US20190169569A1 - Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells - Google Patents

Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells Download PDF

Info

Publication number
US20190169569A1
US20190169569A1 US15/758,314 US201615758314A US2019169569A1 US 20190169569 A1 US20190169569 A1 US 20190169569A1 US 201615758314 A US201615758314 A US 201615758314A US 2019169569 A1 US2019169569 A1 US 2019169569A1
Authority
US
United States
Prior art keywords
cells
rpe
medium
cell
retinal
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/758,314
Other languages
English (en)
Inventor
Kapil Bharti
Lucas Chase
Feng Xuezhu
Balendu Shekhar Jha
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Fujifilm Cellular Dynamics Inc
Original Assignee
US Department of Health and Human Services
Fujifilm Cellular Dynamics Inc
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 US Department of Health and Human Services, Fujifilm Cellular Dynamics Inc filed Critical US Department of Health and Human Services
Priority to US15/758,314 priority Critical patent/US20190169569A1/en
Assigned to CELLULAR DYNAMICS INTERNATIONAL, INC. reassignment CELLULAR DYNAMICS INTERNATIONAL, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: XUEZHU, FENG, CHASE, LUCAS
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BHARTI, Kapil, JHA, BALENDU SHEKHAR
Publication of US20190169569A1 publication Critical patent/US20190169569A1/en
Assigned to FUJIFILM Cellular Dynamics, Inc. reassignment FUJIFILM Cellular Dynamics, Inc. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CELLULAR DYNAMICS INTERNATIONAL, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/16Activin; Inhibin; Mullerian inhibiting substance
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • 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
    • C12N2533/52Fibronectin; Laminin

Definitions

  • the present invention was made as a result of activities undertaken within the scope of a joint research agreement that was in effect at the time the present invention was made.
  • the parties to said joint research agreement are The Government of the United States of America, U.S. Department of Health and Human Services, as represented by the National Eye Institute, an institute of the National Institutes of Health and Cellular Dynamics International, Inc.
  • This disclosure relates generally to the field of stem cell biology. More particularly, it concerns methods of efficient production of stem cell-derived retinal pigment epithelial cell populations for use as a cell therapy.
  • the retina is a light-sensitive layer of tissue that lines the inner surface of the eye.
  • Photoreceptor cells either rods or cones, in the retina are directly sensitive to light and transform chemical light signals into electrical events that trigger nerve impulses.
  • the retinal pigment epithelium (RPE) is a layer of pigmented cells that forms the blood-retinal barrier.
  • the RPE cells play important roles in the maintenance of visual function and the transport of ions, water, and metabolic end products from the subretinal space to the blood (Strauss et al., 2005). Further, RPE cells establish the immune privilege of the eye by secreting immunosuppressive factors.
  • a disorder or injury to the RPE cells can result in degeneration of the retina, loss of visual function, and blindness.
  • Several disorders of the retina including acute and age-related macular degeneration and Best disease, involve degeneration of the RPE; therefore, cell replacement therapy is a possible therapeutic option for preservation of vision (Buchholz et al., 2009).
  • iPSCs induced pluripotent stem cells
  • iPSCs have been shown to give rise to ocular cells, including RPE cells (Hirami et al., 2009).
  • RPE cells Hirami et al., 2009.
  • all of the techniques known to date for the production of iPSC- or ESC-derived RPE cells are dependent on using a starting population of embryoid bodies.
  • the pluripotent stem cells are induced pluripotent stem cells (iPSCs).
  • iPSCs induced pluripotent stem cells
  • a method for producing human RPE cells comprising (a) obtaining a starting population comprising human induced pluripotent stem cells (iPSCs) that are dissociated into essentially single cells; (b) culturing the iPSCs in a retinal induction medium to initiate differentiation of the cells into retinal lineage cells; (c) further culturing the retinal lineage cells in a retinal differentiation medium to further differentiate the retinal lineage cells; (d) culturing the cells in retinal medium to form differentiating RPE cells; and (e) culturing the RPE cells in a RPE maturation medium, thereby producing human RPE cells.
  • the method does not include the formation of embryoid bodies.
  • the RPE cells does not include the formation of embryoid bodies.
  • the RPE cells does not include the formation of embryoid bodies.
  • the iPSCs are cultured on a matrix.
  • the matrix comprises at least one recombinant cellular adhesion protein such as laminin, vitronectin or fibronectin.
  • the at least one cellular adhesion protein is human.
  • the iPSCs are cultured without a feeder layer. In some aspects, the iPSCs are cultured in a fully-defined culture medium. In other aspects, the iPSCs are cultured in a xeno-free culture medium.
  • the retinal induction medium comprises a WNT pathway inhibitor, a BMP pathway inhibitor, a TGF ⁇ pathway inhibitor and insulin growth factor 1 (IGF1).
  • the WNT pathway inhibitor is selected from the group consisting of N-(2-Aminoethyl)-5-chloroisoquinoline-8-sulphonamide dihydrochloride (CKI-7), N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP2), N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-3-(2-methoxyphenyl)-4-oxothieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP4), 2-Phenoxybenzoic acid-[(5
  • the WNT pathway inhibitor is CKI-7.
  • the BMP pathway inhibitor is 4-(6-(4-(piperazin-1-yl)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline hydrochloride (LDN193189), 6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)-pyrazolo[1,5-a]pyrimidine dihydrochloride (Dorsomorphin), 4-[6-[4-(1-Methylethoxy)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]-quinoline (DMH1), 4-[6-[4-[2-(4-Morpholinyl)ethoxy]phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline (DMH-2), or 5-[6-(4-Methoxyphenyl)pyrazolo
  • the BMP pathway inhibitor is LDN193189.
  • the TGF ⁇ pathway inhibitor is 4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]benzamide (SB431542), 6-[2-(1,1-Dimethylethyl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-4-yl]quinoxaline (SB525334), 2-(5-Benzo[1,3]dioxol-5-yl-2-ieri-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride hydrate (SB-505124), 4-(5-Benzol[1,3]dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)-benzamide hydrate, 4-[4-(1,3-Benzodioxol-5-yl)-benzamide
  • the retinal differentiation medium comprises a WNT pathway inhibitor, a BMP pathway inhibitor, a TGF ⁇ pathway inhibitor, a MEK inhibitor and IGF1.
  • the WNT pathway inhibitor is selected from the group consisting of N-(2-Aminoethyl)-5-chloroisoquinoline-8-sulphonamide dihydrochloride (CKI-7), N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP2), N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-3-(2-methoxyphenyl)-4-oxothieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP4), 2-Phenoxybenzoic acid-[(5
  • the WNT pathway inhibitor is CKI-7.
  • the BMP pathway inhibitor is 4-(6-(4-(piperazin-1-yl)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline hydrochloride (LDN193189), 6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)-pyrazolo[1,5-a]pyrimidine dihydrochloride (Dorsomorphin), 4-[6-[4-(1-Methylethoxy)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]-quinoline (DMH1), 4-[6-[4-[2-(4-Morpholinyl)ethoxy]phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline (DMH-2), or 5-[6-(4-Methoxyphenyl)pyrazolo
  • the BMP pathway inhibitor is LDN193189.
  • the TGF ⁇ pathway inhibitor is 4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]benzamide (SB431542), 6-[2-(1,1-Dimethylethyl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-4-yl]quinoxaline (SB525334), 2-(5-Benzo[1,3]dioxol-5-yl-2-ieri-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride hydrate (SB-505124), 4-(5-Benzo[1,3]dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)-benzamide hydrate, 4-[4-(1,3-Benzodioxol-5-yl)
  • the TGF ⁇ pathway inhibitor is SB431542.
  • the MEK inhibitor is N-[(2R)-2,3-Dihydroxypropoxy]-3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]-benzamide (PD0325901), N-[3-[3-cyclopropyl-5-(2-fluoro-4-iodoanilino)-6,8-dimethyl-2,4,7-trioxopyrido[4,3-d]pyrimidin-1-yl]phenyl]acetamide (GSK1120212), 6-(4-bromo-2-fluoroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide (MEK162), N-[3,4-difluoro-2-(2-fluoro-4-iodoanilino)-6-methoxyphenyl]-1-(2,3-
  • the RPE cells are dissociated after culture in the RPE maturation medium.
  • the dissociated RPE cells are seeded and cultured in RPE maturation medium.
  • the RPE maturation medium comprises a MEK inhibitor.
  • the MEK inhibitor is N-[(2R)-2,3-Dihydroxypropoxy]-3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]-benzamide (PD0325901), N-[3-[3-cyclopropyl-5-(2-fluoro-4-iodoanilino)-6,8-dimethyl-2,4,7-trioxopyrido[4,3-d]pyrimidin-1-yl]phenyl]acetamide (GSK1120212), 6-(4-bromo-2-fluoroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide (MEK162), N-[3,4-difluoro-2-(2-fluoro-4-iodoanilino)-6-methoxyphenyl]-1-(2,3-dihydroxypropyl)cyclopropane-1-
  • the RPE cells are dissociated after culture in the RPE medium and reseeded on a degradable scaffold in the RPE maturation medium thereby producing mature RPE cells.
  • the RPE maturation medium may comprise at least one primary cilium inducer.
  • the at least one primary cilium inducer is prostaglandin E2 (PGE2) or aphidicolin.
  • the RPE maturation medium may comprise N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP2) or 4-(1,3,3a,4,7,7a-Hexahydro-1,3-dioxo-4,7-methano-2H-isoindol-2-yl)-N-8-quinolinyl-Benzamide (endo-IWR1).
  • the starting population of iPSCs is pre-confluent cells that have been dissociated into single cells.
  • the iPSCs are cultured at an initial cell density of about 5,000 to 40,000 cells/cm 2 .
  • the iPSCs are cultured at an initial cell density of 5,000, 10,000, 20,000, 30,000, or 40,000 cells/cm 2 .
  • the starting population of iPSCs is MHC haplotype-matched to a subject in need thereof.
  • the iPSCs are homozygous for at least one HLA allele.
  • the iPSCs are homozygous at HLA-A, HLA-B or HLA-DR.
  • the iPSCs are homozygous at HLA-A and HLA-B.
  • a method for producing human retinal pigment epithelial (RPE) cells comprising (a) obtaining a starting population comprising human induced pluripotent stem cells (iPSCs) that are dissociated into essentially single cells in a fully defined medium; (b) culturing the iPSCs on laminin in a retinal induction medium comprising LDN193189, CKI-7, and SB431542 to initiate differentiation of the cells into retinal lineage cells; (c) further culturing the retinal lineage cells in a retinal differentiation medium comprising LDN193189, CKI-7, SB431542, and PD0325901 to further differentiate the retinal lineage cells; (d) culturing the cells in retinal medium comprising nicotinamide and Activin A to form differentiating RPE cells; and (e) culturing the RPE cells in a RPE maturation medium, thereby producing human RPE cells.
  • the method does not include the formation
  • FIGS. 1A-1D A) Image of pre-confluent iPSCs. B) Example image for day 25 of RPE differentiation. C) Example image for day 40 of RPE differentiation. D) Example images at day 60 after a day 40 replate with culture in RPE-MM at 100 ⁇ brightfield.
  • FIGS. 2A-2B Flow-cytometry analysis of relevant markers including MAP2, NES, PAX6, MITF, PMEL17, TYRP1, CRALBP and BEST1 before cell sorting of iPSC-derived RPE cell population.
  • FIGS. 3A-3C Flow-cytometry analysis of relevant markers including MAP2, NES, PAX6, MITF, PMEL17, TYRP1, CRALBP and BEST1 after cell sorting of an iPSC-derived RPE cell population to remove CD24 positive cells, CD24 positive and CD56 positive cells, CD24 positive and CD90 positive cells, and CD24 positive, CD56 positive, and CD90 positive cells.
  • relevant markers including MAP2, NES, PAX6, MITF, PMEL17, TYRP1, CRALBP and BEST1 after cell sorting of an iPSC-derived RPE cell population to remove CD24 positive cells, CD24 positive and CD56 positive cells, CD24 positive and CD90 positive cells, and CD24 positive, CD56 positive, and CD90 positive cells.
  • FIGS. 4A-4D A) Beta catenin and F-Actin staining of iPSC-RPE untreated cells and iPSC-RPE cells treated with PGE2. Beta catenin staining is seen in the cytoplasm of the untreated cells and at the membrane in treated cells.
  • ERM staining is low in the cytoplasm of the untreated cells and high in the cytoplasm of the treated cells while ZO1 staining is seen at the tight junctions in the plasma membrane of both untreated and treated cells.
  • RPE65 staining is low in the cytoplasm of the untreated cells and high in the cytoplasm of the treated cells.
  • FIGS. 5A-5D A) Beta catenin staining of cells treated with IWP2+endo-IWR1, IWP2, or LiCl. Cells treated with IWP2 or IWP2+endo-IWR1 have beta catenin on cell membrane. Cell treated with LiCl have beta catenin in the nucleus and untreated cells have beta catenin in the cytoplasm. B) p27 staining of cells treated with IWP2+endo-IWR1, IWP2, or LiCl. Cells treated with IWP2 or IWP2+endo-IWR1 have higher p27 expression in the nucleus, suggesting that cells have exited cell cycle.
  • Cells treated with LiCl or untreated cells have weak p27 expression in the nucleus.
  • RPE65 is high in the cytoplasm of the IWP2+IWR1 treated cells and IWP2 cells, low in the untreated cells and no staining is seen in the LiCl treated cells.
  • FIGS. 6A-6D A) Multi-operator RPE Differentiation. Data shown represent RPE differentiations set-up using the optimized protocol across three lines with multiple operators as measured by flow cytometry for the RPE marker Retinaldehyde-binding protein 1(Cra1bp). B) Reproducibility of the RPE differentiation protocol is shown among different starting cell line populations including 3D1, AMD1B, BEST1L, BEST3A, BEST8A, AMD Donor3D, AMD Donor3C and HLA Line A. C-D) Reproducibility of the RPE differentiation protocol is shown among different starting cell line populations. Data represent 109 differentiations performed by five operators on 28 iPSC lines derived from 13 donors. The percentage of Cra1bp-positive cells increased to between 90-100% as compared to the varied purity of the pre-purification cell population.
  • FIGS. 7A-7G A) Functionality of the barrier function of RPE cells generated using the RPE differentiation protocol is shown by transepithelial electric potential (TEP) measurement of the ion gradient across the monolayer.
  • C-E Transepithelial electric resistance (TER) and TEP (lighter line) of untreated cells, PGE2 treated cells and IWP2+endo-IWR1 treated cells.
  • F Functional response (TER) from cells matured with 50 ⁇ M vs. 100 ⁇ M PGE2 in the RPE-MM+PGE2 medium from Day 54 to Day 75 of iPSC-derived differentiation protocol.
  • RPE cells can be derived from pluripotent stem cells such as ES cells and iPSC cells; however, current methods are dependent on a starting population of embryoid bodies.
  • the present disclosure provides a highly efficient and reproducible method of differentiating pluripotent stem cells (PSCs) into functional and mature RPE cells without the use of embryoid bodies. Further embodiments and advantages are described below.
  • purified does not require absolute purity; rather, it is intended as a relative term. Thus, a purified population of cells is greater than about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% pure, or, most preferably, essentially free other cell types.
  • essentially or “essentially free,” in terms of a specified component is used herein to mean that none of the specified component has been purposefully formulated into a composition and/or is present only as a contaminant or in trace amounts.
  • the total amount of the specified component resulting from any unintended contamination of a composition is therefore well below 0.05%, preferably below 0.01%.
  • Most preferred is a composition in which no amount of the specified component can be detected with standard analytical methods.
  • cell is herein used to refer to a structural and functional unit of an organism that can replicate independently, is enclosed by a membrane, and contains biomolecules and genetic material.
  • Cells used herein may be naturally-occurring cells or artificially modified cells (e.g., fusion cells, genetically modified cells, etc.).
  • cell population is used herein to refer to a group of cells, typically of a common type.
  • the cell population can be derived from a common progenitor or may comprise more than one cell type.
  • An “enriched” cell population refers to a cell population derived from a starting cell population (e.g., an unfractionated, heterogeneous cell population) that contains a greater percentage of a specific cell type than the percentage of that cell type in the starting population.
  • the cell populations may be enriched for one or more cell types and depleted of one or more cell types.
  • stem cell refers herein to a cell that under suitable conditions is capable of differentiating into a diverse range of specialized cell types, while under other suitable conditions is capable of self-renewing and remaining in an essentially undifferentiated pluripotent state.
  • stem cell also encompasses a pluripotent cell, multipotent cell, precursor cell and progenitor cell.
  • Exemplary human stem cells can be obtained from hematopoietic or mesenchymal stem cells obtained from bone marrow tissue, embryonic stem cells obtained from embryonic tissue, or embryonic germ cells obtained from genital tissue of a fetus.
  • Exemplary pluripotent stem cells can also produced from somatic cells by reprogramming them to a pluripotent state by the expression of certain transcription factors associated with pluripotency; these cells are called “induced pluripotent stem cells” or “iPSCs”.
  • pluripotent refers to the property of a cell to differentiate into all other cell types in an organism, with the exception of extraembryonic, or placental, cells. Pluripotent stem cells are capable of differentiating to cell types of all three germ layers (e.g., ectodermal, mesodermal, and endodermal cell types) even after prolonged culture.
  • a pluripotent stem cell is an embryonic stem cell derived from the inner cell mass of a blastocyst. In other embodiments, the pluripotent stem cell is an induced pluripotent stem cell derived by reprogramming somatic cells.
  • the term “differentiation” refers to the process by which an unspecialized cell becomes a more specialized type with changes in structural and/or functional properties.
  • the mature cell typically has altered cellular structure and tissue-specific proteins. More specifically, in the context of the present methods indicates the process of a human stem cell acquiring the cell type of a retinal pigment epithelial (RPE) cell with features indicative that said RPE cell is a mature, terminally differentiated cell.
  • RPE retinal pigment epithelial
  • undifferentiated refers to cells that display characteristic markers and morphological characteristics of undifferentiated cells that clearly distinguish them from terminally differentiated cells of embryo or adult origin.
  • Embryoid bodies are aggregates of pluripotent stem cells that can undergo differentiation into cells of the endoderm, mesoderm, and ectoderm germ layers.
  • the spheroid structures form when pluripotent stem cells aggregate and enable the non-adherent culture of EBs in suspension.
  • Isolated cell has been substantially separated or purified from others cells in an organism or culture. Isolated cells can be, for example, at least 99%, at least 98% pure, at least 95% pure or at least 90% pure.
  • an “embryo” refers to a cellular mass obtained by one or more divisions of a zygote or an activated oocyte with an artificially reprogrammed nucleus.
  • An “embryonic stem (ES) cell” is an undifferentiated pluripotent cell which is obtained from an embryo in an early stage, such as the inner cell mass at the blastocyst stage, or produced by artificial means (e.g. nuclear transfer) and can give rise to any differentiated cell type in an embryo or an adult, including germ cells (e.g. sperm and eggs).
  • iPSCs are cells generated by reprogramming a somatic cell by expressing or inducing expression of a combination of factors (herein referred to as reprogramming factors). iPSCs can be generated using fetal, postnatal, newborn, juvenile, or adult somatic cells.
  • factors that can be used to reprogram somatic cells to pluripotent stem cells include, for example, Oct4 (sometimes referred to as Oct 3/4), Sox2, c-Myc, and Klf4, Nanog, and Lin28.
  • somatic cells are reprogrammed by expressing at least two reprogramming factors, at least three reprogramming factors, or four reprogramming factors to reprogram a somatic cell to a pluripotent stem cell.
  • An “allele” refers to one of two or more forms of a gene. Diploid organisms such as humans contain two copies of each chromosome, and thus carry one allele on each.
  • homozygous is defined as containing two of the same alleles at a particular locus.
  • heterozygous refers to as containing two different alleles at a particular locus.
  • haplotype refers to a combination of alleles at multiple loci along a single chromosome.
  • a haplotype can be based upon a set of single-nucleotide polymorphisms (SNPs) on a single chromosome and/or the alleles in the major histocompatibility complex.
  • SNPs single-nucleotide polymorphisms
  • haplotype-matched is defined as the cell (e.g. iPSC cell) and the subject being treated share one or more major histocompatibility locus haplotypes.
  • the haplotype of the subject can be readily determined using assays well known in the art.
  • the haplotype-matched iPSC cell can be autologous or allogeneic.
  • the autologous cells which are grown in tissue culture and differentiated to RPE cells inherently are haplotype-matched to the subject.
  • Substantially the same HLA type indicates that the HLA type of donor matches with that of a patient to the extent that the transplanted cells, which have been obtained by inducing differentiation of iPSCs derived from the donor's somatic cells, can be engrafted when they are transplanted to the patient.
  • a defined medium refers to a medium, an extracellular matrix, or a culture condition in which the chemical composition and amounts of approximately all the components are known.
  • a defined medium does not contain undefined factors such as in fetal bovine serum, bovine serum albumin or human serum albumin.
  • Xeno-Free (XF) when used in relation to a medium, an extracellular matrix, or a culture condition, refers to a medium, an extracellular matrix, or a culture condition which is essentially free from heterogeneous animal-derived components. For culturing human cells, any proteins of a non-human animal, such as mouse, would be xeno components. In certain aspects, the Xeno-free matrix may be essentially free of any non-human animal-derived components, therefore excluding mouse feeder cells or MATRIGELTM.
  • Pre-confluent refers to a cell culture in which the proportion of the culture surface which is covered by cells is about 60-80%. Usually, pre-confluent refers to a culture in which about 70% of the culture surface is covered by cells.
  • the “retina” refers to a light-sensitive layer of tissue which lines the inner surface of the eye.
  • Retinal pigment epithelium refers to a monolayer of pigmented cells between the choroid, a layer filled with blood vessels, and the retina.
  • RIM Retinal Induction Medium
  • the “Retinal Differentiation Medium (RDM)” is defined herein as a medium that comprises a WNT pathway inhibitor, a BMP pathway inhibitor and a MEK inhibitor and differentiates retinal cells.
  • the RDM also comprises a TGF ⁇ pathway inhibitor.
  • the “Retinal Medium (RM)” is defined as a growth medium for the culture of retinal cells comprising Activin A and Nicotinamide.
  • RPE-Maturation Medium herein refers to a medium for the maturation of RPE cells comprising taurine and hydrocortisone.
  • the RPE-MM also comprises triiodothyronine.
  • the RPE-MM may also comprise PD0325901 or PGE2.
  • RPE cells are referred to herein as RPE cells which have downregulated expression of immature RPE markers such as Pax6 and upregulated expression of mature RPE markers such as RPE65.
  • a “therapeutically effective amount” used herein refers to the amount of a compound that, when administered to a subject for treatment of a disease or condition, is sufficient to effect such treatment.
  • Inducer is defined herein as a molecule that regulates gene expression such as activating genes within a cell.
  • An inducer can bind to repressors or activators. Inducers functions by disabling repressors.
  • mouse ES cells Methods for producing mouse ES cells are well known.
  • a preimplantation blastocyst from the 129 strain of mice is treated with mouse antiserum to remove the trophoectoderm, and the inner cell mass is cultured on a feeder cell layer of chemically inactivated mouse embryonic fibroblasts in medium containing fetal calf serum. Colonies of undifferentiated ES cells that develop are subcultured on mouse embryonic fibroblast feeder layers in the presence of fetal calf serum to produce populations of ES cells.
  • mouse ES cells can be grown in the absence of a feeder layer by adding the cytokine leukemia inhibitory factor (LIF) to serum-containing culture medium (Smith, 2000).
  • LIF cytokine leukemia inhibitory factor
  • mouse ES cells can be grown in serum-free medium in the presence of bone morphogenetic protein and LIF (Ying et al., 2003).
  • human ES cells can be grown without serum by culturing the ES cells on a feeder layer of fibroblasts in the presence of basic fibroblast growth factor (Amit et al., 2000). In other methods, human ES cells can be grown without a feeder cell layer by culturing the cells on a protein matrix such as MATRIGELTM or laminin in the presence of “conditioned” medium containing basic fibroblast growth factor (Xu et al., 2001).
  • ES cells can also be derived from other organisms including rhesus monkey and marmoset by previously described methods (Thomson, and Marshall, 1998; Thomson et al., 1995; Thomson and Odorico, 2000), as well as from established mouse and human cell lines.
  • established human ES cell lines include MAOI, MA09, ACT-4, HI, H7, H9, H13, H14 and ACT30.
  • mouse ES cell lines that have been established include the CGR8 cell line established from the inner cell mass of the mouse strain 129 embryos, and cultures of CGR8 cells can be grown in the presence of LIF without feeder layers.
  • pluripotency was originally achieved in 2006 using mouse cells (Yamanaka et al. 2006) and in 2007 using human cells (Yu et al. 2007; Takahashi et al. 2007) by reprogramming of somatic cells via the introduction of transcription factors that are linked to pluripotency.
  • Pluripotent stem cells can be maintained in an undifferentiated state and are capable of differentiating into almost any cell type.
  • the use of iPSCs circumvents most of the ethical and practical problems associated with large-scale clinical use of ES cells, and patients with iPSC-derived autologous transplants may not require lifelong immunosuppressive treatments to prevent graft rejection.
  • any cell can be used as a starting point for iPSCs.
  • cell types could be keratinocytes, fibroblasts, hematopoietic cells, mesenchymal cells, liver cells, or stomach cells.
  • T cells may also be used as a source of somatic cells for reprogramming (U.S. Pat. No. 8,741,648).
  • somatic cell is itself a RPE cells such as a human RPE cell.
  • Somatic cells can be reprogrammed to produce induced pluripotent stem cells (iPSCs) using methods known to one of skill in the art.
  • iPSCs induced pluripotent stem cells
  • One of skill in the art can readily produce induced pluripotent stem cells, see for example, Published U.S. Patent Application No. 20090246875, Published U.S. Patent Application No. 2010/0210014; Published U.S. Patent Application No. 20120276636; U.S. Pat. Nos. 8,058,065; 8,129,187; 8,278,620; PCT Publication NO. WO 2007/069666 A1, and U.S. Pat. No. 8,268,620, which are incorporated herein by reference.
  • nuclear reprogramming factors are used to produce pluripotent stem cells from a somatic cell.
  • at least three, or at least four, of Klf4, c-Myc, Oct3/4, Sox2, Nanog, and Lin28 are utilized.
  • Oct3/4, Sox2, c-Myc and Klf4 are utilized.
  • the cells are treated with a nuclear reprogramming substance, which is generally one or more factor(s) capable of inducing an iPSC from a somatic cell or a nucleic acid that encodes these substances (including forms integrated in a vector).
  • the nuclear reprogramming substances generally include at least Oct3/4, Klf4 and Sox2 or nucleic acids that encode these molecules.
  • a functional inhibitor of p53, L-myc or a nucleic acid that encodes L-myc, and Lin28 or Lin28b or a nucleic acid that encodes Lin28 or Lin28b, can be utilized as additional nuclear reprogramming substances.
  • Nanog can also be utilized for nuclear reprogramming. As disclosed in published U.S. Patent Application No.
  • exemplary reprogramming factors for the production of iPSCs include (1) Oct3/4, Klf4, Sox2, L-Myc (Sox2 can be replaced with Sox1, Sox3, Sox15, Sox17 or Sox18; Klf4 is replaceable with Klf1, Klf2 or Klf5); (2) Oct3/4, Klf4, Sox2, L-Myc, TERT, SV40 Large T antigen (SV40LT); (3) Oct3/4, Klf4, Sox2, L-Myc, TERT, human papilloma virus (HPV)16 E6; (4) Oct3/4, Klf4, Sox2, L-Myc, TERT, HPV16 E7 (5) Oct3/4, Klf4, Sox2, L-Myc, TERT, HPV16 E6, HPV16 E7; (6) Oct3/4, Klf4, Sox2, L-Myc, TERT, Bmil; (7) Oct3/4, Klf4, Sox2, L-Myc, TERT, Bmil; (7) Oct3/
  • Oct3/4, Klf4, Sox2, and c-Myc are utilized.
  • Oct4, Nanog, and Sox2 are utilized, see for example, U.S. Pat. No. 7,682,828, which is incorporated herein by reference.
  • these factors include, but are not limited to, Oct3/4, Klf4 and Sox2.
  • the factors include, but are not limited to Oct 3/4, Klf4 and Myc.
  • Oct3/4, Klf4, c-Myc, and Sox2 are utilized.
  • Oct3/4, Klf4, Sox2 and Sal 4 are utilized.
  • telomeres like Nanog, Lin28, Klf4, or c-Myc can increase reprogramming efficiency and can be expressed from several different expression vectors.
  • an integrating vector such as the EBV element-based system can be used (U.S. Pat. No. 8,546,140).
  • reprogramming proteins could be introduced directly into somatic cells by protein transduction.
  • Reprogramming may further comprise contacting the cells with one or more signaling receptors including glycogen synthase kinase 3 (GSK-3) inhibitor, a mitogen-activated protein kinase kinase (MEK) inhibitor, a transforming growth factor beta (TGF- ⁇ ) receptor inhibitor or signaling inhibitor, leukemia inhibitory factor (LIF), a p53 inhibitor, an NF-kappa B inhibitor, or a combination thereof.
  • GSK-3 glycogen synthase kinase 3
  • MEK mitogen-activated protein kinase kinase
  • TGF- ⁇ transforming growth factor beta
  • LIF leukemia inhibitory factor
  • p53 inhibitor a p53 inhibitor
  • NF-kappa B inhibitor a combination thereof.
  • Those regulators may include small molecules, inhibitory nucleotides, expression cassettes, or protein factors. It is anticipated that virtually any iPS cells or cell lines may be used.
  • iPSCs can be cultured in a medium sufficient to maintain pluripotency.
  • the iPSCs may be used with various media and techniques developed to culture pluripotent stem cells, more specifically, embryonic stem cells, as described in U.S. Pat. No. 7,442,548 and U.S. Patent Pub. No. 2003/0211603.
  • LIF Leukemia Inhibitory Factor
  • bFGF basic fibroblast growth factor
  • pluripotent cells may be cultured on fibroblast feeder cells or a medium that has been exposed to fibroblast feeder cells in order to maintain the stem cells in an undifferentiated state.
  • the cell is cultured in the co-presence of mouse embryonic fibroblasts treated with radiation or an antibiotic to terminate the cell division, as feeder cells.
  • pluripotent cells may be cultured and maintained in an essentially undifferentiated state using a defined, feeder-independent culture system, such as a TESRTM medium (Ludwig et al., 2006a; Ludwig et al., 2006b) or E8TM medium (Chen et al., 2011).
  • the iPSC can be modified to express exogenous nucleic acids, such as to include a tyrosinase enhancer operaby linked to a promoter and a nucleic acid sequence encoding a first marker.
  • exogenous nucleic acids such as to include a tyrosinase enhancer operaby linked to a promoter and a nucleic acid sequence encoding a first marker.
  • the tyrosinase gene is disclosed, for example, in GENBANK® Accession No. 22173, as available on Jan. 1, 2013. This sequence aligns to chromosome 7 of mouse strain C57BL/6 location 5286971-5291691 (invert orientation). A 4721 base pair sequence is sufficient for expression in RPE cells, see Murisier et al., Dev. Biol. 303: 838-847, 2007, which is incorporated herein by reference.
  • This construct is expressed in retinal pigment epithelial cells.
  • Other enhancers can be utilized.
  • Other RPE-specific enhancers include D-MITF, DCT, TYRP1, RPE65, VMD2, MERTK, MYRIP, and RAB27A.
  • Suitable promoters include, but are not limited to, any promoter expressed in retinal pigment epithelial cells including the tyrosinase promoter.
  • the construct can also include other elements, such as a ribosome binding site for translational initiation (internal ribosomal binding sequences), and a transcription/translation terminator. Generally, it is advantageous to transfect cells with the construct.
  • Suitable vectors for stable transfection include, but are not limited to retroviral vectors, lentiviral vectors and Sendai virus.
  • Plasmids have been designed with a number of goals in mind, such as achieving regulated high copy number and avoiding potential causes of plasmid instability in bacteria, and providing means for plasmid selection that are compatible with use in mammalian cells, including human cells. Particular attention has been paid to the dual requirements of plasmids for use in human cells. First, they are suitable for maintenance and fermentation in E. coli , so that large amounts of DNA can be produced and purified. Second, they are safe and suitable for use in human patients and animals. The first requirement calls for high copy number plasmids that can be selected for and stably maintained relatively easily during bacterial fermentation. The second requirement calls for attention to elements such as selectable markers and other coding sequences.
  • plasmids that encode a marker are composed of: (1) a high copy number replication origin, (2) a selectable marker, such as, but not limited to, the neo gene for antibiotic selection with kanamycin, (3) transcription termination sequences, including the tyrosinase enhancer and (4) a multicloning site for incorporation of various nucleic acid cassettes; and (5) a nucleic acid sequence encoding a marker operably linked to the tyrosinase promoter.
  • plasmid vectors that are known in the art for inducing a nucleic acid encoding a protein. These include, but are not limited to, the vectors disclosed in U.S. Pat. Nos. 6,103,470; 7,598,364; 7,989,425; and 6,416,998, which are incorporated herein by reference.
  • a viral gene delivery system can be an RNA-based or DNA-based viral vector.
  • An episomal gene delivery system can be a plasmid, an Epstein-Barr virus (EBV)-based episomal vector, a yeast-based vector, an adenovirus-based vector, a simian virus 40 (SV40)-based episomal vector, a bovine papilloma virus (BPV)-based vector, or a lentiviral vector.
  • Markers include, but are not limited to, fluorescence proteins (for example, green fluorescent protein or red fluorescent protein), enzymes (for example, horse radish peroxidase or alkaline phosphatase or firefly/ renilla luciferase or nanoluc), or other proteins.
  • a marker may be a protein (including secreted, cell surface, or internal proteins; either synthesized or taken up by the cell); a nucleic acid (such as an mRNA, or enzymatically active nucleic acid molecule) or a polysaccharide. Included are determinants of any such cell components that are detectable by antibody, lectin, probe or nucleic acid amplification reaction that are specific for the marker of the cell type of interest.
  • a nucleic acid is included that encodes one or more of MITF, PAX6, TFEC, OTX2, LHX2, VMD2, CFTR, RPE65, MFRP, CTRP5, CFH, C3, C2B, APOE, APOB, mTOR, FOXO, AMPK, SIRT1-6, HTRP1, ABCA4, TIMP3, VEGFA, CFI, TLR3, TLR4, APP, CD46, BACE1, ELOLV4, ADAM 10, CD55, CD59, and ARMS2.
  • Major Histocompatibility Complex is the main cause of immune-rejection of allogeneic organ transplants.
  • the HLA loci are highly polymorphic and are distributed over 4 Mb on chromosome 6.
  • the ability to haplotype the HLA genes within the region is clinically important since this region is associated with autoimmune and infectious diseases and the compatibility of HLA haplotypes between donor and recipient can influence the clinical outcomes of transplantation.
  • HLAs corresponding to MHC class I present peptides from inside the cell and HLAs corresponding to MHC class II present antigens from outside of the cell to T-lymphocytes.
  • HLA-matched stem cell lines may overcome the risk of immune rejection.
  • HLA loci are usually typed by serology and PCR for identifying favorable donor-recipient pairs.
  • Serological detection of HLA class I and II antigens can be accomplished using a complement mediated lymphocytotoxicity test with purified T or B lymphocytes. This procedure is predominantly used for matching HLA-A and -B loci.
  • Molecular-based tissue typing can often be more accurate than serologic testing.
  • Low resolution molecular methods such as SSOP (sequence specific oligonucleotide probes) methods, in which PCR products are tested against a series of oligonucleotide probes, can be used to identify HLA antigens, and currently these methods are the most common methods used for Class II-HLA typing.
  • High resolution techniques such as SSP (sequence specific primer) methods which utilize allele specific primers for PCR amplification can identify specific MHC alleles.
  • MHC compatibility between a donor and a recipient increases significantly if the donor cells are HLA homozygous, i.e. contain identical alleles for each antigen-presenting protein. Most individuals are heterozygous for MHC class I and II genes, but certain individuals are homozygous for these genes. These homozygous individuals can serve as super donors and grafts generated from their cells can be transplanted in all individuals that are either homozygous or heterozygous for that haplotype. Furthermore, if homozygous donor cells have a haplotype found in high frequency in a population, these cells may have application in transplantation therapies for a large number of individuals.
  • iPSCs can be produced from somatic cells of the subject to be treated, or another subject with the same or substantially the same HLA type as that of the patient.
  • the major HLAs e.g., the three major loci of HLA-A, HLA-B and HLA-DR
  • the somatic cell donor may be a super donor; thus, iPSCs derived from a MHC homozygous super donor may be used to generate RPE cells.
  • the iPSCs derived from a super donor may be transplanted in subjects that are either homozygous or heterozygous for that haplotype.
  • reprogramming factors are expressed from expression cassettes comprised in one or more exogenous episiomal genetic elements (see U.S. Patent Publication 2010/0003757, incorporated herein by reference).
  • iPSCs can be essentially free of exogenous genetic elements, such as from retroviral or lentiviral vector elements.
  • These iPSCs are prepared by the use of extra-chromosomally replicating vectors (i.e., episomal vectors), which are vectors capable of replicating episomally to make iPSCs essentially free of exogenous vector or viral elements (see U.S. Pat. No. 8,546,140, incorporated herein by reference; Yu et al., 2009).
  • EBV elements are OriP and EBNA-1, or their variants or functional equivalents.
  • An additional advantage of episomal vectors is that the exogenous elements will be lost with time after being introduced into cells, leading to self-sustained iPSCs essentially free of these elements.
  • lymphotrophic herpes virus is a herpes virus that replicates in a lymphoblast (e.g., a human B lymphoblast) and becomes a plasmid for a part of its natural life-cycle.
  • Herpes simplex virus (HSV) is not a “lymphotrophic” herpes virus.
  • Exemplary lymphotrophic herpes viruses include, but are not limited to EBV, Kaposi's sarcoma herpes virus (KSHV); Herpes virus saimiri (HS) and Marek's disease virus (MDV).
  • KSHV Kaposi's sarcoma herpes virus
  • HS Herpes virus saimiri
  • MDV Marek's disease virus
  • episome-based vectors are contemplated, such as yeast ARS, adenovirus, SV40, or BPV.
  • Pluripotent stem cells can be prepared through the method of somatic cell nuclear transfer.
  • Somatic cell nuclear transfer involves the transfer of a donor nucleus into a spindle-free oocyte.
  • donor fibroblast nuclei from skin fibroblasts of a rhesus macaque are introduced into the cytoplasm of spindle-free, mature metaphase II rhesus macaque ooctyes by electrofusion (Byrne et al., 2007).
  • the fused oocytes are activated by exposure to ionomycin, and then incubated until the blastocyst stage.
  • the inner cell mass of selected blastocysts are then cultured to produce embryonic stem cell lines.
  • the embryonic stem cell lines show normal ES cell morphology, express various ES cell markers, and differentiate into multiple cell types both in vitro and in vivo.
  • RPE cells are produced in the methods disclosed herein.
  • the cells in the retina that are directly sensitive to light are the photoreceptor cells.
  • Photoreceptors are photosensitive neurons in the outer part of the retina and can be either rods or cones. In the process of phototransduction, the photoreceptor cells convert incident light energy focused by the lens to electric signals which are then sent via the optic nerve to the brain.
  • Vertebrates have two types of photoreceptor cells including cones and rods. Cones are adapted to detect fine detail, central and color vision and function well in bright light. Rods are responsible for peripheral and dim light vision. Neural signals from the rods and cones undergo processing by other neurons of the retina.
  • the main functions of the specialized RPE cells include: transport of nutrients such as glucose, retinol, and fatty acids from the blood to the photoreceptors; transport of water, metabolic end products, and ions from the subretinal space to the blood; absorption of light and protection against photooxidation; reisomerization of all-trans-retinol into 11-cis-retinal; phagocytosis of shed photoreceptor membranes; and secretion of various essential factors for the structural integrity of the retina.
  • RPE cells express several proteins that can serve as markers for detection by the use of methodologies, such as immunocytochemistry, Western blot analysis, flow cytometry, and enzyme-linked immunoassay (ELISA).
  • RPE-specific markers may include: cellular retinaldehyde binding protein (CRALBP), microphthalmia-associated transcription factor (MITF), tyrosinase-related protein 1 (TYRP-1), retinal pigment epithelium-specific 65 kDa protein (RPE65), premelanosome protein (PMEL17), bestrophin 1 (BEST1), and c-mer proto-oncogene tyrosine kinase (MERTK).
  • CRALBP retinaldehyde binding protein
  • MITF microphthalmia-associated transcription factor
  • TYRP-1 tyrosinase-related protein 1
  • RPE65 retinal pigment epithelium-specific 65 kDa protein
  • PMEL17 premelanosome protein
  • BEST1 bestrophin 1
  • RPE cells do not express (at any detectable level) the embryonic stem cells markers Oct-4, nanog or Rex-2. Specifically, expression of these genes is approximately 100-1000 fold lower in RPE cells than in ES cells or iPSC cells, when assessed by quantitative RT-PCR.
  • embryoid bodies are produced from iPSCs by the addition of a rho-associated coiled-coil kinase (ROCK) inhibitor and cultured in a first medium comprising two WNT pathway inhibitors and a Nodal pathway inhibitor.
  • the EBs are plated on a MATRIGELTM coated tissue culture in a second medium that does not comprise basic fibroblast growth factor (bFGF), comprises a Nodal pathway inhibitor, comprises about 20 ng to about 90 ng of Noggin, and comprises about 1 to about 5% knock out serum replacement to form differentiating RPE cells.
  • the differentiating RPE cells are cultured in a third medium comprising ACTIVIN and WNT3a.
  • the RPE cells are then cultured in RPE medium that includes about 5% fetal serum, a canonical WNT inhibitor, a non-canonical WNT inhibitor, and inhibitors of the Sonic Hedgehog and FGF pathways to produce human RPE cells.
  • methods are provided for producing RPE cells from an essentially single cell suspension of pluripotent stem cells (PSCs) such as human iPSCs.
  • PSCs pluripotent stem cells
  • the PSCs are cultured to pre-confluency to prevent any cell aggregates.
  • the PSCs are dissociated by incubation with a cell dissociation enzyme, such as exemplified by TRYPSINTM or TRYPLETM.
  • PSCs can also be dissociated into an essentially single cell suspension by pipetting.
  • Blebbistatin e.g., about 2.5 ⁇ M
  • a ROCK inhibitor instead of Blebbistatin may alternatively used to increase PSC survival after dissociated into single cells.
  • the single cell suspension of PSCs is generally counted before seeding.
  • the single cell suspension of PSCs is counted by a hemocytometer or an automated cell counter, such as VICELL® or TC20.
  • the cells may be diluted to a cell density of about 10,000 to about 500,000 cells/mL, about 50,000 to about 200,000 cells/mL, or about 75,000 to about 150,000 cells/mL.
  • the single cell suspension of PSCs is diluted to a density of about 100,000 cells/mL in a fully defined cultured medium such as ESSENTIAL 8TM (E8TM) medium.
  • a culture vessel used for culturing the cell(s) can include, but is particularly not limited to: flask, flask for tissue culture, dish, petri dish, dish for tissue culture, multi dish, micro plate, micro-well plate, multi plate, multi-well plate, micro slide, chamber slide, tube, tray, CELLSTACK® Chambers, culture bag, and roller bottle, as long as it is capable of culturing the stem cells therein.
  • the cells may be cultured in a volume of at least or about 0.2, 0.5, 1, 2, 5, 10, 20, 30, 40, 50 ml, 100 ml, 150 ml, 200 ml, 250 ml, 300 ml, 350 ml, 400 ml, 450 ml, 500 ml, 550 ml, 600 ml, 800 ml, 1000 ml, 1500 ml, or any range derivable therein, depending on the needs of the culture.
  • the culture vessel may be a bioreactor, which may refer to any device or system ex vivo that supports a biologically active environment such that cells can be propagated.
  • the bioreactor may have a volume of at least or about 2, 4, 5, 6, 8, 10, 15, 20, 25, 50, 75, 100, 150, 200, 500 liters, 1, 2, 4, 6, 8, 10, 15 cubic meters, or any range derivable therein.
  • the PSCs such as iPSCs, are generally cultured on culture plates coated by one or more cellular adhesion proteins to promote cellular adhesion while maintaining cell viability.
  • preferred cellular adhesion proteins include extracellular matrix proteins such as vitronectin, laminin, collagen and/or fibronectin which may be used to coat a culturing surface as a means of providing a solid support for pluripotent cell growth.
  • extracellular matrix is recognized in the art.
  • Its components include one or more of the following proteins: fibronectin, laminin, vitronectin, tenascin, entactin, thrombospondin, elastin, gelatin, collagen, fibrillin, merosin, anchorin, chondronectin, link protein, bone sialoprotein, osteocalcin, osteopontin, epinectin, hyaluronectin, undulin, epiligrin, and kalinin.
  • the PSCs are grown on culture plates coated with vitronectin or fibronectin.
  • the cellular adhesion proteins are human proteins.
  • the extracellular matrix (ECM) proteins may be of natural origin and purified from human or animal tissues or, alternatively, the ECM proteins may be genetically engineered recombinant proteins or synthetic in nature.
  • the ECM proteins may be a whole protein or in the form of peptide fragments, native or engineered. Examples of ECM protein that may be useful in the matrix for cell culture include laminin, collagen I, collagen IV, fibronectin and vitronectin.
  • the matrix composition includes synthetically generated peptide fragments of fibronectin or recombinant fibronectin.
  • the matrix composition is xeno-free. For example, in the xeno-free matrix to culture human cells, matrix components of human origin may be used, wherein any non-human animal components may be excluded.
  • the total protein concentration in the matrix composition may be about 1 ng/mL to about 1 mg/mL. In some preferred embodiments, the total protein concentration in the matrix composition is about 1 ⁇ g/mL to about 300 ⁇ g/mL. In more preferred embodiments, the total protein concentration in the matrix composition is about 5 ⁇ g/mL to about 200 ⁇ g/mL.
  • Cells such as RPE cells or PSC, can be cultured with the nutrients necessary to support the growth of each specific population of cells.
  • the cells are cultured in growth media including a carbon source, a nitrogen source and a buffer to maintain pH.
  • the medium can also contain fatty acids or lipids, amino acids (such as non-essential amino acids), vitamin(s), growth factors, cytokines, antioxidant substances, pyruvic acid, buffering agents, and inorganic salts.
  • An exemplary growth medium contains a minimal essential media, such as Dulbecco's Modified Eagle's medium (DMEM) or ESSENTIAL 8TM (E8TM) medium, supplemented with various nutrients, such as non-essential amino acids and vitamins, to enhance stem cell growth.
  • DMEM Dulbecco's Modified Eagle's medium
  • E8TM ESSENTIAL 8TM
  • minimal essential media examples include, but are not limited to, Minimal Essential Medium Eagle (MEM) Alpha medium, Dulbecco's modified Eagle medium (DMEM), RPMI-1640 medium, 199 medium, and F12 medium. Additionally, the minimal essential media may be supplemented with additives such as horse, calf or fetal bovine serum. Alternatively, the medium can be serum free. In other cases, the growth media may contain “knockout serum replacement,” referred to herein as a serum-free formulation optimized to grow and maintain undifferentiated cells, such as stem cell, in culture. KNOCKOUTTM serum replacement is disclosed, for example, in U.S. Patent Application No. 2002/0076747, which is incorporated herein by reference. Preferably, the PSCs are cultured in a fully defined and feeder free media.
  • the culturing temperature can be about 30 to 40° C., for example, at least or about 31, 32, 33, 34, 35, 36, 37, 38, 39° C. but particularly not limited to them.
  • the cells are cultured at 37° C.
  • the CO 2 concentration can be about 1 to 10%, for example, about 2 to 5%, or any range derivable therein.
  • the oxygen tension can be at least, up to, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20%, or any range derivable therein.
  • the Retinal Induction Medium comprises a WNT pathway inhibitor and can result in the differentiation of PSCs to retinal lineage cells.
  • the RIM additionally comprises a TGF ⁇ pathway inhibitor and a BMP pathway inhibitor.
  • Table 3 One exemplary RIM medium is shown in Table 3.
  • the RIM can include DMEM and F12 at about a 1:1 ratio.
  • a WNT pathway inhibitor is included in the RIM, such as CKI-7
  • a BMP pathway inhibitor is included, such as LDN193189
  • the TGF ⁇ pathway inhibitor is included, such as SB431542.
  • the RIM comprises about 5 nM to about 50 nM, such as about 10 nM, of LDN193189, about 0.1 ⁇ M to about 5 ⁇ M, such as about 0.5 ⁇ M, of CKI-7, and about 0.5 ⁇ M to about 10 ⁇ M, such as about 1 ⁇ M, of SB431542.
  • the retinal lineage cells can then be cultured in Retinal Differentiation Medium (RDM) for further differentiation.
  • RDM comprises a WNT pathway inhibitor, a BMP pathway inhibitor, a TGF ⁇ pathway inhibitor and a MEK inhibitor.
  • the RDM comprises a WNT pathway inhibitor, such as CKI-7, a BMP pathway inhibitor, such as LDN193189, a TGF ⁇ pathway inhibitor, such as SB431542, and a MEK inhibitor, such as PD0325901.
  • the RDM can comprise a WNT pathway inhibitor, a BMP pathway inhibitor, a TGF ⁇ pathway inhibitor and a bFGF inhibitor.
  • the concentrations of the Wnt pathway inhibitor, BMP pathway inhibitor and TGF ⁇ pathway inhibitor are higher in the RDM as compared to the RIM, such as about 9 to about 11 times higher, such as about 10 times higher.
  • the RDM comprises about 50 nM to about 200 nM, such as about 100 nM of LDN193189, about 1 ⁇ M to about 10 ⁇ M, such as about 5 ⁇ M, of CKI-7, about 1 ⁇ M to about 50 ⁇ M, such as about 10 ⁇ M, of SB431542, and about 0.1 ⁇ M to about 10 ⁇ M, such as about 1 ⁇ M, 2 ⁇ M, 3 ⁇ M, 4 ⁇ M, 5 ⁇ M, 6 ⁇ M, 7 ⁇ M, 8 ⁇ M, or 9 ⁇ M of PD0325901.
  • Table 3 One exemplary RDM medium is shown in Table 3.
  • the RDM comprises DMEM and F12 at about a 1:1 ratio, knockout serum replacement (e.g., about 1% to about 5%, such as about 1.5%), MEM NEAA, sodium pyruvate, N-2 supplement, B-27 supplement, ascorbic acid and IGF1 (e.g., about 1 ng/mL to about 50 ng/mL, such as about 10 ng/mL).
  • knockout serum replacement e.g., about 1% to about 5%, such as about 1.5%
  • MEM NEAA sodium pyruvate
  • N-2 supplement e.g., sodium pyruvate
  • N-2 supplement e.g., sodium pyruvate
  • B-27 supplement e.g., sodium pyruvate
  • ascorbic acid e.g., about 1 ng/mL to about 50 ng/mL, such as about 10 ng/mL
  • IGF1 e.g., about 1 ng/mL to about 50 ng/
  • the differentiated retinal cells can be even further differentiated by culturing the cells in Retinal Medium (RM).
  • the Retinal Medium comprises Activin A and can additionally comprise Nicotinamide.
  • the RM can comprise about 50 to about 200 ng/mL, such as about 100 ng/mL, of ACTIVIN A, and about 1 mM to about 50 mM, such as about 10 mM, of nicotinamide.
  • the RM can comprise other TGF- ⁇ pathway activators such as GDF1 and/or WNT pathway activators such as WAY-316606, IQ1, QS11, SB-216763, BIO (6-bromoindirubin-3′-oxime), or 2-amino-4-[3,4-(methylenedioxy)benzyl-amino]-6-(3-methoxyphenyl) pyrimidine.
  • the RM can additionally comprise WNT3a.
  • One exemplary RM medium is shown in Table 3.
  • the RM can include DMEM and F12 at about a 1:1 ratio, knockout serum replacement at about 1% to about 5%, such as about 1.5%, MEM non-essential amino acids (NEAA), sodium pyruvate, N-2 supplement, B-27 supplement, and ascorbic acid.
  • the medium can be changed daily with room temperature RM.
  • the cells are generally cultured in the RM for about 8, 9, 10, 11, 12, 13, 14, 15, 16 or 17 days, such as for about 10 days to derive differentiating RPE cells.
  • the cells are preferably cultured in RPE Maturation Medium (RPE-MM).
  • RPE-MM media are shown in Table 3.
  • the RPE-Maturation Medium can comprise about 100 ⁇ g/mL to about 300 ⁇ g/mL, such as about 250 ⁇ g/mL, of taurine, about 10 ⁇ g/L to about 30 ⁇ g/L, such as about 20 ⁇ g/L, of hydrocortisone and about 0.001 ⁇ g/L to about 0.1 ⁇ g/L, such as about 0.013 ⁇ g/L, of triiodothyronine.
  • the RPE-MM can comprise MEM Alpha, N-2 supplement, MEM non-essential amino acids (NEAA), and sodium pyruvate, and fetal bovine serum (e.g., about 0.5% to about 10%, such as about 1% to about 5%).
  • the medium can be changed every other day with room temperature RPE-MM.
  • the cells are generally cultured in RPE-MM for about 5 to about 10 days, such as about 5 days.
  • the cells can then be dissociated, such as with a cell dissociation enzyme, reseeded, and cultured for an additional period of time, such as an additional about 5 to about 30 days, such as about 15 to 20 days, for further differentiation into RPE cells.
  • the RPE-MM does not include a WNT pathway inhibitor.
  • RPE cells can be cryopreserved at this stage.
  • the RPE cells can then be cultured in the RPE-MM for a continued period of time for maturation.
  • the RPE cells are grown in wells, such as a 6-well, 12-well, 24-well, or 10 cm plate.
  • the RPE cells can be maintained in RPE medium for about four to about ten weeks, such as for about six to eight weeks, such as for six, seven, or eight weeks.
  • the cells can be dissociated in a cell dissociated enzyme such as TRYPLETM and reseeded on a degradable scaffold assembly such as in a specialized SNAPWELLTM design for about one to two weeks in RPE-MM with a MEK inhibitor such as PD0325901.
  • the RPE-MM can comprise a bFGF inhibitor instead of the MEK inhibitor.
  • the methods for culturing RPE cells on a degradable scaffold are taught and described in PCT Publication No. WO 2014/121077, which is incorporated herein by reference in its entirety. Briefly, the main components of this method are a CORNING® COSTAR® SNAPWELLTM plate, a bioinert 0-ring, and a biodegradable scaffold. SNAPWELLTM plates provide the structure and platform for the biodegradable scaffolds. The microporous membrane that creates an apical and basal side is ideal for providing support to the scaffold as well as isolating the distinct sides of the polarized layer of cells.
  • the ability of the SNAPWELLTM insert to detach the membrane allows the support ring of the insert to be used an anchor for the scaffold.
  • the resulting differentiated, polarized, and confluent monolayers of functional RPE cells can be cryopreserved at this stage (e.g., in xenofree CS10 medium).
  • mature RPE cells can be further developed into functional RPE cell monolayers that behave as intact RPE tissue by continued culture in the RPE-MM with additional chemicals or small molecules that promote RPE maturation.
  • these small molecules are primary cilium inducers such as prostaglandin E2 (PGE2) or aphidicolin.
  • PGE2 may be added to the medium at a concentration of about 25 ⁇ M to about 250 ⁇ M, such as about 50 ⁇ M to about 100 ⁇ M.
  • the RPE-MM can comprise canonical WNT pathway inhibitors.
  • Exemplary canonical WNT pathway inhibitors are N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP2) or 4-(1,3,3a,4,7,7a-Hexahydro-1,3-dioxo-4,7-methano-2H-isoindol-2-yl)-N-8-quinolinyl-Benzamide (endo-IWR1).
  • the cells are can be cultured in this medium for an additional period of time, such as an additional about one week to about five weeks, such as about another two to four weeks to obtain mature and functional RPE cell monolayers.
  • an additional period of time such as an additional about one week to about five weeks, such as about another two to four weeks to obtain mature and functional RPE cell monolayers.
  • the retinal pigment epithelial cells produced by the methods disclosed herein can be cryopreserved, see for example, PCT Publication No. 2012/149484 A2, which is incorporated by reference herein.
  • the cells can be cryopreserved with or without a substrate.
  • the storage temperature ranges from about ⁇ 50° C. to about ⁇ 60° C., about ⁇ 60° C. to about ⁇ 70° C., about ⁇ 70° C. to about ⁇ 80° C., about ⁇ 80° C. to about ⁇ 90° C., about ⁇ 90° C. to about ⁇ 100° C., and overlapping ranges thereof.
  • lower temperatures are used for the storage (e.g., maintenance) of the cryopreserved cells.
  • liquid nitrogen or other similar liquid coolant
  • the cells are stored for greater than about 6 hours.
  • the cells are stored about 72 hours.
  • the cells are stored 48 hours to about one week.
  • the cells are stored for about 1, 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the cells are stored for 1, 2, 3, 4, 5, 67, 8, 9, 10, 11 or 12 months.
  • the cells can also be stored for longer times.
  • the cells can be cryopreserved separately or on a substrate, such as any of the substrates disclosed herein.
  • cryoprotectants can be used.
  • the cells can be cryopreserved in a cryopreservation solution comprising one or more cryoprotectants, such as DM80, serum albumin, such as human or bovine serum albumin.
  • the solution comprises about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% DMSO.
  • the solution comprises about 1% to about 3%, about 2% to about 4%, about 3% to about 5%, about 4% to about 6%, about 5% to about 7%, about 6% to about 8%, about 7% to about 9%, or about 8% to about 10% dimethylsulfoxide (DMSO) or albumin.
  • DMSO dimethylsulfoxide
  • the solution comprises 2.5% DMSO.
  • the solution comprises 10% DMSO.
  • Cells may be cooled, for example, at about 1° C. minute during cryopreservation.
  • the cryopreservation temperature is about ⁇ 80° C. to about ⁇ 180° C., or about ⁇ 125° C. to about ⁇ 140° C.
  • the cells are cooled to 4° C. prior to cooling at about 1° C./minute.
  • Cryopreserved cells can be transferred to vapor phase of liquid nitrogen prior to thawing for use. In some embodiments, for example, once the cells have reached about ⁇ 80° C., they are transferred to a liquid nitrogen storage area. Cryopreservation can also be done using a controlled-rate freezer.
  • Cryopreserved cells may be thawed, e.g., at a temperature of about 25° C. to about 40° C., and typically at a temperature of about 37° C.
  • WNT is a family of highly conserved secreted signaling molecules that regulate cell-to-cell interactions and are related to the Drosophila segment polarity gene, wingless.
  • the WNT family of genes encodes 38 to 43 kDa cysteine rich glycoproteins.
  • the WNT proteins have a hydrophobic signal sequence, a conserved asparagine-linked oligosaccharide consensus sequence (see e.g., Shimizu et al Cell Growth Differ 8: 1349-1358 (1997)) and 22 conserved cysteine residues. Because of their ability to promote stabilization of cytoplasmic beta-catenin, WNT proteins can act as transcriptional activators and inhibit apoptosis. Overexpression of particular WNT proteins has been shown to be associated with certain cancers.
  • a WNT inhibitor herein refers to WNT inhibitors in general.
  • a WNT inhibitor refers to any inhibitor of a member of the WNT family proteins including Wnt1, Wnt2, Wnt2b, Wnt3, Wnt4, Wnt5A, Wnt6, Wnt7A, Wnt7B, Wnt8A, Wnt9A, Wnt10a, Wnt11, and Wnt16.
  • Certain embodiments of the present methods concern a WNT inhibitor in the differentiation medium.
  • WNT inhibitors include N-(2-Aminoethyl)-5-chloroisoquinoline-8-sulphonamide dihydrochloride (CKI-7), N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-4-oxo-3-phenylthieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP2), N-(6-Methyl-2-benzothiazolyl)-2-[(3,4,6,7-tetrahydro-3-(2-methoxyphenyl)-4-oxothieno[3,2-d]pyrimidin-2-yl)thio]-acetamide (IWP4), 2-Phenoxybenzoic acid-[(5-methyl-2-furanyl)methylene]hydrazide (PNU 74654) 2,4-diamino-quinazoline, quercetin, 3,5,
  • Bone morphogenic proteins are multi-functional growth factors that belong to the transforming growth factor beta (TGF ⁇ ) superfamily BMPs are considered to constitute a group of pivotal morphogenetic signals, orchestrating architecture through the body. The important functioning of BMP signals in physiology is emphasized by the multitude of roles for dysregulated BMP signaling in pathological processes.
  • BMP pathway inhibitors may include inhibitors of BMP signaling in general or inhibitors specific for BMP1, BMP2, BMP3, BMP4, BMP5, BMP6, BMP7, BMP8a, BMP8b, BMP10 or BMP15.
  • Exemplary BMP inhibitors include 4-(6-(4-(piperazin-1-yl)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline hydrochloride (LDN193189), 6-[4-[2-(1-Piperidinyl)ethoxy]phenyl]-3-(4-pyridinyl)-pyrazolo[1,5-a]pyrimidine dihydrochloride (Dorsomorphin), 4-[6-[4-(1-Methylethoxy)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]-quinoline (DMH1), 4-[6-[4-[2-(4-Morpholinyl)ethoxy]
  • TGF ⁇ Transforming growth factor beta
  • TGF- ⁇ is a secreted protein that controls proliferation, cellular differentiation, and other functions in most cells. It is a type of cytokine which plays a role in immunity, cancer, bronchial asthma, lung fibrosis, heart disease, diabetes, and multiple sclerosis. TGF- ⁇ exists in at least three isoforms called TGF- ⁇ 1, TGF- ⁇ 2 and TGF- ⁇ 3.
  • the TGF- ⁇ family is part of a superfamily of proteins known as the transforming growth factor beta superfamily, which includes inhibins, activin, anti-müllerian hormone, bone morphogenetic protein, decapentaplegic and Vg-1.
  • TGF ⁇ pathway inhibitors may include any inhibitors of TGF ⁇ signaling in general.
  • the TGF ⁇ pathway inhibitor is 4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]benzamide (SB431542), 6-[2-(1,1-Dimethylethyl)-5-(6-methyl-2-pyridinyl)-1H-imidazol-4-yl]quinoxaline (SB525334), 2-(5-Benzo[1,3]dioxol-5-yl-2-ieri-butyl-3H-imidazol-4-yl)-6-methylpyridine hydrochloride hydrate (SB-505124), 4-(5-Benzol[1,3]dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)-benzamide hydrate, 4-[4-(1,3-Benzodioxol
  • MEK inhibitor is a chemical or drug that inhibits the mitogen-activated protein kinase enzymes MEK1 or MEK2. They can be used to affect the MAPK/ERK pathway.
  • MEK inhibitors include N-[(2R)-2,3-Dihydroxypropoxy]-3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]-benzamide (PD0325901), N-[3-[3-cyclopropyl-5-(2-fluoro-4-iodoanilino)-6,8-dimethyl-2,4,7-trioxopyrido[4,3-d]pyrimidin-1-yl]phenyl]acetamide (GSK1120212), 6-(4-bromo-2-fluoroanilino)-7-fluoro-N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide (MEK162), N-[3,4-difluoro
  • Basic fibroblast growth factor also known as bFGF, FGF2 or FGF- ⁇
  • bFGF is present in basement membranes and in the subendothelial extracellular matrix of blood vessels.
  • bFGF is a common component of human ESC culture medium in which it is necessary for the cells to remain in an undifferentiated state.
  • bFGF inhibitors herein refer to bFGF inhibitors in general.
  • bFGF inhibitors include, but are not limited to N-[2-[[4-(Diethylamino)butyl]amino-6-(3,5-dimethoxyphenyl)pyrido[2,3-d]pyrimidin-7-yl]-N′-(1,1-dimethylethyl)urea (PD173074), 2-(2-Amino-3-methoxyphenyl)-4H-1-benzopyran-4-one (PD 98059), 1-tert-Butyl-3-[6-(2,6-dichlorophenyl)-2-[[4-(diethylamino)butyl]amino]pyrido[2,3-d]pyrimidin-7-yl]urea (PD161570), 6-(2,6-Dichlorophenyl)-2-[[4-[2-(diethylamino)ethoxy]phenyl]
  • Certain aspects provide a method to produce an RPE or RPE-enriched cell population which can be used for a number of important research, development, and commercial purposes.
  • the methods disclosed herein result in a cell population of at least or about 10 6 , 10 7 , 10 8 , 5 ⁇ 10 8 , 10 9 , 10 10 cells (or any range derivable therein) comprising at least or about 90% (for example, at least or about 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, or any range derivable therein) RPE cells.
  • starting cells for the present methods may comprise the use of at least or about 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 cells or any range derivable therein.
  • the starting cell population may have a seeding density of at least or about 10, 10 1 , 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 cells/ml, or any range derivable therein.
  • RPE cells produced by the methods disclosed herein may be used in any methods and applications currently known in the art for RPE cells.
  • a method of assessing a compound may be provided, comprising assaying a pharmacological or toxicological property of the compound on the RPE cell.
  • a method of assessing a compound for an effect on a RPE cell comprising: a) contacting the RPE cells provided herein with the compound; and b) assaying an effect of the compound on the RPE cells.
  • RPE cells can be used commercially to screen for factors (such as solvents, small molecule drugs, peptides, oligonucleotides) or environmental conditions (such as culture conditions or manipulation) that affect the characteristics of such cells and their various progeny.
  • factors such as solvents, small molecule drugs, peptides, oligonucleotides
  • environmental conditions such as culture conditions or manipulation
  • test compounds may be chemical compounds, small molecules, polypeptides, growth factors, cytokines, or other biological agents.
  • a method includes contacting a RPE cell with a test agent and determining if the test agent modulates activity or function of RPE cells within the population.
  • screening assays are used for the identification of agents that modulate RPE cell proliferation or alter RPE cell differentiation. Screening assays may be performed in vitro or in vivo. Methods of screening and identifying ocular agents or RPE agents include those suitable for high-throughput screening.
  • RPE cells can be positioned or placed on a culture dish, flask, roller bottle or plate (e.g., a single multi-well dish or dish such as 8, 16, 32, 64, 96, 384 and 1536 multi-well plate or dish), optionally at defined locations, for identification of potentially therapeutic molecules. Libraries that can be screened include, for example, small molecule libraries, siRNA libraries, and adenoviral transfection vector libraries.
  • screening applications relate to the testing of pharmaceutical compounds for their effect on retinal tissue maintenance or repair. Screening may be done either because the compound is designed to have a pharmacological effect on the cells, or because a compound designed to have effects elsewhere may have unintended side effects on cells of this tissue type.
  • RPE cells can also provide use of RPE cells to enhance ocular tissue maintenance and repair for any condition in need thereof, including retinal degeneration or significant injury.
  • the cells can first be tested in a suitable animal model.
  • the RPE cells are evaluated for their ability to survive and maintain their phenotype in vivo.
  • Cell compositions are administered to immunodeficient animals (e.g., nude mice or animals rendered immunodeficient chemically or by irradiation). Tissues are harvested after a period of growth, and assessed as to whether the pluripotent stem cell-derived cells are still present.
  • RPE cell suitability and survival can be determined by transplantation (e.g. subcutaneous or subretinal) in matrigel in immunodeficient animals such as NOG mice (Kanemura et al., 2014).
  • the human RPE cells described herein, or a pharmaceutical composition including these cells can be used for the manufacture of a medicament to treat a condition in a patient in need thereof.
  • the RPE cells can be previously cryopreserved.
  • the disclosed RPE cells are derived from iPSCs, and thus can be used to provide “personalized medicine” for patients with eye diseases.
  • somatic cells obtained from patients can be genetically engineered to correct the disease causing mutation, differentiated into RPE, and engineered to form an RPE tissue. This RPE tissue can be used to replace the endogenous degenerated RPE of the same patient.
  • iPSCs generated from a healthy donor or from HLA homozygous “super-donors” can be used.
  • RPE cells can be treated in vitro with certain factors, such as pigment epithelium-derived factor (PEDF), transforming growth factor (TGF)-beta, and/or retinoic acid to generate an anti-inflammatory and immunosuppressive environment in vivo.
  • PEDF pigment epithelium-derived factor
  • TGF transforming growth factor
  • retinoic acid to generate an anti-inflammatory and immunosuppressive environment in vivo.
  • Various eye conditions may be treated or prevented by the introduction of the RPE cells obtained using the methods disclosed herein.
  • the conditions include retinal diseases or disorders generally associated with retinal dysfunction or degradation, retinal injury, and/or loss of retinal pigment epithelium.
  • Conditions that can be treated include, without limitation, degenerative diseases of the retina, such as Stargardt's macular dystrophy, retinitis pigmentosa, macular degeneration (such as age related macular degeneration), glaucoma, and diabetic retinopathy.
  • Additional conditions include Lebers congenital amaurosis, hereditary or acquired macular degeneration, Best disease, retinal detachment, gyrate atrophy, choroideremia, pattern dystrophy, other dystrophies of the RPE, and RPE and retinal damage due to damage caused by any one of photic, laser, inflammatory, infectious, radiation, neovascular or traumatic injury.
  • methods are provided for treating or preventing a condition characterized by retinal degeneration, comprising administering to a subject in need thereof an effective amount of a composition comprising RPE cells. These methods can include selecting a subject with one or more of these conditions, and administering a therapeutically effective amount of the RPE cells sufficient to treat the condition and/or ameliorate symptoms of the condition.
  • the RPE cells may be transplanted in various formats.
  • the RPE cells may be introduced into the target site in the form of cell suspension, or adhered onto a matrix, extracellular matrix or substrate such as a biodegradable polymer, as a monolayer, or a combination.
  • the RPE cells may also be transplanted together (co-transplantation) with other retinal cells, such as with photoreceptors.
  • the RPE cells are produced from iPSCs from the subject to be treated, and thus are autologous.
  • the RPE cells are produced from an MHC-matched donor.
  • the RPE cells can be used for autologous RPE grafts to those subjects suitable for receiving regenerative medicine.
  • the RPE cells may be transplanted in combination with other retinal cells, such as with photoreceptors.
  • Transplantation of the RPE cells produced by the disclosed methods can be performed by various techniques known in the art. For example, methods for performed RPE transplants are described in U.S. Pat. Nos. 5,962,027 and 6,045,791, each of which is incorporated herein by reference in its entirety.
  • the transplantation is performed via pars pana vitrectomy surgery followed by delivery of the cells through a small retinal opening into the sub-retinal space or by direct injection.
  • the RPE cells can be introduced into the target site in the form of cell suspension, adhered onto a matrix, such as extracellular matrix, or provided on substrate such as a biodegradable polymer.
  • the RPE cells can also be transplanted together (co-transplantation) with other cells, such as retinal cells with photoreceptors.
  • a composition comprising RPE cells obtained by the methods disclosed herein is provided.
  • these RPE cells include a tyrosinase enhancer operably linked to a promoter and a nucleic acid encoding a marker.
  • the RPE cells also include a second constitutive promoter operably linked to a nucleic acid encoding a second marker.
  • compositions of the RPE cells produced by the methods disclosed herein can include at least about 1 ⁇ 10 3 RPE cells, about 1 ⁇ 10 4 RPE cells, about 1 ⁇ 10 5 RPE cells, about 1 ⁇ 10 6 RPE cells, about 1 ⁇ 10 7 RPE cells, about 1 ⁇ 10 8 RPE cells, or about 1 ⁇ 10 9 RPE cells.
  • the compositions are substantially purified (with respect to non-RPE cells) preparations comprising differentiated RPE cells produced by the methods disclosed herein.
  • Compositions are also provided that include a scaffold, such as a polymeric carrier and/or an extracellular matrix, and an effective amount of the RPE cells produced by the methods disclosed herein.
  • the cells are provided as a monolayer of cells.
  • the matrix material if generally physiologically acceptable and suitable for use in in vivo applications.
  • the physiologically acceptable materials include, but are not limited to, solid matrix materials that are absorbable and/or non-absorbable, such as small intestine submucosa (SIS), crosslinked or non-crosslinked alginate, hydrocolloid, foams, collagen gel, collagen sponge, polyglycolic acid (PGA) mesh, fleeces and bioadhesives.
  • SIS small intestine submucosa
  • PGA polyglycolic acid
  • Suitable polymeric carriers also include porous meshes or sponges formed of synthetic or natural polymers, as well as polymer solutions.
  • the matrix is a polymeric mesh or sponge, or a polymeric hydrogel.
  • Natural polymers that can be used include proteins such as collagen, albumin, and fibrin; and polysaccharides such as alginate and polymers of hyaluronic acid.
  • Synthetic polymers include both biodegradable and non-biodegradable polymers.
  • biodegradable polymers include polymers of hydroxy acids such as polyactic acid (PLA), polyglycolic acid (PGA) and polylactic acid-glycolic acid (PGLA), polyorthoesters, polyanhydrides, polyphosphazenes, and combinations thereof.
  • Non-biodegradable polymers include polyacrylates, polymethacrylates, ethylene vinyl acetate, and polyvinyl alcohols.
  • a hydrogel is a substance formed when an organic polymer (natural or synthetic) is cross-linked via covalent, ionic, or hydrogen bonds to create a three-dimensional open-lattice structure which entraps water molecules to form a gel.
  • materials which can be used to form a hydrogel include polysaccharides such as alginate, polyphosphazines, and polyacrylates, which are crosslinked ionically, or block copolymers such as PLURONICSTM or TETRONICSTM, polyethylene oxide-polypropylene glycol block copolymers which are crosslinked by temperature or H, respectively.
  • Other materials include proteins such as fibrin, polymers such as polyvinylpyrrolidone, hyaluronic acid and collagen.
  • a reagent system in some embodiments, includes a set or combination of cells comprising a RPE-enriched cell population that exists at any time during manufacture, distribution or use.
  • the cell sets comprise any combination of the cell population disclosed herein in combination with undifferentiated pluripotent stem cells or other differentiated cell types, often sharing the same genome.
  • Each cell type may be packaged together, or in separate containers in the same facility, or at different locations, at the same or different times, under control of the same entity or different entities sharing a business relationship.
  • compositions may optionally be packaged in a suitable container with written instructions for a desired purpose, such as the reconstitution of RPE cell function to improve a disease or injury of the ocular tissue.
  • kits that can include, for example, one or more media and components for the production of RPE cells.
  • the reagent system may be packaged either in aqueous media or in lyophilized form, where appropriate.
  • the container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there is more than one component in the kit, the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial.
  • the components of the kit may be provided as dried powder(s).
  • kits When reagents and/or components are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means.
  • the kits also will typically include a means for containing the kit component(s) in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained.
  • the kit can also include instructions for use, such as in printed or electronic format, such as digital format.
  • a starting population of RPE cells can be derived from pluripotent stem cells such as ES cells and iPSCs.
  • the RPE cells were derived from human iPSCs reprogrammed from somatic cells by methods known in the art such as U.S. Pat. Nos. 8,546,140, 8,741,648, 8,691,574, Published U.S. Patent Application No. 20090246875, Published U.S. Pat. No. 8,278,104, Published U.S. Pat. Nos. 9,005,967, 8,058,065, 8,129,187, PCT Publication NO. WO 2007/069666 A1, U.S. Pat. Nos. 8,183,038 and 8,268,620, which are incorporated herein by reference.
  • the iPSCs were grown without mouse or human feeder layers in fully defined-culture medium, such as ESSENTIAL 8TM (E8TM) medium, on a plate coated by vitronectin.
  • the vitronectin was diluted 1:200 in DPBS without calcium or magnesium and the culture plates were coated with the vitronectin and incubated at room temperature for about 1 hour.
  • the iPSCs were split when they were pre-confluent and not allowed to overgrow to prevent unhealthy and/or differentiated cells ( FIG. 1A ).
  • the iPSCs were dissociated into a single cell suspension to remove any aggregates or embryoid bodies.
  • the cells were washed with DPBS and incubated in a cell dissociation enzyme such as TRYPLETM for about 10 min at 37° C.
  • the cells were then detached by pipetting with a serological pipet and the cell suspension was collected in a conical tube. If the cells did not detach with gently pipetting, the cultures were allowed to incubate longer, such as 2-3 additional minutes.
  • the culture vessel was washed with room temperature E8TM medium and the medium was then added to the tube containing the cell suspension.
  • Blebbistatin e.g.
  • E8TM Medium 2.5 ⁇ M was added to the E8TM Medium to increase PSC survival after dissociation into single cells while the cells are not adhered to a culture vessel. To collect the cells, they were centrifuged at 400 ⁇ g for about 5 minutes, the supernatant was aspirated and the cells were resuspended in an appropriate volume of E8TM medium.
  • the single cell iPSCs seeded at the appropriate cells density were cultured for about 2 days as in Example 1, they were cultured in various differentiation media for deriving RPE cells.
  • the E8TM medium was aspirated and room temperature Retinal Induction Medium (RIM) (e.g., Table 3) was added.
  • RIM comprised DMEM and F12 at about a 1:1 ratio, knockout serum replacement, MEM non-essential amino acids (NEAA), sodium pyruvate, N-2 supplement, B-27 supplement, and ascorbic acid.
  • the RIM comprised a WNT pathway inhibitor, a BMP pathway inhibitor, a TGF ⁇ pathway inhibitor and insulin growth factor 1 (IGF1).
  • IGF1 insulin growth factor 1
  • the cells were next cultured in Retinal Differentiation Medium (RDM) for about seven to fourteen days.
  • RDM Retinal Differentiation Medium
  • Table 2 comprised DMEM and F12 at about a 1:1 ratio, knockout serum replacement, MEM NEAA, sodium pyruvate, N-2 supplement, B-27 supplement, and ascorbic acid.
  • the RDM comprised a WNT pathway inhibitor (e.g., CKI-7), a BMP pathway inhibitor (e.g., LDN193189), a TGF ⁇ pathway inhibitor (e.g., SB431542), and a MEK inhibitor (e.g., PD325901).
  • the concentration of the Wnt pathway inhibitor, BMP pathway inhibitor and TGF ⁇ pathway inhibitor was ten times higher in the RDM as compared to the RIM. Each day the media was aspirated and room temperature RDM was added to the cells to produce differentiated retinal cells.
  • the cells were then cultured in Retinal Medium (RM) for seven to ten days.
  • the RM comprised DMEM and F12 at about a 1:1 ratio, knockout serum replacement, MEM NEAA, sodium pyruvate, N-2 supplement, B-27 supplement and ascorbic acid.
  • the RM comprised Nicotinamide and Activin A. The medium was changed daily with room temperature RM resulting in RPE cells.
  • RPE-MM RPE Maturation Medium
  • the RPE-MM (Table 2) comprised MEM Alpha, fetal bovine serum, N-2 supplement, MEM NEAA, and sodium pyruvate.
  • the RPE-MM contained Taurine, Hydrocortisone and 3,3′,5-Triiodo-L-thyronin ( FIG. 1C ).
  • the medium was changed every other day with room temperature RPE-MM.
  • the cells were then dissociated in a cell dissociation enzyme and reseeded on vitronectin coated plates.
  • the derived PRE cells can be cryopreserved in xenofree CS10 medium.
  • plated cells are cultured for another approximately fifteen days.
  • the cells were dissociated in a cell dissociation enzyme such as TRYPLETM and reseeded on a degradable scaffold assembly in a specialized SNAPWELLTM design for 1-2 weeks in the RPE-MM with a MEK inhibitor such as PD325901.
  • a cell dissociation enzyme such as TRYPLETM
  • MEK inhibitor such as PD325901.
  • the mature RPE cells were further developed into functional RPE cell monolayers that function as an intact RPE tissue by continued culture in the RPE-MM with additional small molecules such as primary cilium inducers like PGE2 or aphidicolin. Without being bound by theory, these primary cilium inducers suppress the canonical WNT pathway, induce cell cycle exit in the cells, and induce apical-basal polarization in the RPE monolayer.
  • RPE maturity can alternatively be induced by canonical WNT pathway inhibitors such as IWP2 and endo-IWR1 that also induce cell cycle exit in RPE cells to promote RPE maturation. The cells were cultured in this medium for another 2-3 weeks to obtain mature and functional RPE cell monolayers.
  • the presently disclosed methods provide mature RPE cells from pluripotent cells that can be consistently reproduced at a large scale for clinical applications.
  • the medium was aspirated and the cells were washed twice with Dulbecco's Phosphate-Buffered Saline (DPBS).
  • DPBS Dulbecco's Phosphate-Buffered Saline
  • the cells were then incubated with a cell dissociation enzyme and the cell suspension was pipetted into a conical tube.
  • the cells were centrifuged, the supernatant aspirated and the cells resuspended in room temperature RPE-MM.
  • the cell suspension was then filtered through a STERIFLIP® cell strainer and the cells were counted.
  • the cells were centrifuged and resuspended at an appropriate density (e.g. 1 ⁇ 10 7 cells/mL) in cold CryoStor® CS10.
  • the cell suspension was aliquoted into pre-labeled cryovials which were placed in a cold freezing container and transferred to a ⁇ 80° C. freezer for 12-24 hours. The vials were then transferred
  • Example 5 MACS Depletion of Contaminating Non-RPE Cells and Enrichment of Starting Population of RPE Cells by CD24, CD56, and/or CD90 Depletion
  • the contaminating cells of the starting population of RPE cells have specific cell surface markers which can be used to separate contaminating cells from the desired mature RPE cells.
  • CD24, CD56, and/or CD90 are cell surface antigens expressed on (but not limited to) pluripotent stem cells and other neural cell types.
  • CD24 is a glycoprotein expressed on the surface of pluripotent stem cells, some B lymphocytes and differentiating neuroblasts.
  • CD56 or neural cell adhesion molecule (NCAM) is a glycoprotein expressed on the surface of neurons and natural killer cells.
  • CD90 or Thy-1 is a marker expressed on the surface of a variety of stem cells as well as neurons.
  • the expression of CD24, CD56 and/or CD90 is lost during differentiation of stem cells to many mature cell types including RPE cells. Therefore, removal of cells positive for CD24, CD56, and/or CD90 results in the depletion of the residual contaminating cells.
  • the cell suspension was collected in RPE-MM Plating Medium and centrifuged at 400 ⁇ g for 5 min.
  • the cell pellet was resuspended in RPE-MM Plating Medium and the cell suspension was filtered through a cell strainer (e.g., 20 ⁇ M steriflip cell strainer) to dissociate any remaining cell clusters.
  • the cell suspension was counted (e.g., using a ViCell counter) for viable cells to obtain a cell concentration.
  • the counted cell suspension provided a single cell suspension that could be used for sorting or the flow cytometry purity assay.
  • MACS was used to deplete the CD24 positive cells, the CD56 positive cells, and/or the CD90 positive cells.
  • MACS buffer such as at 1 ⁇ 10 7 cells/mL.
  • An example MACS buffer is included in Table 3.
  • the cells were stained with an anti-CD24 antibody, an anti-CD56 antibody, and/or an anti-CD90 antibody (each diluted 1:500) and incubated at 4° C. for 20 min to allow the antibody to bind to the antigen on the cells.
  • the antibodies used should be tagged with a label (e.g., FITC) that will bind to the secondary antibody.
  • a label e.g., FITC
  • 20 mL of MACS buffer was added and the cells were centrifuged at 400 ⁇ g for 5 min.
  • the cell pellet was resuspended in 20 mL MACS buffer, vigorously mixed, and centrifuged at 400 ⁇ g for 5 min to remove any unbound antibody.
  • the cell pellet was resuspended in MACS buffer (e.g., at 1.11 ⁇ 10 8 cells/mL), microbeads coated with the diluted (1:10) secondary antibody (e.g., anti-FITC) were added, and the cells were incubated at 4° C. for 20 min.
  • the collected unlabeled cell suspension was centrifuged (400 ⁇ g for 5 min) and replated in RPE-MM Plating Medium, and an aliquot of the cell suspension was used for the flow cytometry purity assay.
  • the MACS cell sorting resulted in a RPE-enriched cell population depleted of cells positive for CD24, CD56, and/or CD90. It is noted that the use of this method is not limited to the starting population resulting from the method detailed in Example 2 and may be employed to remove contaminating cells from a RPE population produced by other methods such as, but not limited to, the methods described in U.S. application Ser. Nos. 12/523,444, and 14/405,730.
  • the pre-sorting percentages of cells positive for the RPE-markers are those present in the starting population of RPE cells of Example 2. Depletion of the combination of CD24 positive cells and CD56 positive cells resulted in greater enrichment of RPE cells than the depletion of only CD24 positive cells. Depletion of CD24 positive cells, CD56 positive cells and CD90 positive cells resulted in more than 99% RPE cells in the cell population.
  • characterization of the RPE cells by a panel of relevant markers including BEST1, CRALBP, TYRP1, PMEL17, MAP2, NES, and MITF was performed (e.g. pre-sorting and post-sorting).
  • the flow cytometry purity assay was performed to obtain a measurement of the percentages (Table 1) of cells positive for each marker before and after removal of CD24 positive cells, CD56 positive cells, and/or CD90 positive cells by MACS ( FIGS. 2 and 3 ).
  • the flow cytometry purity assay was performed to determine the percentage of RPE cells obtained by a sorting method of the present disclosure.
  • An aliquot of the cell suspension collected from the MACS assay (2 ⁇ 10 6 cells in a 5 mL FACS tube per sample) was centrifuged at 400 ⁇ g for 3 min.
  • the cell pellet was resuspended in 1 mL of a stain (e.g., Live-Dead Red stain) and incubated in the dark at room temperature for 15 min. After incubation, 2 mL of wash buffer was added and the cells were centrifuged at 400 ⁇ g for 3 min to remove any unbound stain.
  • a stain e.g., Live-Dead Red stain
  • the cell pellet was resuspended in fixation buffer and incubated in the dark at room temperature for 15 min After incubation, 2 mL of wash buffer were added and the cells were centrifuged at 400 ⁇ g for 3 min and the supernatant was decanted. The cell pellet was resuspended in 2 mL of wash buffer to make a 1 ⁇ 10 6 cells/mL suspension and 200 ⁇ L of the cell suspension was transferred to a FACS tube. To each tube, 2 mL of perm buffer was added and cells were centrifuged at 400 ⁇ g for 3 min. Primary antibodies for the RPE-specific markers were diluted in perm buffer and 100 ⁇ L diluted antibody solution was added to each tube. After incubation overnight at 4° C.
  • the flow cytometry purity assay showed that the MACS sorting to deplete the contaminating cells positive for CD24, CD56, and/or CD90 resulted in a RPE cell-enriched population (95-99%) as compared to the starting cell population (78.6%) as determined by the BEST1 marker.
  • the inclusion of PD0325901 at a concentration of 1 ⁇ M in media for certain windows of time beginning on Day 2 post iPSC plating through the end of the differentiation process, including post-MACS culture may improve both purity of the RPE population (meaning a decrease in contaminating cells) as well as maturity of the resulting RPE population.
  • Inclusion of 1 ⁇ M PD 0325901 has been shown to improve both purity and maturity of the RPE population when included in RDM as well as in RPE-MM (approximately Days 42 through 50) of the RPE process described herein.
  • a decrease in the percentage of fetal bovine serum from 5 percent to 0.5-1 percent in RPE-MM and RPE-MM Plating Medium may improve both purity of the RPE population (meaning a decrease in contaminating cells) as well as maturity of the resulting RPE population.
  • transepithelial electrical potential TEP
  • TER transepithelial electrical resistance
  • RPE differentiation process three iPSC lines were differentiated to RPE cells by multiple operators ( FIG. 6A ).
  • the average purity of the resultant RPE cells was characterized by measurement of the RPE marker Retinaldehyde-binding protein 1(Crap1bp) by flow cytometry (Table 2).
  • the RPE differentiation process was found to be highly reproducible between different starting cell populations as well as different operators.
  • the reproducibility was confirmed by RPE differentiation from different starting cell lines including 3D1, AMD1B, BEST1L, BEST3A, BEST8A, AMD Donor3D and HLA Line A (FIG. 6 B).
  • HLA Line A (21525.102) is an iPSC line produced from a donor homozygous for HLA-A*01 and HLA-B*08 that could provide a beneficial match to 11.38% of the US population. Furthermore, RPE has also been successfully produced using this process using an iPSC line produced from a donor homozygous for HLA-A*03 and HLA-B*07 called HLA Line C (21526.101) that could potentially provide a beneficial match to 7.63% of the US population.
  • HLA Line A (21525.102) and HLA Line C (21526.101) homozygous at HLA-A and HLA-B as described above are the property of Cellular Dynamics International, Inc.
  • the flow cytometry wash buffer was prepared by adding 20 mL FBS to 1000 mL of DPBS (i.e. without calcium and magnesium). The buffer was filter sterilized and can be stored at 4° C. for up to 4 weeks.
  • the flow cytometry perm buffer was prepared by adding 20 mL FBS to 1000 mL DPBS (i.e. without calcium and magnesium). One gram of Saponin was added and mixed well. The buffer was filter sterilized and can be stored at 4° C. for up to 4 weeks.
  • the flow cytometry Live-Dead Red stain was prepared by diluting Live-Dead Stain 1:1000 in DPBS (i.e. without calcium and magnesium). One mL of the stain was prepared per 1 ⁇ 10 6 cells being assayed. The stain was prepared fresh before use.
  • the flow cytometry fixation buffer was prepared by adding 1 mL of 36.5% Formaldehyde to 8.1 mL of DPBS (i.e. without calcium and magnesium). One mL of stain was prepared per 1 ⁇ 10 6 cells being assayed. The buffer was prepared fresh before use.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Neurosurgery (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Neurology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Ophthalmology & Optometry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Materials For Medical Uses (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US15/758,314 2015-09-08 2016-09-07 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells Abandoned US20190169569A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/758,314 US20190169569A1 (en) 2015-09-08 2016-09-07 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201562215579P 2015-09-08 2015-09-08
US15/758,314 US20190169569A1 (en) 2015-09-08 2016-09-07 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells
PCT/US2016/050543 WO2017044483A1 (fr) 2015-09-08 2016-09-07 Méthode de différenciation reproductible de cellules de l'épithélium pigmentaire rétinien de qualité clinique

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/050543 A-371-Of-International WO2017044483A1 (fr) 2015-09-08 2016-09-07 Méthode de différenciation reproductible de cellules de l'épithélium pigmentaire rétinien de qualité clinique

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/931,003 Continuation US20210139847A1 (en) 2015-09-08 2020-07-16 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells

Publications (1)

Publication Number Publication Date
US20190169569A1 true US20190169569A1 (en) 2019-06-06

Family

ID=56936541

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/758,314 Abandoned US20190169569A1 (en) 2015-09-08 2016-09-07 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells
US16/931,003 Pending US20210139847A1 (en) 2015-09-08 2020-07-16 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/931,003 Pending US20210139847A1 (en) 2015-09-08 2020-07-16 Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells

Country Status (9)

Country Link
US (2) US20190169569A1 (fr)
EP (2) EP3347456B1 (fr)
JP (3) JP6983762B2 (fr)
KR (1) KR20180042437A (fr)
CN (2) CN108473962B (fr)
AU (2) AU2016321170B2 (fr)
CA (1) CA2997952A1 (fr)
DK (1) DK3347456T3 (fr)
WO (1) WO2017044483A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110709504A (zh) * 2017-06-05 2020-01-17 梅约医学教育与研究基金会 培养、增殖和分化干细胞的方法和材料
US10865381B2 (en) 2015-10-20 2020-12-15 FUJIFILM Cellular Dynamics, Inc. Multi-lineage hematopoietic precursor cell production by genetic programming
US11162070B2 (en) 2015-09-08 2021-11-02 FUJIFILM Cellular Dynamics, Inc. MACS-based purification of stem cell-derived retinal pigment epithelium
CN115873797A (zh) * 2021-09-29 2023-03-31 北京干细胞与再生医学研究院 一种视网膜色素上皮细胞的扩增培养基及培养方法
US11679180B2 (en) 2016-12-07 2023-06-20 Mayo Foundation For Medical Education And Research Methods and materials for using fibrin supports for retinal pigment epithelium transplantation

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3347456B1 (fr) * 2015-09-08 2023-12-27 The United States of America, as represented by The Secretary, Department of Health and Human Services Méthode de différenciation reproductible de cellules de l'épithélium pigmentaire rétinien de qualité clinique
US11717298B2 (en) 2016-11-09 2023-08-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tissue clamp and implantation method
US11458225B2 (en) 2016-11-09 2022-10-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services 3D vascularized human ocular tissue for cell therapy and drug discovery
PL236298B1 (pl) * 2017-09-21 2020-12-28 Univ Jagiellonski Sposób otrzymywania upigmentowanych komórek in vitro poprzez różnicowanie ludzkich indukowanych pluripotencjalnych komórek macierzystych
CN108531443A (zh) * 2018-04-04 2018-09-14 温州医科大学附属第二医院、温州医科大学附属育英儿童医院 小分子诱导多能性干细胞分化为视网膜色素上皮细胞的方法
WO2019204817A1 (fr) * 2018-04-20 2019-10-24 FUJIFILM Cellular Dynamics, Inc. Procédé pour la differentiation de cellules oculaires et son utilisation
SG11202104401RA (en) * 2018-11-19 2021-05-28 The United States Of America As Represented By The Secretary Biodegradable tissue replacement implant and its use
US20220233602A1 (en) * 2019-07-05 2022-07-28 Novo Nordisk A/S Generation of neural stem cell lines derived from human pluripotent stem cells
CA3156678A1 (fr) 2019-10-09 2021-04-15 Bluerock Therapeutics Lp Cellules a expression transgenique soutenue
KR102224115B1 (ko) 2020-01-10 2021-03-09 경북대학교병원 3차원 바이오프린팅 기술을 이용한 망막 세포 배양용 구조체 및 이의 활용
EP4314249A1 (fr) 2021-03-25 2024-02-07 Bluerock Therapeutics LP Procédés d'obtention de cellules souches pluripotentes induites
CN115261301A (zh) * 2021-04-30 2022-11-01 深圳华大生命科学研究院 一种视网膜色素上皮细胞体外诱导及培养方法
WO2022251499A1 (fr) * 2021-05-28 2022-12-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Procédés pour produire des cellules épithéliales pigmentaires rétiniennes maculaires, centrales et périphériques
WO2023196577A1 (fr) * 2022-04-08 2023-10-12 New York Stem Cell Foundation, Inc. Procédés de production d'ipscs

Family Cites Families (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG49267A1 (en) 1989-08-14 1998-05-18 Photogenesis Inc Surgical instrument and cell isolation and transplantation
US6045791A (en) 1992-03-06 2000-04-04 Photogenesis, Inc. Retinal pigment epithelium transplantation
US6416998B1 (en) 1992-09-02 2002-07-09 Baylor College Of Medicine Plasmid encoding a modified steroid hormone
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5763270A (en) 1995-06-07 1998-06-09 Genemedicine, Inc. Plasmid for delivery of nucleic acids to cells and methods of use
WO1998030679A1 (fr) 1997-01-10 1998-07-16 Life Technologies, Inc. Substitut de serum pour cellules souches embryonnaires
US6458589B1 (en) * 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
WO2001088104A2 (fr) 2000-05-17 2001-11-22 Geron Corporation Populations de progeniteurs neuronaux
US20030211603A1 (en) 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
US7989425B2 (en) 2002-09-27 2011-08-02 Genexine Inc. Vaccine enhancing the protective immunity to hepatitis c virus using plasmid DNA and recombinant adenovirus
US7682828B2 (en) 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
EP1781776A2 (fr) * 2004-07-29 2007-05-09 Stem Cell Innovations, Inc. Differenciation de cellules souches
US7442548B2 (en) 2004-09-08 2008-10-28 Wisconsin Alumni Research Foundation Culturing human embryonic stem cells in medium containing pipecholic acid and gamma amino butyric acid
EP2186823A1 (fr) 2005-11-14 2010-05-19 Merial Limited Thérapie génique pour traiter l'insuffisance rénale
MX2008007654A (es) 2005-12-13 2008-09-26 Univ Kyoto Factor de reprogramacion nuclear.
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
AU2008231020B2 (en) 2007-03-23 2013-09-05 Wisconsin Alumni Research Foundation Somatic cell reprogramming
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
CN101981446B (zh) 2008-02-08 2016-03-09 医疗探索公司 用于使用支持向量机分析流式细胞术数据的方法和系统
ES2587395T3 (es) 2008-06-04 2016-10-24 Cellular Dynamics International, Inc. Procedimientos para la producción de células IPS usando un enfoque no vírico
EP3450545B1 (fr) 2008-10-24 2023-08-23 Wisconsin Alumni Research Foundation Cellules souches pluripotentes obtenues par reprogrammation non virale
EP2548950B1 (fr) 2009-06-05 2017-10-25 Cellular Dynamics International, Inc. Reprogrammation de lymphocytes T et cellules hématopoïétiques
CA2770412C (fr) 2009-08-07 2018-09-11 Kyoto University Procede permettant d'etablir de facon efficace des cellules souches pluripotentes induites
US9359592B2 (en) * 2009-10-06 2016-06-07 Snu R&Db Foundation Method for differentiation into retinal cells from stem cells
US9005967B2 (en) 2010-01-22 2015-04-14 Kyoto University Myc variants improve induced pluripotent stem cell generation efficiency
US8278620B2 (en) 2010-05-03 2012-10-02 Thermo Finnigan Llc Methods for calibration of usable fragmentation energy in mass spectrometry
CN103003416B (zh) 2010-06-15 2016-11-16 细胞动力学国际有限公司 从小体积的外周血产生诱导性多潜能干细胞
JP5896360B2 (ja) * 2010-07-21 2016-04-13 国立大学法人京都大学 ヒト多能性幹細胞から中間中胚葉細胞への分化誘導方法
EP2702135B1 (fr) 2011-04-29 2019-04-17 University of Southern California Procédé de cryopréservation de cellules épithéliales pigmentaires de la rétine issues de cellules souches sur un substrat polymère
US10519422B2 (en) * 2012-02-29 2019-12-31 Riken Method of producing human retinal pigment epithelial cells
WO2013151186A1 (fr) * 2012-04-06 2013-10-10 国立大学法人京都大学 Méthode d'induction de cellules produisant de l'érythropoïétine
EP2951290B1 (fr) * 2013-02-01 2017-11-29 The United States of America, as represented by The Secretary, Department of Health and Human Services Procédé de génération de cellules épithéliales pigmentaires rétiniennes (epr) à partir de cellules souches pluripotentes induites (cspi)
CN105814192A (zh) * 2013-10-09 2016-07-27 加利福尼亚大学董事会 哺乳动物视网膜干细胞产生方法和应用
AU2014345110B2 (en) * 2013-11-11 2021-01-14 Riken Method for producing retinal pigment epithelial cells
US20150159134A1 (en) * 2013-12-11 2015-06-11 Pfizer Limited Method for producing retinal pigment epithelial cells
AU2014363032A1 (en) * 2013-12-11 2016-06-09 Pfizer Limited Method for producing retinal pigment epithelial cells
EP3347456B1 (fr) * 2015-09-08 2023-12-27 The United States of America, as represented by The Secretary, Department of Health and Human Services Méthode de différenciation reproductible de cellules de l'épithélium pigmentaire rétinien de qualité clinique

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11162070B2 (en) 2015-09-08 2021-11-02 FUJIFILM Cellular Dynamics, Inc. MACS-based purification of stem cell-derived retinal pigment epithelium
US10865381B2 (en) 2015-10-20 2020-12-15 FUJIFILM Cellular Dynamics, Inc. Multi-lineage hematopoietic precursor cell production by genetic programming
US11679180B2 (en) 2016-12-07 2023-06-20 Mayo Foundation For Medical Education And Research Methods and materials for using fibrin supports for retinal pigment epithelium transplantation
CN110709504A (zh) * 2017-06-05 2020-01-17 梅约医学教育与研究基金会 培养、增殖和分化干细胞的方法和材料
CN115873797A (zh) * 2021-09-29 2023-03-31 北京干细胞与再生医学研究院 一种视网膜色素上皮细胞的扩增培养基及培养方法

Also Published As

Publication number Publication date
US20210139847A1 (en) 2021-05-13
WO2017044483A1 (fr) 2017-03-16
AU2016321170A1 (en) 2018-04-12
EP3347456B1 (fr) 2023-12-27
DK3347456T3 (da) 2024-02-19
CN108473962A (zh) 2018-08-31
CA2997952A1 (fr) 2017-03-16
EP4345160A2 (fr) 2024-04-03
AU2022275469A1 (en) 2023-01-05
JP2021176335A (ja) 2021-11-11
KR20180042437A (ko) 2018-04-25
JP6983762B2 (ja) 2021-12-17
CN114807035A (zh) 2022-07-29
CN108473962B (zh) 2022-04-26
AU2016321170B2 (en) 2022-09-01
CN114807035B (zh) 2024-02-02
JP2018526014A (ja) 2018-09-13
EP3347456A1 (fr) 2018-07-18
JP2023162441A (ja) 2023-11-08

Similar Documents

Publication Publication Date Title
US20220033770A1 (en) Macs-based purification of stem cell-derived retinal pigment epithelium
US20210139847A1 (en) Method for reproducible differentiation of clinical-grade retinal pigment epithelium cells
US20210238546A1 (en) Method for differentiation of ocular cells and use thereof
US20230201267A1 (en) Retinal pigmented epithelium and photoreceptor dual cell aggregates and methods of use thereof
US20200248138A1 (en) Method for inducing differentiation of pluripotent stem cells into germline stem cell-like cells
WO2023196577A1 (fr) Procédés de production d'ipscs

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLULAR DYNAMICS INTERNATIONAL, INC., WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHASE, LUCAS;XUEZHU, FENG;SIGNING DATES FROM 20160923 TO 20161008;REEL/FRAME:045189/0193

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY TH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BHARTI, KAPIL;JHA, BALENDU SHEKHAR;SIGNING DATES FROM 20160928 TO 20161011;REEL/FRAME:045189/0873

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

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: FUJIFILM CELLULAR DYNAMICS, INC., WYOMING

Free format text: CHANGE OF NAME;ASSIGNOR:CELLULAR DYNAMICS INTERNATIONAL, INC.;REEL/FRAME:050826/0718

Effective date: 20180323

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

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

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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