WO2017023955A1 - Tissu cornéen mis au point par génie biologique - Google Patents

Tissu cornéen mis au point par génie biologique Download PDF

Info

Publication number
WO2017023955A1
WO2017023955A1 PCT/US2016/045216 US2016045216W WO2017023955A1 WO 2017023955 A1 WO2017023955 A1 WO 2017023955A1 US 2016045216 W US2016045216 W US 2016045216W WO 2017023955 A1 WO2017023955 A1 WO 2017023955A1
Authority
WO
WIPO (PCT)
Prior art keywords
keratocytes
functional
temperature
cells
substrate
Prior art date
Application number
PCT/US2016/045216
Other languages
English (en)
Inventor
Steven Yu
Original Assignee
Steven Yu
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 Steven Yu filed Critical Steven Yu
Publication of WO2017023955A1 publication Critical patent/WO2017023955A1/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
    • A61L27/36Materials 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0621Eye cells, e.g. cornea, iris pigmented 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
    • A61L2430/16Materials or treatment for tissue regeneration for reconstruction of eye parts, e.g. intraocular lens, cornea
    • 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
    • C12N2513/003D culture
    • 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
    • C12N2523/00Culture process characterised by temperature
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue
    • 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
    • C12N2535/00Supports or coatings for cell culture characterised by topography
    • 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
    • C12N2535/00Supports or coatings for cell culture characterised by topography
    • C12N2535/10Patterned coating
    • 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
    • C12N2539/00Supports and/or coatings for cell culture characterised by properties
    • C12N2539/10Coating allowing for selective detachment of cells, e.g. thermoreactive coating

Definitions

  • This invention relates to bioengineered corneal tissue.
  • FIG. 1 shows a cross-section of the cornea of the human eye.
  • the corneal epithelium is a thin layer of non-keratinized stratified squamous epithelium. This thin layer is composed of several layers of cells which are shed constantly on the exposed layer and are regenerated by multiplication in the basal layer.
  • Bowman's layer (also known as the anterior limiting membrane) is the subepithelial basement membrane.
  • the corneal stroma is a thick, transparent, middle layer of the cornea. The stroma constitutes up to 90% of the corneal thickness and is constructed of highly regular collagenous lamellae and extracellular matrix components along with sparsely distributed, interconnected keratocytes.
  • the stroma has about 300 to 500 such collagenous lamellae, which are generally parallel to the corneal surface.
  • Corneal keratocytes are specialized fibroblasts residing in the stroma. They are the principal cell component of corneal stroma and are responsible for the stromal matrix. Through cell-cell dendritic connections, the keratocytes have an extensive 3D network.
  • the cornea stroma is relatively avascular and receives its essential nutrients primarily by diffusion.
  • the corneal keratocytes produce the extracellular matrix containing collagen, proteoglycans (such as keratocan), and glycosaminoglycans (such as keratan sulfate or dermatan sulfate).
  • the keratocytes constitute approximately 5 - 10% of the stromal volume and are distributed in a helical network from the anterior to the posterior surface.
  • Descemet's membrane also posterior limiting membrane
  • the corneal endothelium is a monolayer of simple squamous or low cuboidal cells. These cells are responsible for regulating fluid and solute transport between the aqueous and corneal stromal compartments.
  • the invention is method of constructing cornea tissue.
  • the method comprises: (a) using a cell culture substrate having a surface, wherein the surface has a grooved pattern and comprises a temperature-responsive polymer in which its hydrophilicity or hydrophobicity can be controlled by a change in temperature; (b) culturing functional keratocytes on the substrate at a first temperature; (c) lowering the temperature to a second temperature to make the temperature-responsive polymer more hydrophilic; (d) detaching the functional keratocytes and extracellular matrix from the substrate surface as a sheet; (e) harvesting the sheet of functional keratocytes and extracellular matrix. In some embodiments, the harvested sheet is transparent.
  • the functional keratocytes are non-randomly oriented. In some embodiments, the method comprises culturing the functional keratocytes until they have a non-random orientation. In some embodiments, the mean orientation angle of the functional keratocytes on the sheet is 30° or less. In some embodiments, at least 75% of the functional keratocytes on the sheet have an orientation angle of 30° or less.
  • the collagen fibrils in the extracellular matrix are non-randomly oriented. In some embodiments, the alignment of the collagen fibrils in the extracellular matrix are influenced by the direction of the grooves. In some embodiments, the method comprises culturing the functional keratocytes until they produce extracellular matrix comprising collagen fibrils having a non-random orientation. In some embodiments, at least 50% of the collagen fibrils in the extracellular matrix have an orientation angle of 30° or less. In some embodiments, at least 75% of the collagen fibrils have a diameter in the range of 15 - 90 nm. In some embodiments, the fibril-to-fibril spacing of the collagen fibrils are spaced with an average spacing of less than 400 nm.
  • the functional keratocytes are cultured until they stratify to form multiple layers of functional keratocytes. In some embodiments, the functional keratocytes are cultured for at least two weeks. In some embodiments, the sheet is detached and harvested prior to the functional keratocytes reaching full confluency.
  • the functional keratocytes are corneal keratocytes or corneal stromal cells that are from an autologous or donor source.
  • the method further comprises: seeding the substrate with progenitor cells or stem cells; and inducing the cells to differentiate into functional keratocytes.
  • the substrate surface is a hydrogel comprising the temperature- responsive polymer.
  • the hydrogel is non-soluble in an aqueous solution.
  • the temperature-responsive polymer is covalently cross-linked.
  • the temperature-responsive polymer is covalently grafted onto the substrate.
  • the method further comprises: (i) lowering the temperature to make the temperature-responsive polymer more hydrophilic and cause it to swell; and (ii) before the sheets completely detach, raising the temperature to make the temperature-responsive polymer more hydrophobic and cause it to shrink.
  • the method further comprises repeating steps (i) and (ii) one or more times.
  • the method further comprises repeating steps (a) - (e) to create multiple sheets and stacking the sheets on top of one another to form a multi-layer tissue construct.
  • This repeating of steps (a) - (e) could be performed according to any suitable time schedule (e.g. in parallel, series, staggered timing, etc.) to produce the multiple cell sheets.
  • the method further comprises culturing the multi-layer tissue construct until at least some of the functional keratocytes migrate from one layer of the multilayer tissue construct to another layer. In some embodiments, the method further comprises culturing the multi-layer tissue construct until at least some of the functional keratocytes interconnect with a functional keratocyte on a different layer of the multi-layer tissue construct. In some embodiments, the multi-layer tissue construct is cultured for a further duration of at least five days.
  • the method further comprises inducing the functional keratocytes to undergo apoptosis.
  • the functional keratocytes are exposed to an apoptosis-inducing agent that induces apoptosis of the functional keratocytes.
  • the functional keratocytes in a cultured sheet are induced to undergo apoptosis prior to detaching and harvesting the cell sheet.
  • the functional keratocytes in a multilayer tissue construct are induced to undergo apoptosis.
  • compression is applied onto the cultured sheet or the multi-layer tissue construct in order to induce keratocyte apoptosis.
  • the invention is a cell sheet comprising functional keratocytes and extracellular matrix.
  • the cell sheet has a round, oval, or elliptical shape.
  • the cell sheet is scaffold-free.
  • the cell sheet is sufficiently sturdy to hold a suture.
  • the functional keratocytes occupy an area of less than 50% of the area of the sheet.
  • the functional keratocytes on the cell sheet are aligned in a non- random orientation.
  • the collagen fibrils in the extracellular matrix are aligned in a non-random orientation.
  • the invention is a tissue construct comprising two or more stacked sheets of functional keratocytes and extracellular matrix.
  • the tissue construct has a round, oval, or elliptical shape.
  • the tissue construct is scaffold-free.
  • the tissue construct is sufficiently sturdy to hold a suture.
  • at least some of the functional keratocytes are in physical contact with a functional keratocyte located in a different sheet.
  • each sheet has functional keratocytes that are aligned in a non- random orientation.
  • each sheet has an extracellular matrix comprising collagen fibrils that are aligned in a non-random orientation
  • the alignment orientation of the functional keratocytes are at an angle with respect to the alignment orientation of the functional keratocytes in an adjacent sheet.
  • the alignment orientation of the collagen fibrils in the extracellular matrix are at an angle with respect to the alignment orientation of the collagen fibrils in the extracellular matrix in an adjacent sheet.
  • the functional keratocytes occupy a volume of less than 50% of the volume of the multi-layer tissue construct. In some embodiments, the average density of the functional keratocytes in the multi-layer tissue construct is less than 75,000 cells/mm 3 . In some embodiments, the average density of the functional keratocytes in the multi-layer tissue construct is less than 38,000 cells in a 1 mm 2 column.
  • the invention is a method of performing a corneal replacement surgery in a patient.
  • the method comprises: removing cornea tissue from the patient's eye, and implanting a bioengineered cornea tissue construct described herein into the patient's eye.
  • the bioengineered cornea tissue construct is optically transparent.
  • the step of implanting comprises suturing the bioengineered cornea tissue construct into the patient's eye.
  • the method comprises, after implanting the bioengineered cornea tissue construct, applying a contact lens on the eye over the implanted tissue construct to result in keratocyte apoptosis in the tissue construct.
  • FIG. 1 shows a cross-section of the cornea of the human eye.
  • FIG. 2 shows an enlarged view of a portion of an example substrate surface having a parallel groove/ridge topography.
  • FIG. 3 shows how the pitch, depth, width of grooves, and width of ridges are measured.
  • FIGs. 4A and 4B show an example of how cell attachment and detachment on a temperature-responsive surface can be controlled by changes in temperature.
  • FIGs. 5A and 5B show an example of how a cell sheet can be cultured and harvested from a temperature-responsive surface.
  • FIG. 6 shows an example of how a thermoresponsive polymer film can be applied onto a patterned substrate.
  • FIG. 7 shows an example of forming a pattern directly onto a layer of thermoresponsive polymer.
  • FIG. 8 shows an example of how alignment of the cells can be measured.
  • FIG. 9 shows keratocytes being cultured on a grooved substrate.
  • FIG. 10 shows how stromal sheets can be stacked from multiple separately cultured sheets.
  • FIG. 11 shows an example of a multi-layer corneal tissue construct.
  • FIGs. 12A - 12C show an example of a corneal replacement surgery.
  • a sheet of cornea stromal tissue can be made by in vitro culturing of functional keratocytes on a substrate such that they deposit extracellular matrix components including collagen fibrils.
  • "Functional keratocytes” are cells that have the ability to deposit an organized transparent extracellular matrix similar to that of the native corneal stroma. As such, they would produce corneal stromal extracellular matrix components, such as collagen fibrils (type I, type V, or type l/V hybrid), proteoglycans (e.g. keratocan), or glycosaminoglycans (e.g. keratan sulfate and dermatan sulfate).
  • collagen fibrils type I, type V, or type l/V hybrid
  • proteoglycans e.g. keratocan
  • glycosaminoglycans e.g. keratan sulfate and dermatan sulfate.
  • the functional keratocytes are corneal keratocytes as they natively exist in the human cornea.
  • “functional keratocytes” may or may not be corneal keratocytes per se as they exist in the human body with precisely the same phenotypic markers as natively found in the human body. They may be corneal stromal cells or
  • differentiated cells that are not literally keratocytes, but exhibiting the ability to function as keratocytes in their ability to deposit organized extracellular matrix components, such as collagen, keratan sulfate, or keratocan. I n addition to their ability to produce extracellular matrix in an organized manner, there are other markers that can be used to identify cells as being functiona l keratocytes.
  • functional keratocytes may also be characterized by their cytoplasmic content of crystallins or aldehyde dehydrogenase that contribute to the transparency of the cornea. Other characteristic markers that can be used to identify functional keratocytes are described in
  • the identification of the cells as "functional keratocytes” may occur at any point during the culturing the process.
  • the functional keratocytes are seeded onto the substrate at the beginning of culturing.
  • corneal stromal cells (such as quiescent keratocytes, activated keratocytes, fibroblasts, repair fibroblasts, or myofibroblasts) are seeded onto the substrate at the beginning of culturing. Under appropriate culture conditions, fibroblasts, repair fibroblasts, or myofibroblasts can be changed into keratocytes.
  • more primitive cells i.e. at an earlier step along a developmental progression to functional keratocytes
  • progenitor cells or stem cells including induced stem cells
  • the functional keratocytes may result from the differentiation of progenitor cells or stem cells that are seeded onto the substrate.
  • Progenitor cells are early descendants of stem cells that can differentiate to form one or more kinds of cells, but cannot divide and reproduce indefinitely.
  • a progenitor cell being more specific than a stem cell, is often more limited than a stem cell in the kinds of cells it can become.
  • the progenitor cell is a mesenchymal fibroblast cell or a primary corneal fibroblast (usually a de-differentiated corneal keratocyte extracted from donor tissue).
  • Some keratocyte progenitor cells are known to express the ocular development gene Pox6, which is not expressed by the resident stromal keratocytes. This may be one possible way to identify a progenitor cell for a functional keratocyte.
  • stem cells examples include human corneal stromal stem cells, human adipose-derived stem cells, and human dental pulp stem cells. See [Syed-Picard et al, "Dental pulp stem cells: a new cellular resource for corneal stromal regeneration” (2015 Mar) Stem Cells Transl Med., 4(3):276-85].
  • the stem cells are stem cells that originate from the cranial neural crest (i.e. of cranial neural crest lineage).
  • the stem cells of neural crest lineage can be co-cultured with mouse PA6 fibroblasts to induce differentiation into adult stromal stem cells, which can then be induced to differentiate into corneal keratocytes by culturing in serum-free medium containing ascorbate. See [Syed-Picard et al, supra].
  • the stem cells are mesenchymal stem cells (e.g. neural-crest derived mesenchymal stem cells).
  • the cells seeded onto the substrate may be functional keratocytes (including native keratocytes, quiescent keratocytes, and activated keratocytes), progenitor cells, stem cells, corneal stromal cells (including fibroblasts, repair fibroblasts, and myofibroblasts that can be induced to differentiate into keratocytes), or corneal mesenchymal cells.
  • the source of the cells may be from any suitable source, including autologous source, allogeneic source (e.g. from a donor), or xenogeneic. Because cornea stroma is relatively immunologically privileged, there is some flexibility in the selection of the source.
  • the cells may be cultured in a culture medium that maintains the viability of the cells or supports their growth. In some embodiments, the culture medium contains an agent that enhances production of extracellular matrix. In some embodiments, the culture medium contains an agent that induces differentiation of the cell into a functional keratocyte (i.e.
  • agents that can do one or both include ascorbic acid or one of the more stable derivatives or analogs of ascorbic acid having the same functional activity as a co-factor in collagen synthesis, such as ascorbyl 2-phosphate, ascorbyl 2-sulfate, ascorbyl 2-glucoside, 2-O-a-D-glucopyranosyl-L-ascorbic acid, ethyl ascorbic acid, and 6-0- alkanoyl ascorbic acids such as ascorbyl palmitate, ascorbyl stearate, ascorbyl octanoate, and ascorbyl dipalmitate.
  • ascorbyl 2-phosphate ascorbyl 2-sulfate
  • ascorbyl 2-glucoside 2-O-a-D-glucopyranosyl-L-ascorbic acid
  • ethyl ascorbic acid ethyl ascorbic acid
  • 6-0- alkanoyl ascorbic acids such as ascorbyl palmitate, ascor
  • the composition of the culture medium may remain the same or change throughout the culturing of the cells.
  • the culture medium may comprise growth factors such as fibroblast growth factor-2 (FGF-2 or basic fibroblast growth factor), transforming growth factor such as TGF- 2 and TGF- 3, platelet-derived growth factor (such as PDGF BB), insulin, or insulin-like growth factor 2 (IGF-2).
  • FGF-2 or basic fibroblast growth factor transforming growth factor such as TGF- 2 and TGF- 3
  • platelet-derived growth factor such as PDGF BB
  • insulin insulin-like growth factor 2
  • Culture medium containing insulin or IGF-2 has been shown to promote cell proliferation while maintaining keratocyte cell morphology. See [Musselmann, "Developing culture conditions to study keratocyte phenotypes in vitro" (2006) graduate Thesis at University of South Florida; see http://scholarcommons. usf.edu/etd/641].
  • the culture medium contains heparin (optionally, along with FGF-2). In some embodiments, the culture medium contains bovine serum albumin. In some embodiments, the culture medium contains corneal extract or corneal stromal extract. Any suitable technique for making corneal extract or corneal stromal extract may be used, including the technique described in [Musselmann, supra].
  • the culture medium may contain serum (e.g. fetal bovine serum or platelet poor horse serum).
  • serum e.g. fetal bovine serum or platelet poor horse serum.
  • fetal bovine serum is known to induce the conversion of keratocytes from their quiescent phenotype to a fibroblastic phenotype and fibroblasts may be reversed to a keratocyte-like morphology by removal of the serum in the medium.
  • the culture medium is serum-free.
  • the harvested sheets may have relatively lower cellular content.
  • the functional keratocytes occupy an area of less than 50% of the area of the sheet. Relatively lower cellular content can be achieved by culturing the cells sufficiently long for them to produce extracellular matrix, but before the cells become fully confluent.
  • the sheet of functional keratocytes are detached and harvested before the functional keratocytes reach confluency of 85% (e.g. upon reaching confluence of 68%); in some cases, before reaching confluency of 70%.
  • the functional keratocytes are cultured until they stratify to form multiple (two or more) layers of the cells in the sheet. In some embodiments, the functional keratocytes are cultured for at least two weeks; in some cases, at least three weeks; and in some cases, at least four weeks. In some embodiments, the harvested sheet is scaffold-free (at the time when the sheet is harvested, the sheet contains only cultured cells and the material produced by the cells).
  • the substrate on which the cells are grown has a pattern of grooves and ridges. Surface patterns are known to play a role in cell alignment. On grooved substrates, cells generally align in the direction of the grooves on the substrate, known as "contact guidance.” See [Gil et al, "Response of Human Corneal Fibroblasts on Silk Film Surface Patterns” (2010 June) Macromol Biosci., 10(6):664-673].
  • the substrate may be part of articles such as microplates, culture dishes, microscope slides, chips, etc.
  • the substrate may be constructed from any suitable material, including plastic, metal, glass, or ceramic.
  • Examples of materials for constructing the substrate include poly(dimethylsiloxan), polystyrene, poly(lactic acid), poly(glycolic acid), poly(hydroxybutyrate), polycarbonate, polycaprolactone, polymethylmethacrylate, or other thermoplastic polymers.
  • the substrate topography can be fabricated by any suitable micro- or nano-patterning technique, such as photolithography, electron beam lithography, soft lithography (including capillary force lithography), etching (e.g. laser etching, chemical etching, dry etching, etc.), stamping, imprinting, embossing, vapor deposition, inkjet printing, etc. Examples of such patterning techniques are described in WO 2013/151755 (Kim et al, Univ.
  • FIG. 2 shows an enlarged view of a portion of an example substrate surface having a parallel groove/ridge topography.
  • FIG. 3 shows how the pitch, depth, width of grooves, and width of ridges are measured. In some embodiments, the pitch of the grooves is between 400 - 4,000 nm.
  • the depth of the groove is between 100 - 1,000 nm. In some embodiments, the width of the groove is between 70 - 2,000 nm. In some embodiments, the width of the ridge is between 70 - 2,000 nm.
  • the dimensions of the grooves and ridges may or may not be uniform across the entirety of the substrate. In some embodiments, the grooves are substantially parallel. In some embodiments, the grooves are in a uniformly repeating pattern.
  • one way to create the surface topography is to make a master containing the patterned relief structure prepared via traditional photolithography techniques. Cast an elastomer, such as polydimethylsiloxane (PDMS), against the master to make a mold or stamp. Use the elastomer stamp for hot embossing against a thermoplastic polymer (e.g.
  • PDMS polydimethylsiloxane
  • polystyrene Press the patterned mold against a thin sheet of polystyrene and heat it above the glass transition temperature of polystyrene. This will transfer the pattern on the mold to the polystyrene substrate.
  • the substrate on which the cells are grown has a surface that comprises a temperature- responsive polymer (also commonly known as thermoresponsive polymers), which undergoes a drastic and discontinuous change in hydrophilicity/hydrophobicity with a change in
  • Temperature-responsive polymers undergo this reversible change below and above a lower critical solution temperature (LCST).
  • LCST critical solution temperature
  • the temperature-responsive polymer changes to become more hydrophilic as temperature falls below its LCST and becomes more hydrophobic as temperature rises above the LCST. This allows the hydrophilicity/hydrophobicity of the polymer to be controlled by a change in temperature. Because cells tend to attach more readily to hydrophobic surfaces, this allows for the control of cell attachment and detachment to produce intact cell sheets.
  • FIGs. 4A and 4B show an example of how cell attachment and detachment on a temperature-responsive surface can be controlled by changes in temperature.
  • the substrate 12 is covered with a film of poly(N-isopropylacrylamide) ('poly-NIPAAm'), which is covalently grafted onto the substrate.
  • Poly-NIPAAm has an LCST of 32° C and is relatively hydrophobic at temperatures above its LCST.
  • FIG. 4A shows the culture-substrate interaction at the 37° C temperature under which the cells are cultured. At this temperature, poly-NIPAAm is in a relatively hydrophobic state and the polymer chains 14 are collapsed on the substrate surface. This allows the surface to support cell attachment and growth, similar to conventional culture dishes.
  • the temperature is reduced to room temperature below poly-NIPAAm's 32° C LCST such that the polymer 14 becomes more hydrophilic. Because the poly-NIPAAm is grafted onto the substrate surface, this polymer layer also acts like a gel and becomes swollen as it becomes hydrated. With this transition, the polymer chains 14 expand and swell to create a hydration layer between the cultured cells 10 and the substrate surface, allowing for cell detachment.
  • the cells are grown on the substrate at a temperature in which the temperature- responsive polymer is relatively hydrophobic such that the cells attach to the surface.
  • the temperature is reduced to cause the temperature- responsive polymer to become relatively more hydrophilic. This change in the temperature- responsive polymer causes the cells to detach.
  • Cells are harvested from the substrate as an intact sheet comprising the cells and their associated extracellular matrix.
  • FIGs. 5A and 5B show an example of how a cell sheet can be cultured and harvested from a temperature-responsive surface. Shown here is a cross-section side view of a sheet of cells being cultured on a temperature-responsive surface of poly-NIPAAm.
  • FIG. 5A shows a monolayer of cells 20 grown on the substrate 24 along with its extracellular matrix 22. At 37° C, the cells attach to the temperature-responsive substrate. As shown in FIG. 5B, when the temperature is reduced to 25° C, the temperature-responsive substrate 24 becomes hydrophilic, causing the cells to detach. The monolayer of cells 20 along with the extracellular matrix 22 can be detached and harvested as an intact sheet without disturbing cell-cell and cell- ECM connections. As seen here, the temperature-responsive substrate 24 is left intact.
  • the monolayer of cells 20 is free of any scaffolding.
  • thermoresponsive polymers examples include thermoresponsive polymers that can be used are described in [Ward et al, "Thermoresponsive Polymers for Biomedical Applications” (2011) Polymers, 3:1215-1242]. Particularly useful thermoresponsive polymers are those that show thermoresponsivity in aqueous solution. Particularly useful thermoresponsive polymers are those that have its LCST in the range of 0 to 100° C, and more particularly, at a relatively low temperature, i.e. in the range of 15 - 35° C.
  • Thermoresponsive polymers that could be used include poly(/V,/V- diethylacrylamide) with an LCST over the range of 25 - 32° C, poly(/V-vinlycaprolactam) with an LCST over the range 25 - 35° C, poly[2-(dimethylamino)ethyl methacrylate)] with a LCST of around 50° C, and poly(ethylene glycol) with a LCST of around 85° C, poly(vinylmethyl ether), poly(hydroxyethyl methacrylate), and hydroxypropylcellulose.
  • thermoresponsive polymer can be tuned by selecting its backbone com position (e.g. incorporating different hydrophilic or hydrophobic co-monomers or copolymers), its molecular weight, side groups or grafted groups, or the architecture of the polymer network (e.g. cross-linking, etc.). Properties that can be altered include its phase temperature (such as LCST), pH responsiveness, swelling/deswelling kinetics, speed of thermoresponsiveness, and degradability. In particular, it may be useful to tune the phase temperature of the polymer to around physiological temperature (37° C) to tailor its use for biological applications. In the case of poly-NI PAAm, its LCST can be tailored by incorporating hydrophilic or hydrophobic co-monomers into the polymer structure. See [Tekin et a I,
  • poly-N IPAAm “Thermoresponsive Platforms for Tissue Engineering and Regenerative Medicine” (2011 Dec) AIChE Journal, 57(12):3249-3258].
  • Other examples of poly-N IPAAm that can be used are described in [Ta ng et al, "Recent development of temperature-responsive surfaces and their application for cell sheet engineering” (2014 Nov) Regenerative Biomaterials, 1(1):91-102].
  • the LCST of poly-N IPAAm can be modified by incorporating hydrophilic co- monomers such as acrylamide, /V-methyl-/V-vinylacetamide, /V-vinylacetamide, and /V-vinyl-2- pyrrolidinone via free-radical polymerization.
  • Copolymers of poly-NIPAAm with two distinct LCSTs can be fabricated by using oligomers, such as carboxy-terminated oligo-NIPAAm, oligo(/V- vinylcaprolactam), and a random co-oligomer of NIPAAm and acrylamide.
  • the thermoresponsive polymer is formed into a hydrogel layer.
  • Hydrogels are polymer networks dispersed in an aqueous solution such that they take on semisolid states containing a high water-to-polymer ratio. Hydrogels can be either a physical gel or a covalently cross-linked gel. See [Ward et al, supra]. In cross-linked gels, the polymer chains are covalently cross-linked together. Physical gels, on the other hand, are formed by the physical entanglement of polymer chains in solution and not from covalently linked polymer chains. Both of these gels, crosslinked and physical, have the ability to swell in a solvent depending on their compatibility with the solvent.
  • thermoresponsive polymers swell/shrink with hydration in response to temperature
  • physical gels show a sol-gel transition with hydration.
  • a cross-linked hydrogel swells in an aqueous solution but does not dissolve therein.
  • Another way to make the thermoresponsive polymers insoluble in an aqueous solution and give them gel-like properties is to covalently graft them onto the substrate.
  • the substrate surface can be made with a temperature-responsive polymer in any suitable way. Examples of techniques for making the temperature-responsive polymer layer are described in WO 2013/151755 (Kim et al, Univ.
  • thermoresponsive polymer film 30 deposited onto a grooved foundation 32.
  • thermoresponsive polymer film 30 constitutes the surface of the substrate. This can be performed by various techniques including in-situ polymerization of precursor monomers, depositing a film of the thermoresponsive polymer onto the substrate surface, or grafting the thermoresponsive polymer onto the substrate surface. Coating methods that can be used include electrostatic coating and physical coating. One possible technique for in-situ
  • polymerization and grafting is UV irradiation.
  • this can be performed by applying a thin layer of NIPAAm monomer and benzophenone (a photoinitiator) dissolved in a solvent (e.g. 2-propanol) onto a textured polystyrene surface.
  • UV irradiation causes polymerization of the NIPAAm monomers and grafting onto the polystyrene surface.
  • the grafted polymer thickness can be controlled by monomer concentrations and radiation energy.
  • EB electron beam
  • This can be performed by applying a thin layer of NIPAAm monomer solution spread on a textured polystyrene surface. Exposing the monomer solution to electron beam irradiation causes polymerization of the NIPAAm monomers and grafting onto the polystyrene surface.
  • the grafted polymer thickness can be controlled by monomer concentrations and radiation energy.
  • thermoresponsive polymer bed 40 on a glass dish base 42.
  • the grooves are imprinted directly into the thermoresponsive polymer bed 40.
  • This direct patterning of the thermoresponsive polymer bed 40 can be performed by any of a variety of possible techniques including soft lithography, photolithography, photopolymerization, micromoulding, embossing, or any of the patterning techniques described above.
  • the temperature-responsive polymer layer may have any suitable thickness (e.g. in the range of 5 - 200 nm). Among the factors that may affect the cell adhesion and detachment are film thickness, surface wettability, polymer chain mobility, grafting density, and swelling ratio. These parameters can be optimized for the best effect.
  • the substrate surface may comprise other materials to tune the properties of the surface, including biodegradable materials such as peptides, proteins (e.g. zein protein), alginate, and gelatin. Such additional materials may be incorporated into the network of temperature-responsive polymers without chemical cross-linking.
  • the functional keratocytes are cultured on the grooved substrate to influence the alignment of the functional keratocytes.
  • the alignment of the cells can be measured by their orientation angle, which is the angle between the longest axis 48 of the cell boundary and the groove direction, ranging from 0 to 90°.
  • an ellipse 36 can be drawn around the cell 38 to approximate the shape of the cell 38.
  • the long axis 48 of the approximated ellipse can be considered the long axis of the cell 38.
  • the orientation angle could also be measured between the long axis of the cell nucleus 46 and the direction of the grooves.
  • the orientation angle of the cells can be determined by any other suitable cell alignment analysis, such as those described in [Koo et al, "Human Corneal Keratocyte Response to Micro- and Nano-Gratings on Chitosan and PDMS” (2011 Sept) Cellular and Molecular Bioengineering, 4(3):399-410], [Teixeira et a I, "Responses of human keratocytes to micro- and nanostructured substrates” (2004 Dec) J Biomed Mater Res A, 71(3):369-376], [Pot et al, "Nanoscale
  • the functional keratocytes are non-randomly oriented (i.e.
  • the method comprises culturing the functional keratocytes until they have a non-random orientation.
  • the mean orientation angle of the functional keratocytes on the sheet is 30° or less.
  • the orientation angle of the cells can also be characterized by the distribution of the cell orientation angles.
  • at least 75% of the functional keratocytes on the sheet have an orientation angle of 30° or less.
  • the grooved substrate may also promote the alignment of extracellular matrix components produced by the functional keratocytes. This may help imitate the structure of the stromal lamellae in which the collagen fibrils are tightly packed, aligned, and parallel. Proper organization of the extracellular matrix components is thought to contribute to optical transparency.
  • the collagen fibrils in the extracellular matrix produced by the cells may imitate the lattice arrangement in the native stroma in one or more ways, such as fibril diameter size, fibril diameter uniformity, fibril-to- fibril spacing, etc.
  • the collagen fibrils (type I, type V, or type l/V hybrid) in the extracellular matrix are non-randomly oriented (i.e. generally tending to be oriented in a particular axial direction).
  • the method comprises culturing the functional keratocytes until they produce extracellular matrix comprising collagen fibrils (type I, type V, or type l/V hybrid) having a non-random orientation.
  • at least one of the functional keratocytes are non-random orientation.
  • FIG. 9 shows keratocytes 50 being cultured on a grooved substrate.
  • the keratocytes 50 have produced extracellular matrix 52 that contains collagen fibrils 54, among other components.
  • the keratocytes 50 are generally aligned in the direction of the parallel grooves 56.
  • the collagen fibrils 54 are generally aligned in the direction of the parallel grooves 56.
  • Collagen fibril measurements can be performed by any suitable technique, such as those described in [Karamichos et al, "TGF- 3 Stimulates Stromal Matrix Assembly by Human Corneal Keratocyte-Like Cells” (2013 Oct) Investigative Ophthalmology & Visual Science, 54:6612-6619], [Ka ramichos et al., "A Role for Topographic Cues in the Organization of Collagenous Matrix by Corneal Fibroblasts and Stem Cells” (2014 Jan) PLoS ONE, 9(l):e86260], [Wu et al, "The engineering of organized human corneal tissue through the spatial guidance of corneal stromal stem cells” (2012 Feb) Biomoteriols, 33(5):1343-1352], and [Gil et al, supra] .
  • the collagen fibrils may have a relatively small diameter (less than 250 nm) and be monodisperse in diameter size distribution. I n some embodiments, at least 75% of the collagen fibrils have a diameter in the range of 15 - 90 nm.
  • the fibril-to-fibril spacing of the collagen fibrils may be spaced with an average spacing of less than 400 nm; but other spacing distances are possible as well.
  • the sheet may contribute to the sheet being transparent.
  • the sheet is transparent.
  • the transparent sheet transmits at least 80% of incident white light.
  • the harvested cell sheets can be used individually, or may be stacked on top of one another to form a multi-layer (two or more) tissue construct.
  • Each sheet may be 1 - 15 ⁇ in thickness, but other thicknesses are possible as well. Many of such sheets (e.g. up to 400) may be stacked to form the multi-layer tissue construct.
  • the multi-layer tissue construct is transparent. In some cases, the multi-layer tissue construct transmits at least 60% of incident white light.
  • Adjacent sheets in the stack may be oriented at an angle (non-parallel) relative to each other (e.g. orthogonally).
  • the direction of orientation for a sheet may be the mean axis of orientation for the functional keratocytes or the collagen fibers, or it may be the orientation of the grooves on the substrate on which the cell sheets are cultu red.
  • the functional keratocytes in different layers may form interconnections with each other (e.g. dendrites of neighboring cells contacting each other).
  • the functional keratocytes may also migrate to different layers (i.e. vertical migration).
  • the multi-layer tissue construct may be maintained in culture for a sufficient duration for such interconnections or cell migrations to occur.
  • the multi-layer tissue construct is further cultured for at least five days to allow for such interconnections or cell migrations.
  • compression is applied to the multi-layer tissue construct in order to promote adhesion between the layers. This can be performed in any suitable way.
  • the below-described cell sheet manipulator can be used to apply gentle compression on the multi-layer tissue construct.
  • the cell sheets may be harvested or stacked using any suitable technique or apparatus.
  • the cells sheets may be harvested and stacked using a sheet manipulator device, such as those described in [Tang et al, supra] and the automated robotic apparatus described in [Kikuchi et al, "Automatic fabrication of 3-dimensional tissues using cell sheet manipulator technique” (2014) Biomoteriols, 35:2428-35].
  • FIG. 10 shows how stromal sheets can be stacked from multiple separately cultured sheets.
  • construct a cell sheet manipulation device using a plunger 60 and a gelatin hydrogel matrix 64 coated on the bottom of the plunger 60.
  • Cell sheets #1, #2, and #3 are simultaneously and independently cultured at 37° C on cell culture dishes 61, 62, and 63.
  • the hydrogel-coated plunger 60 On the surface of cell sheet #1 and reduce the temperature to 23° C for harvesting. Harvest the cell sheet by lifting the plunger 60. This will recover the cell sheet while avoiding shrinking or folding of the sheet.
  • Moving to culture dish 62 for cell sheet #2 place the plunger 60 on cell sheet #2 so that cell sheet #1 is transferred on top of cell sheet #2.
  • Sheet orientation can be controlled by rotating the plunger 60 as desired (e.g. so that the two sheets are orthogonal to each other). Continue incubating at 37° C to promote adhesion between the two cell sheets.
  • An epithelium or endothelium can also be added to the multi-layer tissue construct, which could be incorporated into the multi-layer tissue construct in any suitable way.
  • One possible way is to separately grow the epithelium cell layer and stack the epithelium cell layer on the harvested stromal sheets, or the reverse; similarly for the endothelium layer.
  • Another possible way is to seed epithelium or endothelial cells on top of the stack of stromal sheets.
  • corneal endothelial cells could be seeded on top of the stromal stack and cultured in endothelial growth medium.
  • limbal epithelial cells could be seeded on the other side of the stromal stack and cultured in epithelial cell medium.
  • FIG. 11 shows an example of a multi-layer corneal tissue construct made according to the invention.
  • Three stromal sheets of functional keratocytes are stacked on top of one another such that they are oriented at an angle relative to each other. This stack of stromal sheets is sandwiched between a sheet of epithelial cells and a sheet of endothelial cells.
  • the multi-layer tissue construct may have relatively lower cellular content.
  • the functional keratocytes occupy a volume of less than 50% of the volume of the multi-layer tissue construct.
  • the average density of the functional keratocytes in the multi-layer tissue construct is less than 75,000 cells/mm 3 . In some embodiments, the average density of the functional keratocytes in the multi-layer tissue construct is less than 38,000 cells in a 1 mm 2 column.
  • Mechanical stimuli may enhance the organization, strength, or optical properties of corneal tissue. See [Ruberti et a I, "Prelude to corneal tissue engineering - Gaining control of collagen organization” (2008 Sept) Prog Retin Eye Res., 27(5):549-577] .
  • the cell sheet or the multi-layer tissue construct is subject to mechanical stimuli.
  • the temperature-responsive polymer could be exploited to apply the mechanical stimuli.
  • the temperature could be reduced to cause the substrate's temperature-responsive surface to swell, applying mechanical stress on the sheet.
  • the temperature could then be raised back to physiologic temperature to cause the temperature-responsive polymer to shrink and continue culturing the cell sheet. This cycle could be repeated multiple times while culturing the cell sheet to repeatedly apply mechanical stress to the sheet.
  • the functional keratocytes may be induced to undergo apoptosis. This may reduce the number or density of cells in the cell sheet or the multi-layer tissue construct, which may enhance optical transparency. Any suitable way of inducing apoptosis can be used, including exposing the functional keratocytes to an apoptosis- inducing agent.
  • an apoptosis-inducing agent is a proinflammatory cytokine, such as interleukin-1 (e.g.
  • IL-la platelet-activating factor
  • PAF platelet-activating factor
  • FAS-ligand also known as FasL or CD95L
  • IFN- ⁇ interferon gamma
  • TNF-a tumor necrosis factor alpha
  • an apoptosis-inducing agent is a growth factor.
  • Certain growth factors such as FGF-2 (basic fibroblast growth factor) and PDGF-BB (platelet-derived growth factor) have been shown to induce apoptosis.
  • Another potential apoptosis-inducing agent is nitric oxide.
  • the functional keratocytes are exposed to an apoptosis-inducing agent that induces apoptosis of the functional keratocytes.
  • the functional keratocytes in a cell sheet are induced to undergo apoptosis.
  • the functional keratocytes in a multi-layer tissue construct are induced to undergo apoptosis. This may involve exposing the functional keratocytes to an apoptosis-inducing agent.
  • compression is applied onto the sheet or the multi-layer tissue construct in order to promote keratocyte apoptosis.
  • This can be performed in any suitable way.
  • the above-described cell sheet manipulator can be used to apply gentle compression on the cell sheet or the multi-layer tissue construct.
  • the harvested sheet or the multi-layer tissue construct could be used for implantation into a patient's eye.
  • the harvested sheet or the multi-layer tissue construct may be processed into a structure that is suitable for implantation.
  • the harvested sheet or the multi-layer tissue construct could be made or processed to have a round, oval, or elliptical shape (e.g. by cutting or punching).
  • the harvested sheet or multi-layer tissue construct could be used in corneal
  • the harvested sheet or the multilayer tissue construct may be sufficiently sturdy to be sutured into place (i.e. sufficiently sturdy to hold a suture).
  • implanting the harvested sheet or the multi-layer tissue construct comprises suturing it into the patient's eye.
  • FIGs. 12A - 12C show a corneal replacement surgery using an example engineered corneal tissue of the invention.
  • FIG. 12A shows a cloudy and scratched cornea 70 that needs replacement.
  • the surgeon cuts an incision 74 and removes the damaged cornea 70.
  • the engineered corneal tissue 76 is implanted into the eye 72 and anchored into place with sutures 78.
  • the functional keratocytes may be induced to undergo apoptosis after implantation. Applying a contact lens onto the eye has been shown to cause keratocyte apoptosis. See
  • the method further comprises applying a contact lens on the eye over the implanted corneal tissue.
  • Substrate fabrication Fabricate the cell culture substrate as described in [Jiao et al, supra] and [Kim et al (2010), supra], which are incorporated by reference herein. In more detail, make the substrate with polyurethane-acrylate:polyglycidyl-methacrylate (pUA-pGMA), imprint the grooved pattern by capillary force lithography, and apply a coating of poly-NIPAAm onto the substrate by grafting.
  • pUA-pGMA polyurethane-acrylate:polyglycidyl-methacrylate
  • Tissue collection and cell culture Isolate human corneal keratocytes and passage them as previously described in [Karamichos et al, "TGF- ⁇ Stimulates Stromal Matrix Assembly by Human Corneal Keratocyte-Like Cells” (2013 Oct) Investigative Ophthalmology & Visual Science, 54:6612-6619], which is incorporated by reference herein. Seed the corneal keratocytes on the polyNIPAAm-coated grooved substrates at the appropriate optimized density (e.g. 5,000 - 15,000 cells/cm 2 ). Culture the cells in Eagle's Minimum Essential Medium with fetal bovine serum, TGF- 3, and 2-O-a-D-glucopyranosyl-L-ascorbic acid. Continue the culture for a suitable duration (e.g. 4 weeks). Examine the morphology of the cultures under a microscope (e.g. brightfield or phase contrast microscopy).
  • a microscope e.g. brightfield or phase contrast microscopy.
  • Cell sheet adhesion If improved cell sheet adhesion is needed, this can be achieved by adding an adhesive mixture into the gelatin solution. Add to the gelatin solution, fibrinogen (for example, about 20% wt/vol) and riboflavin-5'-phosphate (for example, about 2.5% wt/vol). After the three sheets are stacked, irradiate with long wavelength UV-A light (365 nm) at 3 mW/cm 2 intensity. This treatment protocol has been shown to adhere stromal layers in a corneal flap.
  • fibrinogen for example, about 20% wt/vol
  • riboflavin-5'-phosphate for example, about 2.5% wt/vol
  • corneal stromal stem cells Alternate cell source using corneal stromal stem cells. Isolate human corneal stromal stem cells as previously described in [Wu et al, "The engineering of organized human corneal tissue through the spatial guidance of corneal stromal stem cells” (2012 Feb) Biomaterials, 33(5):1343-1352], which is incorporated by reference herein. From donor corneas, isolate human corneal stromal stem cells by collagenase-digestion of limbal stromal tissue.
  • SCGM stem cell growth medium
  • DMEM Dulbecco's Modified Eagle Medium
  • MCDB-201 fetal bovine serum
  • epidermal growth factor epidermal growth factor
  • platelet-derived growth factor insulin
  • transferrin selenous acid
  • linoleic acid-bovine serum albumin ascorbic acid-2-phosphate
  • dexamethasone penicillin, streptomycin, gentamicin, and cholera toxin. Passage the cells several times to use in the experiments.
  • thermoresponsive grooved substrates Seed the human corneal stromal stem cells onto the thermoresponsive grooved substrates at the appropriate optimized density (e.g. 7.0 ⁇ 10 4 cells/cm 2 ) and incubate in SCGM. After three days incubation in SCGM, change the culture medium to keratocyte differentiation medium consisting of Advanced DMEM (Life Technologies) supplemented with basic fibroblast growth factor, ascorbic acid-2-phosphate, L-glutamine, gentamicin, and penicillin. Change the media twice weekly and continue culture for a suitable duration (e.g. up to six weeks).
  • Advanced DMEM Advanced DMEM
  • any use of the word “or” herein is intended to be inclusive and is equivalent to the expression “and/or,” unless the context clearly dictates otherwise.
  • the expression “A or B” means A, or B, or both A and B.
  • the expression “A, B, or C” means A, or B, or C, or any combination thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Ophthalmology & Optometry (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Transplantation (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Dermatology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Abstract

On décrit un tissu cornéen mis au point par génie biologique, qui est obtenu par la culture de kératinocytes fonctionnels sur un substrat rainuré revêtu d'un polymère sensible à la température, tel qu'un poly(N-isopropylacrylamide). Pour récolter la feuille cellulaire, la température est abaissée de sorte que la feuille cellulaire se détache du substrat. Le substrat rainuré peut favoriser l'alignement des kératocytes fonctionnels ou des constituants de la matrice extracellulaire produits par les kératocytes fonctionnels. Les feuilles cellulaires récoltées peuvent être empilées les unes sur les autres pour former une structure tissulaire multicouche. Les kératocytes fonctionnels peuvent résulter de l'ensemencement du substrat avec des cellules progénitrices ou des cellules souches, et amener les cellules à se différencier en kératocytes fonctionnels.
PCT/US2016/045216 2015-08-02 2016-08-02 Tissu cornéen mis au point par génie biologique WO2017023955A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562200087P 2015-08-02 2015-08-02
US62/200,087 2015-08-02

Publications (1)

Publication Number Publication Date
WO2017023955A1 true WO2017023955A1 (fr) 2017-02-09

Family

ID=57943611

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/045216 WO2017023955A1 (fr) 2015-08-02 2016-08-02 Tissu cornéen mis au point par génie biologique

Country Status (1)

Country Link
WO (1) WO2017023955A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112512601A (zh) * 2018-07-31 2021-03-16 罗基医疗保健公司 多层细胞膜片的制备方法及利用其制备的多层细胞膜片
US11186819B2 (en) * 2018-06-06 2021-11-30 Zhongshan Ophthalmic Center, Sun Yat-Sen University Methods of serum-free culturing corneal limbal stromal stem cells and inducing sphere formation and differentiation in vitro

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1600177A1 (fr) * 2003-02-20 2005-11-30 Cellseed Inc. Feuille de cellules endotheliales pour regeneration de la cornee, leurs methodes de preparation et d'utilisation

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1600177A1 (fr) * 2003-02-20 2005-11-30 Cellseed Inc. Feuille de cellules endotheliales pour regeneration de la cornee, leurs methodes de preparation et d'utilisation

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
NARA SHARDA ET AL.: "Strategies for faster detachment of corneal cell sheet using micropatterned thermoresponsive matrices.", JOURNAL OF MATERIALS CHEMISTRY B, vol. 3, 2015, pages 4155 - 4169, XP055362583 *
NISHIDA KOHJI ET AL.: "Functional bioengineered corneal epithelial sheet grafts from corneal stem cells expanded ex vivo on a temperature-responsive cell culture surface.", TRANSPLANTATION, vol. 77, no. 3, 1 February 2004 (2004-02-01), pages 379 - 385, XP002988152 *
TANG ZHONGLAN ET AL.: "Recent development of temperature-responsive surfaces and their application for cell sheet engineering.", REGENERATIVE BIOMATERIALS, 2014, pages 91 - 102, XP055362594 *
YANG JOSEPH ET AL.: "Corneal epithelial stem cell delivery using cell sheet engineering: Not lost in transplantation.", JOURNAL OF DRUG TARGETING, vol. 14, no. 7, 2006, pages 471 - 482 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11186819B2 (en) * 2018-06-06 2021-11-30 Zhongshan Ophthalmic Center, Sun Yat-Sen University Methods of serum-free culturing corneal limbal stromal stem cells and inducing sphere formation and differentiation in vitro
CN112512601A (zh) * 2018-07-31 2021-03-16 罗基医疗保健公司 多层细胞膜片的制备方法及利用其制备的多层细胞膜片
EP3831419A4 (fr) * 2018-07-31 2022-04-20 Rokit Healthcare Inc. Procédé de fabrication d'une feuille de cellules multicouche, et feuille de cellules multicouche fabriquée au moyen dudit procédé

Similar Documents

Publication Publication Date Title
JP6140240B2 (ja) 組織工学的に作製された絹製臓器
CA2848405C (fr) Fabrication de feuille d'hydrogel de gelatine pour transplantation d'endothelium corneen
JP4921353B2 (ja) 培養細胞シート、製造方法及びその利用方法
JP5946046B2 (ja) ヒト角膜内皮細胞シート
US20050019488A1 (en) Layered aligned polymer structures and methods of making same
MX2015002710A (es) Metodos de generacion de tejidos.
WO2019198086A1 (fr) Greffon cornéen biologiquement modifié et ses procédés de préparation
JP6744008B2 (ja) 構造化セルロースパッチ又は要素の製造方法、及びそのような方法を使用して製造されたデバイス
Lai et al. Functional biomedical polymers for corneal regenerative medicine
WO2007043255A1 (fr) Feuillet endothélial cornéen cultivé et son procédé de production
JP6240997B2 (ja) ガラス化後のハイドロゲル膜の製造方法、ハイドロゲル材料の製造方法、ガラス化後のハイドロゲル膜、ガラス化後のハイドロゲル膜の乾燥体および細胞シート
Jia et al. Advances in 3D bioprinting technology for functional corneal reconstruction and regeneration
WO2017023955A1 (fr) Tissu cornéen mis au point par génie biologique
Hussain et al. Scaffold-free and scaffold-based cellular strategies and opportunities for cornea tissue engineering
Hassan et al. Amine functional hydrogels as selective substrates for corneal epithelialization
Green et al. New biomimetic directions in regenerative ophthalmology
Kishore et al. Application of hydrogels in ocular tissue engineering
Thomas et al. Print me a cornea-Are we there yet?
CN109464705B (zh) 一种rpe细胞片及其应用和制备方法
Evans et al. Synthetic corneal implants
Venugopal et al. Stem cell–based therapeutic approaches toward corneal regeneration
US20230035013A1 (en) Ophthalmological device for the treatment of lscd and substrate for use in same
Shrestha Two-photon polymerization of poly (caprolactone) and extracellular matrix polymer scaffolds for retinal tissue engineering
CN116831781A (zh) 角膜基质透镜制作方法及角膜基质透镜
Li et al. Cultivation of periodontal tissue cell sheet by a new way for cell sheet engineering

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: 16833751

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: 16833751

Country of ref document: EP

Kind code of ref document: A1