), which would include collagen and fibrinogen; biocompatible materials or polymers which may be highly charged for the purpose to complex (i.e. aggregate/coagulate, and the like) with structural proteins or other microcapsule components.
  • the biocompatible, biodegradable polymer being agarose or a material with similar physical characteristics
  • structural components including proteins such as serum-derived proteins, which may be selected from extracellular matrix proteins, or derivatives thereof, such as gelatin, or synthetic materials emulating structural proteins, as well as all synthetically modified structural proteins (such as alkylated collagen and the like), which would include
  • one purpose of the components of the microstructure/macrostructure as described herein is to emulate glycosaminoglycan and extracellular matrix structures, which the inventors of the present disclosure found to be surprisingly beneficial in providing optimal cell microenvironment that leads to growth and cell viability even six weeks post transplantation (see Fig. 7).
  • the components of the microstructure/macrostructure as described herein has a role in retaining small molecular weight components like growth factors (VEGF) and present them to the encapsulated cells or to achieve a steady release of these small molecules components.
  • VEGF growth factors
  • heparin or heparin sulphate mediates interaction of VEGF with its receptors.
  • the structure as described herein may comprise a biocompatible, biodegradable polymer that is alginate or agarose or combinations thereof; a structural protein that is collagen I or hydrolysed collagen or combinations thereof; another structural protein that is fibrinogen; and a negatively charged polymer that is dextran sulfate.
  • Fig. 7 of the present disclosure illustrates the growth and viability of mesenchymal stem cells in rat hearts even after six weeks post-transplantation.
  • the organs and tissue may include, but are not limited to, liver, bone, heart, pancreas, liver, lung, skeletal muscle, skin, cartilage, tendon, ligaments, adipose tissue, tissue of the lymphatic circulatory system, tissue of the vascular system, neural cells, periphery neurons, tissue of the central nervous system, and the like.
  • the biologically active material may comprise stem cells.
  • the biologically active material may comprise mesenchymal stem cells.
  • the present disclosure also provides the use of a structure as described herein in the manufacture of a medicament for the treatment of an ischemic disease, muscular dystrophy and diseases arising from genetic deficiencies.
  • the ischemic disease may include, but is not limited to, biliary ischemia, bone-related ischemia, cerebral ischemia, colonic ischemia, coronary ischemia, foot-related ischemia, hepatic ischemia, mesenteric ischemia, myocardial ischemia, optical nerve ischemia, retinal ischemia, spinal ischemia and the like.
  • the ischemic disease may be myocardial infarction.
  • the form of the biologically active material and the delivery route of the structure would define the physical requirements of the structure, and hence influence the form of the structure. Additionally, it is also understood by the skilled person in the art that the structure may be prepared in in liquid form prior to transplantation for ease of injection. Upon injection into the body, the liquid structure may subsequently solidify within the host subject.
  • CaC3 ⁇ 4 dihydrate may be used to prevent the clotting of fibrinogen during the formation of structures such as microcapsules and/or macrocapsules.
  • suitable cations would be non-toxic.
  • strontium is one example of non-toxic suitable cations.
  • the anion or hydrate water may or may not be crucial to facilitate the process (various anions or hydrates are possible).
  • a method of manufacturing the structures as described herein comprising emulsifying methods, polymerization methods, forming methods, moulding methods, casting methods and coating methods.
  • the term "moulding" implies the use of an actual mould, whereas forming implies forming a structure without the use of a mould.
  • the polymerization and forming methods may refer to methods that polymerize units of pre-formed microstructures. As would be understood by the skilled person in the art, a polymerization itself does not indicate the formation of structures but rather the build-up of a macromolecule in some way.
  • curing for example the process to solidify silicon polymers into patches, implants, microfluidic devices are known as curing silicon (e.g.PDMS), that may also involve a curing agent that induces the process in some cases.
  • methods used to form units of desired structures by division from raw material, pre- or post-solidification may also be utilised.
  • a method of manufacturing the structures as described herein may comprise the steps of (a) mixing a biologically active material with a biocompatible, biodegradable polymer, at least one structural component capable of emulating ECM and a negatively charged polymer, resulting in a biomaterial mix; (b) adding said biomaterial mix to an emulsifier to form an emulsion; (c) isolating the formed structures from the emulsion; and optionally (d) incubating the formed structures with a polymerization agent.
  • the method may comprise the steps of (a) mixing an aqueous phase, wherein the aqueous phase comprises a solution of biologically active material and/or cellular cargo with an oil or non-aqueous phase containing a emulsifier; (b) forming microcapsules and/or macrocapsules, microstructures and/or macrostructures or beads by shaking, membrane emulsification, droplet generator, microfluidics or any other method that is essentially emulsion based or forming the aqueous phase containing biomaterial components and cellular cargo into droplets that subsequently solidify in liquid phase, air phase or at the solid/air or solid liquid interface; (c) generating desired shapes and sizes using an extrusion or mould-based method; (d) allowing the aqueous phase containing biologically active material to solidify prior to a process
  • the emulsifier may be SPAN ® 80 (IUPAC name is: [2-(3,4- dihydroxyoxolan-2-yl)-2-hydroxyethyl]octadec-9-enoate).
  • the polymerization agent may be thrombin.
  • the term "about”, in the context of a diameter of a microstructure, means +/- 5% of the stated value, +/- 4% of the stated value, +/- 3% of the stated value, +/- 2% of the stated value, +/- 1% of the stated value, or +/- 0.5% of the stated value.
  • the invention illustratively described herein may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein. Thus, for example, the terms “comprising”, “including”, “containing”, etc. shall be read expansively and without limitation.
  • Human bone marrow MSCs (ATCC), rat bone marrow MSCs (isolated from rat femur) and IMR-90 fibroblasts (ATCC) were cultured in DMEM supplemented with 10% FBS and 1% penicillin-streptomycin at 37°C and 5% C0 2 in a humidified incubator.
  • Low temperature gelling agarose (Sigma) was dissolved and autoclaved in PBS at a stock concentration of 2% (w/v).
  • Dextran sulfate (DxS) with a molecular weight of 500kDa was prepared as a stock solution of lOmg/ml in deionized water.
  • a concentrated CaCi 2 dihydrate solution (2M) was prepared in deionized water.
  • Collagen I (bovine skin, Advanced Biomatrix) was neutralized using 80% collagen stock solution 10% lOxPBS and 10% 0.1M NaOH. Final neutralized collagen stock had a concentration of 2.48 mg/ml.
  • Fibrinogen stock was prepared fresh every time at a concentration of 40mg/ml in PBS.
  • Thrombin aliquots were stored at a concentration of 2 mg/ml at -20°C.
  • MSCs or fibroblasts were encapsulated at a cell concentration of 0.5-1 x 10 6 cells in 100 ⁇ biomaterial.
  • cells were harvested and the exact number of cells per sample was collected in a pellet. All components of the biomaterial mixture were brought to and kept at 37°C just before mixing.
  • PBS or collagen I and/or fibrinogen were warmed to 37°C first, and then pre-warmed agarose was added at a final concentration of 0.4% (w/V). Next DxS and CaC3 ⁇ 4 were added and the biomaterial composition was gently mixed (Exact biomaterial composition details can be found in table 1).
  • CaC3 ⁇ 4 increased cell viability during encapsulation and allowed isolation of fibrinogen-containing microcapsules, which otherwise strongly aggregated during the first purification step of the microcapsules.
  • biomaterial mixtures were prepared, they were mixed immediately with the cell pellet.
  • the cell- biomaterial suspension was added into 1ml of pre-warmed soya bean oil with 0.5% (w/v) Span ® 8 (Sigma Aldrich, USA).
  • An emulsification was formed by manually shaking. Whole vial containing the emulsification was placed on ice for 10 min to allow solidification of agarose. The emulsion was layered on top of 1 ml of HBSS and centrifuged at 200g for 10 minutes.
  • Solidified agarose acted as a scaffold for pre-mature microcapsules.
  • the oil phase was removed and the microcapsules pellet gently collected and placed into fresh HBSS.
  • HBSS was no fibrinogen present in the microcapsule material
  • encapsulated cells are washed twice with HBSS.
  • Fibrinogen was one of the components of the microcapsule material, 100 ⁇ of cell-biomaterial suspension were added to 1 ml of HBSS and incubated with 10 ⁇ thrombin at room temperature. Thrombin incubation took place on a horizontal shaker (400 rpm) for 30 minutes.
  • encapsulated cells were collected and washed twice with full DMEM media. All encapsulated cells were plated into adherent surface plates over night to allow attachment of cells, which were not permanently trapped within the microcapsules. On the next day all microcapsules were transferred into non-adherent plates.
  • Table 1 Composition of one example of a microcapsule material
  • Fibrino gen [ ⁇ )mg/m 40 ⁇ ⁇ 5/ 40 ⁇ 2 ⁇ ⁇ 0.1-10 ⁇ ( 2 ⁇
  • Fibrinc gen (2( )mg/m _ 40 ⁇ Col / 40 ⁇ 2 ⁇ ⁇ 0.1-10 ⁇ 2 ⁇
  • Microcapsules were collected and washed with PBS. They were incubated in 200 ⁇ staining solution consisting of PBS with Calcein AM (1: 1600 dilution) and ETDH-1 (1:400 dilution) for 45 minutes. Microcapsules were pelleted and 15 ⁇ of pellet was pipetted onto a glass slide, covered with a cover slip and sealed with nail polish. Samples were imaged immediately afterwards using the Nikon upright Ni-E microscope. Staining areas were quantified using ImageJ software.
  • Microcapsules were collected, washed twice with PBS and fixed with 4% paraformaldehyde for 10 minutes with 3 subsequent PBS washes.
  • DxS tracking FITC-conjugated DxS was used and no further staining was necessary for imaging. The images were taken with the Nikon N-STORM/TIRF microscope at 40x magnification and an exposure time of 10 seconds. Samples without added DxS did not give any signal.
  • phalloidin staining alone microcapsules were dried on a glass slide. Immunostaining took place in suspension. For phalloidin and immunostaining, fixed microcapsules were blocked for 1 hour with 3%BSA and incubated for 1.5 hours with the primary antibody.
  • Encapsulated mesenchymal stem cells were cultured either alone or in the presence of 10 ⁇ CPX (ciclopirox olamine) for 24 hours. Supernatants of encapsulated cells were analyzed for secreted VEGF using the DuoSet ELISA kits (R&D System) following the manufacturer's instructions. Results were normalized to cell numbers of originally encapsulated MSCs.
  • Encapsulated cells were seeded into adherent surface 24 well-plate wells and allowed to migrate out of capsules for 1 week. Osteogenic differentiation took place directly after that. For adipogenic differentiation, cells were cultured until confluence before differentiation. For osteoblast differentiation, media composed of HG DMEM containing 10% FBS and 1% P/S with dexamethasone (1 x 10-7 M), ascorbic acid at 100 ⁇ and ⁇ -glycerophosphate at 10 mM, was prepared freshly every time. Media was changed every 3 to 4 days for 4 weeks. Cells were washed 2 times with PBS, fixed using 4% formaldehyde and washed 2 more times with PBS.
  • hydroxyapatite was stained using 40 mM alizarin red. Wells were washed 3 times with water and air- dried.
  • induction media was composed of HG DMEM containing 10% FBS and 1% P/S with 0.5 mM 3-isobutyl-l-methylxanthine (IBMX), ⁇ dexamethasone, 0.2 mM indomethacin and 10 ⁇ g/ml insulin and the maintenance media of HG DMEM 10% FBS and 1% P/S and 10 ⁇ g/ml insulin. Cells were induced for 4 days and then kept in maintenance media for 3 days.
  • IBMX 3-isobutyl-l-methylxanthine
  • Cell counting Kit-8 (CCk-8) assay (Sigma Aldrich, USA) was performed according to manufacturer's instructions. Encapsulated cells were cultured in non-adherent plates and were transferred into adherent surface plates one day before CCk-8 assay was performed to isolate single cells. On the day of the assay encapsulated cells were re-suspended in fresh media and 150 ⁇ of cell suspension were mixed with 15 ⁇ of CCk-8 solution in a 96 wells-plate. Incubation took place for 4 hours at 37°C and 5% CO 2 in a humidified incubator. Cell suspensions were collected and centrifuged and 100 ⁇ of the supernatant were placed into a new 96 well plate. Absorbance readings were taken at 450 nm and 650 nm for reference. Results are plotted as a fold-change from absorbance reading on day 1.
  • lxlO 6 labelled cells (suspended in 100 ⁇ of PBS) were implanted into both the lesioned and peri-infarcted areas (three sites of injection; 50 ⁇ into infarcted area and 25 ⁇ into two peri-infracted areas) with a 27-gauge needle. Animals received analgesics and antibiotics for three days post-surgery.
  • Post-implantation stem cell localization and heart function were assessed in anaesthetized rats using high-resolution MRI acquired on a 7T horizontal-bore animal scanner (Cliniscan, Bruker) 2 days post-implantation and then on week 1, 2, 4 and 6 using a cardiac array coil.
  • Microcapsules were generated by suspending biomaterial-cell mixtures in a water-in-oil emulsion, using soybean oil containing 0.5% SPAN ® 80. Agarose concentration as low as 0.4% was sufficient to generate stable microcapsules. Therefore all biomaterial compositions containing dextran sulfate (DxS), collagen I and/or fibrinogen were mixed with 0.4% agarose. Microcapsules containing fibrinogen were incubated additionally with thrombin to yield fibrin. Live-dead cell staining at early time-points were used to evaluate the ability of the material to support mesenchymal stem cells survival (Fig. 1).
  • the present disclosure compared microcapsules containing collagen I and DxS (concentrations 0-100 ⁇ g/ml) or collagen I mixed with fibrin and 100 ⁇ g/ml DxS.
  • Cell survival in capsules increased in a dose-dependent manner with the DxS concentration (Fig. Id).
  • Highest cell survival rates were found in microcapsules composed of a mixture of collagen I, fibrin and 100 ⁇ g/ml DxS (Fig. Id).
  • Resulting collagen hydrogels were imaged in 3D via confocal reflection microscopy according to methods known in the art, for example Jawerth LM, Munster S, Vader DA, Fabry B, Weitz DA.
  • a blind spot in confocal reflection microscopy the dependence of fiber brightness on fiber orientation in imaging biopolymer networks. Biophys J 2010; 98(3): Ll-3.
  • the presence of DxS markedly influenced the architecture of gels, reducing the amount of visible fibers for DxS concentrations between 0 and 10 ⁇ g/ml (Fig. 2c). No structure was visible for DxS concentrations of 20 ⁇ g/ml and above.
  • Agarose affected collagen fibrillogenesis only in the absence of DxS as assessed by turbidimetry. Agarose (0.4%) induced an early nucleation in the absence of DxS, but had no significant effect in the presence of 100 ⁇ g/ml DxS (Fig 2d).
  • DxS aggregates with collagen I and enhances cell-microenvironment interactions.
  • FITC-conjugated DxS at the optimal concentration of 100 ⁇ g/ml was tracked in microcapsules, with and without supplementation of collagen I (Fig. 3a).
  • DxS could only be tracked in agarose-collagen I capsules, whereas in agarose capsules alone fluorescent signal was below the detection limit (Fig. 3a).
  • Fluorescent signal from DxS co-localized with collagen fibrils within the microcapsules, indicating DxS-collagen I interactions (Fig 3a, magnified). Overall this indicates that DxS aggregated with collagen monomers and inhibited fiber elongation.
  • Aggregation of collagen I monomers by DxS will lead to a different presentation of binding sites to the cells.
  • Phalloidin staining of the actin cytoskeleton confirmed encapsulated cells form extrusion in the presence of collagen I, which are further enhanced by DxS supplementation (Fig. 3b). Therefore co-localization of collagen and DxS, and aggregation of collagen I monomers by DxS enhances cell-ECM interactions.
  • Microcapsules composed of collagen I, fibrin and 100 ⁇ ⁇ DxS demonstrate optimal characteristics and functionality
  • Live-dead cell staining allows the monitoring of cell survival over only a short period of time. In order to monitor cell numbers over a longer period, proliferation assays are necessary. Human MSC survival and proliferation over three weeks were tested with a metabolic assay (CCK-8) under selected conditions. The measured enzymatic activity of dehydrogenases is considered to be directly proportional to cell numbers in this assay. Starting at day 2 post encapsulation, cell numbers in microcapsules containing only agarose remained constant over 3 weeks (Fig. 4a), indicating that majority of cell death was occurring shortly post encapsulation (Fig. 1 a,c). MSCs in collagen I - agarose microcapsules with or without DxS displayed no significant increase in cell number (Fig.
  • Microcapsules had an average diameter of 106 ⁇ + 26 ⁇ (average + SD) (Fig 4 b, and c).
  • Fig 4 b, and c average + SD
  • cells in microcapsules were cultured on adherent tissue culture plates, cells were able to migrate out of capsules, indicating the presence of large pore sizes allowing the exit of mesenchymal stem cells (Fig. 4b).
  • attached microcapsules could be washed off easily from the plates, leaving cells and cell debris behind, indicating that microcapsules did not absorb on to the surface, but were only partially attached via outwardly migrated cells.
  • Encapsulated human MSCs were shown to secrete VEGF, an important growth factor for angiogenesis and therefore tissue repair (Fig. 5a). Hypoxic conditions were emulated using ciclopirox olamine (CPX), a prolyl hydroxylase inhibitor. Emulating hypoxic conditions, CPX administration stabilizes the transcription factor HIF- ⁇ , which then activates various down-stream angiogenic processes. Therefore, incubation with CPX resulted in a significant (24%) increase in VEGF secretion, indicating that encapsulated MSCs secrete paracrine factors and respond to external/environmental stimuli (Fig. 5a). Furthermore, migrated MSCs were subjected to standard differentiation protocols into adipocytes and osteoblasts (Fig. 5b). Successful differentiation was confirmed by both Nile red staining of accumulated fat droplets in adipocytes, and alizarin red staining of hydroxyapatite deposited in osteoblast cultures (Fig. 5b).
  • encapsulated cells were stained with phalloidin (green fluorescent - seen as bright spots) and dapi (blue nucleus- seen as dull spots).
  • ECM proteins collagen I, collagen IV, fibronectin and heparin sulfate proteoglycans (HSPG)
  • HSPG heparin sulfate proteoglycans
  • Microcapsules composed of collagen I, fibrin and 100 ⁇ DxS do not induce a fibrotic body response and are biodegradable in vivo
  • microcapsules were injected into rat calf muscles, and animals euthanized four weeks later. Upon initiation of a foreign body response, a fibrotic capsules should form around the implant after this period of time. Microcapsules were generated using TRITC- conjugated agarose to allow easier tracking of capsules. Strong signal from TRITC could be detected in small areas in the muscle (Fig. 6b). However, no spherical shape was observed, indicating that microcapsules disintegrated over time. Hematoxylin and Eosin (H&E) staining confirmed an infiltration of cells (Fig. 6b).
  • H&E Hematoxylin and Eosin
  • USPIO-labelled rat bmMSCs encapsulated in collagen I, fibrin and 100 g/ml DxS (Col-Fb-DxSlOO) microcapsules can be tracked long-term by MRI, when injected into the myocardium.
  • Col-Fb-DxS 100 microcapsules showed a reasonable response in vivo, they were tested for the suitability to deliver MSCs into the myocardium.
  • MSCs were isolated from wistar rat femur bone marrow, expanded and labelled with TAT peptide derivatized ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles coated with dextran (IODEX-TAT-FITC). 10 weeks old wistar rats underwent a thoracotomy at the fourth intercostal space with subsequent left anterior descending coronary artery (LAD) ligation.
  • UAT peptide derivatized ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles coated with dextran (IODEX-TAT-FITC) 10 weeks old wistar rats underwent a thoracotomy at the fourth intercostal space with subsequent left anterior descending coronary artery (LAD) ligation.
  • LAD left anterior descending coronary artery
  • Sections were co-stained against CDl lb to ensure that nanoparticles were still within the transplanted rat MSCs and not taken up by macrophages (Fig. 7c). Staining for nanoparticles co-localized or overlaid with oval-shaped intact nuclei in both conditions, indicating integrated stromal cells (Fig. 7c). An overlapping with CDl lb staining was very rare (MSCs suspension, white arrow), but confirmed that dead transplanted MSCs were scavenged by macrophages. Interestingly, in cardiac sections with injected encapsulated MSCs some of the larger patches of stained nanoparticles were surrounded or in proximity to macrophages (CDl lb staining).
  • CDl lb staining was often not in direct contact with staining for nanoparticles and showed a pattern comparable to the one observed around the biomaterial composition in the host response study (Fig. 6c). Therefore CDl lb staining was an indirect indication of the retention of microcapsule.
  • a fraction of transplanted rat MSCs had migrated out of the microcapsules and integrated into the host tissue, whereas some of the transplanted MSCs were still found within the biomaterial (Fig. 6c). Therefore Col-Fb-DxSlOO capsules were proven suitable to deliver therapeutic cells into the myocardium.
  • the optimized microcapsule biomaterial for cell encapsulation is a finely tuned composite of agarose, collagen I, fibrin and DxS. All components play a role for the performance of the microcapsule. Although materials such as agarose (or substituted by alginate) in combination with collagen, fibrin and fibronectin have been investigated before, the resulting microcapsule biomaterials had limited performance. Most encapsulated cells (MSCs or fibroblast) had a decreased viability over time; cell-material interaction, cell functionality post encapsulation, and in vivo applications were often not examined. Furthermore, this proves that the supplementation of microcapsules with extracellular matrix (ECM) proteins alone is not sufficient for the optimal performance of cells and biomaterial.
  • ECM extracellular matrix
  • GAGs glycosaminoglycans
  • DxS dextran sulfate
  • DxS an off-the-shelf sulfated glucose polymer
  • Dextran and DxS are degraded into glucose and non-toxic. Both are approved for medical applications.
  • DxS plays a role for production of microcapsules containing fibrin, as the absence of DxS leads to aggregation of fibrin-containing microcapsules during purification from the oil phase.
  • DxS has additional functionality, in that it interacts with the supplemented proteins, enhancing cell- microenvironment interaction with downstream effects on cell survival. Nevertheless, it is the combination of all the components, which allows for the optimal functionality of the biomaterial.
  • Collagen I and fibrin provide mechanical stability and anchorage for the cells, and are both approved for biomedical application.
  • Span ® 80 and agarose are food additives and generally recognized as safe (GRAS) by FDA.
  • MSCs show not only increased survival, but also proliferation rates within the microcapsules. MSCs perform all functions that are reported to contribute to tissue repair when transplanted, as they are able to differentiate into other lineages and secrete paracrine factors.
  • the microcapsule composition On translation into an in vivo setting, the microcapsule composition exhibited sufficient mechanical stability for intramuscular injection and a reasonable body response with phagocytizing macrophages, but without a fibrotic capsule formation. It is notable that the body response was studied in the absence of MSCs, which possess inherent immunosuppressive properties. It was demonstrated previously that the strong foreign body response towards alginate-PLL microcapsules could be alleviated by encapsulated MSCs.
  • Microcapsules degrade slowly in vivo, therefore allowing a slow and gradual release of cells into the host tissue. Moreover, when injected into the myocardium of immune-competent rats, MR signal from encapsulated MSCs could be tracked for the duration of the study. In contrast, injections of single cell suspension resulted in no or a weak signal at early time-points post surgery. Therefore Col-Fb-DxSlOO microcapsules proved to be suitable for stem cell delivery into the myocardium. Furthermore, stem cell delivery in Col-Fb-DxS 100 microcapsules seems to overcome current shortcoming of limited cell retention in cell-based therapies of heart diseases and therefore could improve the therapeutic effect of MSCs.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Dermatology (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Transplantation (AREA)
  • Developmental Biology & Embryology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Urology & Nephrology (AREA)
  • Botany (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Preparation (AREA)
  • Materials For Medical Uses (AREA)

Abstract

L'invention concerne des macro- et micro-structures. Ces structures peuvent convenir à l'encapsulage de matière biologiquement active, la structure comportant un polymère à charge négative. L'invention concerne également des procédés d'utilisation des structures, des utilisations des structures et leurs procédés de production.
PCT/SG2015/050142 2014-06-05 2015-06-05 Matière et structures destinées à l'adminisatration de cellules WO2015187098A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
SG11201602828YA SG11201602828YA (en) 2014-06-05 2015-06-05 Material and structures for cell delivery

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SG10201402976V 2014-06-05
SG10201402976V 2014-06-05

Publications (1)

Publication Number Publication Date
WO2015187098A1 true WO2015187098A1 (fr) 2015-12-10

Family

ID=54767058

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG2015/050142 WO2015187098A1 (fr) 2014-06-05 2015-06-05 Matière et structures destinées à l'adminisatration de cellules

Country Status (2)

Country Link
SG (1) SG11201602828YA (fr)
WO (1) WO2015187098A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102056391B1 (ko) 2017-03-14 2019-12-16 울산과학기술원 하이드로젤 패치
CN111330072A (zh) * 2020-03-03 2020-06-26 南京鼓楼医院 一种仿生多孔MSCs微球的制备方法及其应用
JP2022526302A (ja) * 2019-03-21 2022-05-24 アンテロジェン シーオー.,エルティーディー. 間葉系幹細胞-ヒドロゲルを含有する注射用組成物、並びにその製造、凍結及び解凍方法
CN114870091A (zh) * 2022-05-17 2022-08-09 哈尔滨医科大学 一种生物友好型心肌敷料及其制备方法与应用
CN117771220A (zh) * 2024-02-28 2024-03-29 山东第二医科大学 一种右美托咪定中药多糖口溶膜剂及其制备方法和应用
US11944723B2 (en) 2018-03-13 2024-04-02 Institut Quimic De Sarria Cets Fundacio Privada Vascular repair patch

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006042132A2 (fr) * 2004-10-08 2006-04-20 Georgia Tech Research Corporation Microencapsulation de cellules dans des hydrogels a l'aide de potentiels electrostatiques
WO2010004018A2 (fr) * 2008-07-11 2010-01-14 Eth Zurich Microcapsules dégradables
US8518682B2 (en) * 2005-03-07 2013-08-27 Boston Scientific Scimed, Inc. Microencapsulated compositions for endoluminal tissue engineering
US20130295012A1 (en) * 2010-08-30 2013-11-07 President And Fellows Of Harvard College Shear controlled release for stenotic lesions and thrombolytic therapies

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006042132A2 (fr) * 2004-10-08 2006-04-20 Georgia Tech Research Corporation Microencapsulation de cellules dans des hydrogels a l'aide de potentiels electrostatiques
US8518682B2 (en) * 2005-03-07 2013-08-27 Boston Scientific Scimed, Inc. Microencapsulated compositions for endoluminal tissue engineering
WO2010004018A2 (fr) * 2008-07-11 2010-01-14 Eth Zurich Microcapsules dégradables
US20130295012A1 (en) * 2010-08-30 2013-11-07 President And Fellows Of Harvard College Shear controlled release for stenotic lesions and thrombolytic therapies

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102056391B1 (ko) 2017-03-14 2019-12-16 울산과학기술원 하이드로젤 패치
US11944723B2 (en) 2018-03-13 2024-04-02 Institut Quimic De Sarria Cets Fundacio Privada Vascular repair patch
JP2022526302A (ja) * 2019-03-21 2022-05-24 アンテロジェン シーオー.,エルティーディー. 間葉系幹細胞-ヒドロゲルを含有する注射用組成物、並びにその製造、凍結及び解凍方法
CN111330072A (zh) * 2020-03-03 2020-06-26 南京鼓楼医院 一种仿生多孔MSCs微球的制备方法及其应用
CN114870091A (zh) * 2022-05-17 2022-08-09 哈尔滨医科大学 一种生物友好型心肌敷料及其制备方法与应用
CN114870091B (zh) * 2022-05-17 2023-07-21 哈尔滨医科大学 一种生物友好型心肌敷料及其制备方法与应用
CN117771220A (zh) * 2024-02-28 2024-03-29 山东第二医科大学 一种右美托咪定中药多糖口溶膜剂及其制备方法和应用
CN117771220B (zh) * 2024-02-28 2024-05-28 山东第二医科大学 一种右美托咪定中药多糖口溶膜剂及其制备方法和应用

Also Published As

Publication number Publication date
SG11201602828YA (en) 2016-05-30

Similar Documents

Publication Publication Date Title
Blocki et al. Microcapsules engineered to support mesenchymal stem cell (MSC) survival and proliferation enable long-term retention of MSCs in infarcted myocardium
Majid et al. Natural biomaterials for cardiac tissue engineering: a highly biocompatible solution
Wei et al. A self-healing hydrogel as an injectable instructive carrier for cellular morphogenesis
Luo et al. Time-responsive osteogenic niche of stem cells: a sequentially triggered, dual-peptide loaded, alginate hybrid system for promoting cell activity and osteo-differentiation
Xu et al. Injectable biodegradable hybrid hydrogels based on thiolated collagen and oligo (acryloyl carbonate)–poly (ethylene glycol)–oligo (acryloyl carbonate) copolymer for functional cardiac regeneration
Santo et al. Chitosan–chondroitin sulphate nanoparticles for controlled delivery of platelet lysates in bone regenerative medicine
Li et al. The effect of layer-by-layer assembly coating on the proliferation and differentiation of neural stem cells
WO2015187098A1 (fr) Matière et structures destinées à l'adminisatration de cellules
Fan et al. A new class of biological materials: cell membrane-derived hydrogel scaffolds
Webber et al. Emerging peptide nanomedicine to regenerate tissues and organs
Jiao et al. Bidirectional differentiation of BMSCs induced by a biomimetic procallus based on a gelatin-reduced graphene oxide reinforced hydrogel for rapid bone regeneration
Choi et al. Gelatin-based micro-hydrogel carrying genetically engineered human endothelial cells for neovascularization
Farhat et al. Hydrogels for advanced stem cell therapies: a biomimetic materials approach for enhancing natural tissue function
Teng et al. A chondrogenesis induction system based on a functionalized hyaluronic acid hydrogel sequentially promoting hMSC proliferation, condensation, differentiation, and matrix deposition
US11998658B2 (en) Injectable porous hydrogels
Lu et al. Human urine-derived stem cell exosomes delivered via injectable GelMA templated hydrogel accelerate bone regeneration
Yan et al. Advances in the application of supramolecular hydrogels for stem cell delivery and cartilage tissue engineering
Kim et al. Temperature responsive chemical crosslinkable UV pretreated hydrogel for application to injectable tissue regeneration system via differentiations of encapsulated hMSCs
Sun et al. Biodegradable mesoporous silica nanocarrier bearing angiogenic QK peptide and dexamethasone for accelerating angiogenesis in bone regeneration
Aminabhavi et al. Polysaccharide-based hydrogels as biomaterials
Sánchez et al. Biologically active and biomimetic dual gelatin scaffolds for tissue engineering
Meco et al. Impact of elastin-like protein temperature transition on PEG-ELP hybrid hydrogel properties
Abourehab et al. Chondroitin sulfate-based composites: a tour d’horizon of their biomedical applications
Yang et al. The development of laminin-alginate microspheres encapsulated with Ginsenoside Rg1 and ADSCs for breast reconstruction after lumpectomy
Molina-Peña et al. Nanoparticle-containing electrospun nanofibrous scaffolds for sustained release of SDF-1α

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15803942

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15803942

Country of ref document: EP

Kind code of ref document: A1