US20110274662A1 - Methods of Producing RPE Cells and Compositions of RPE Cells - Google Patents

Methods of Producing RPE Cells and Compositions of RPE Cells Download PDF

Info

Publication number
US20110274662A1
US20110274662A1 US12/682,712 US68271208A US2011274662A1 US 20110274662 A1 US20110274662 A1 US 20110274662A1 US 68271208 A US68271208 A US 68271208A US 2011274662 A1 US2011274662 A1 US 2011274662A1
Authority
US
United States
Prior art keywords
cells
rpe
rpe cells
cell
human
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
US12/682,712
Other languages
English (en)
Inventor
Christopher Malcuit
Linda Lemieux
William Holmes
Pedro Huertas
Lucy Vilner
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.)
Astellas Institute for Regenerative Medicine
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US12/682,712 priority Critical patent/US20110274662A1/en
Assigned to ADVANCED CELL TECHNOLOGY, INC. reassignment ADVANCED CELL TECHNOLOGY, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEMIEUX, LINDA, HOLMES, WILLIAM, MALCUIT, CHRISTOPHER, VILNER, LUCY
Publication of US20110274662A1 publication Critical patent/US20110274662A1/en
Assigned to ADVANCED CELL TECHNOLOGY, INC. reassignment ADVANCED CELL TECHNOLOGY, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUERTAS, PEDRO
Assigned to CAMOFI MASTER LDC, CAMHZN MASTER LDC reassignment CAMOFI MASTER LDC SECURITY AGREEMENT Assignors: ADVANCED CELL TECHNOLOGY, INC., MYTOGEN, INC.
Assigned to ADVANCED CELL TECHNOLOGY, INC. reassignment ADVANCED CELL TECHNOLOGY, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOLMES, WILLIAM, MALCUIT, CHRISTOPHER, VILNER, LUCY, LEMIEUX, LINDA
Assigned to ADVANCED CELL TECHNOLOGY, INC., MYTOGEN, INC. reassignment ADVANCED CELL TECHNOLOGY, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: CAMHZN MASTER LDC, CAMOFI MASTER LDC
Assigned to ADVANCED CELL TECHNOLOGY, INC. reassignment ADVANCED CELL TECHNOLOGY, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUERTAS, PEDRO
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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • 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
    • 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
    • C12N2501/70Enzymes
    • C12N2501/73Hydrolases (EC 3.)
    • C12N2501/734Proteases (EC 3.4.)
    • 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
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin

Definitions

  • the retinal pigment epithelium is the pigmented cell layer just outside the neurosensory retina. This layer of cells nourishes retinal visual cells, and is attached to the underlying choroid (the layer of blood vessels behind the retina) and overlying retinal visual cells.
  • the RPE acts as a filter to determine what nutrients reach the retina from the choroid. Additionally, the RPE provides insulation between the retina and the choroid. Breakdown of the RPE interferes with the metabolism of the retina, causing thinning of the retina. Thinning of the retina can have serious consequences. For example, thinning of the retina may cause “dry” macular degeneration and may also lead to the inappropriate blood vessel formation that can cause “wet” macular degeneration).
  • RPE cells produced in vitro could be used to study the developments of the RPE, to identify factors that cause the RPE to breakdown, or to identify agents that can be used to stimulate repair of endogenous RPE cells. Additionally, RPE cells produced in vitro could themselves be used as a therapy for replacing or restoring all or a portion of a patient's damaged RPE cells. When used in this manner, RPE cells may provide an approach to treat macular degeneration, as well as other diseases and conditions caused, in whole or in part, by damage to the RPE.
  • RPE cells produced in vitro for screening or as a therapeutic relies on methods that can be used to produce large numbers of RPE cells in a systematic, directed manner. Such systematized differentiation methods would provide significant advantages over previous schemes based on, for example, spontaneous differentiation of RPE cells from transformed cell lines or other sources.
  • the present invention provides a method for differentiating RPE cells from human pluripotent stem cells, such as human embryonic stem cells and human induced pluripotent stem cells.
  • the method is used to produce large numbers of differentiated RPE cells for use in screening assays, to study the basic biology of the RPE, and as therapeutics.
  • RPE cells differentiated from pluripotent stem cells, such as human embryonic stem cells using this approach are molecularly distinct from human embryonic stem cells, as well as from adult and fetal-derived RPE cells.
  • the present invention also provides preparations and pharmaceutical preparations of RPE cells derived from human pluripotent stem cells.
  • RPE cell preparations are molecularly distinct from human embryonic stem cells, as well as from adult and fetal-derived RPE cells.
  • the present invention provides, for the first time, a detailed molecular characterization of RPE cells differentiated from human embryonic stem cells.
  • the detailed characterization includes comparisons to RPE cells derived from other sources (e.g., adult RPE cells and fetal RPE cells), as well as to human embryonic stem cells. This analysis not only provides a deeper understanding of RPE cells, but it also revealed that RPE cells differentiated from human embryonic stem cells have distinct molecular properties that distinguish these cells from previously described RPE cells.
  • the present invention provides preparations of RPE cells, including substantially purified preparations of RPE cells.
  • exemplary RPE cells are differentiated from human pluripotent stem cells, such as human embryonic stem cells or iPS cells.
  • Human pluripotent stem cell-derived RPE cells can be formulated and used to treat retinal degenerative diseases.
  • human pluripotent stem cell-derived RPE cells can be used in screening assays to identify agents that modulate RPE cell survival (in vitro and/or in vivo), to study RPE cell maturation, or to identify agents that modulate RPE cell maturation. Agents identified using such screening assays may be used in vitro or in vivo and may provide additional therapeutics that can be used alone or in combination with RPE cells to treat retinal degenerative diseases.
  • the present invention provides improved methods for the production of RPE cells from embryonic stem cells or other pluripotent stem cells.
  • the methods of the invention can be used to produce differentiated RPE cells.
  • the level of maturation, as assessed by pigmentation levels, of the differentiated RPE cells can be modulated so that differentiated RPE cells, mature RPE cells, or mixtures thereof are produced.
  • improved methods for the treatment of eye disorders involve the use of RPE cells derived from human embryonic stem cells to treat or ameliorate the symptoms of eye disorders, particularly eye disorders caused or exacerbated, in whole or in part, by damage to or breakdown of the endogenous RPE layer.
  • the invention provides a method for producing a culture of retinal pigment epithelial (RPE) cells.
  • the culture is a substantially purified culture containing at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater than 99% differentiated RPE cells (at least 75% of the culture is a differentiated RPE cell, regardless of level of maturity).
  • the substantially purified culture contains at least 30%, 35%, 40% or 45% mature differentiated RPE cells.
  • the substantially purified culture contains at least 50% mature differentiated RPE cells.
  • the substantially purified culture contains at least 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater than 99% mature differentiated RPE cells.
  • the differentiated RPE cells are derived from human embryonic stem cells, human iPS cells, or other pluripotent stem cells.
  • the method comprising the steps of
  • the invention provides a method of producing a mature retinal pigment epithelial (RPE) cell, said method comprising the steps of
  • step (c) culturing the adherent culture of cells of step (c) in nutrient rich, low protein medium, which medium does not contain serum free B-27 supplement;
  • the substantially purified culture of RPE cells may contain both differentiated RPE cells and mature differentiated RPE cells.
  • the level of pigment may vary.
  • the mature RPE cells can be distinguished visually from the RPE cells based on the increased level of pigmentation and the more columnar shape.
  • the percentage of mature differentiated RPE cells in the culture can be reduced by decreasing the density of the culture.
  • the method further comprises subculturing a population of mature RPE cells to produce a culture containing a smaller percentage of mature RPE cells.
  • the medium used when culturing the cells as embryoid bodies may be selected from any medium appropriate for culturing cells as embryoid bodies.
  • any medium that is capable of supporting high-density cultures may be used, such as medium for viral, bacterial, or eukaryotic cell culture.
  • the medium may be high nutrient, protein-free medium or high nutrient, low protein medium.
  • the human embryonic stem cells may be cultured in MDBK-GM, OptiPro SFM, VP-SFM, EGM-2, or MDBK-MM.
  • the medium may also contain B-27 supplement.
  • the medium described herein may also be supplemented with one or more growth factors.
  • Growth factors that may be used include, for example, EGF, bFGF, VEGF, and recombinant insulin-like growth factor.
  • the medium may also contain supplements such as heparin, hydrocortisone, ascorbic acid, serum (such as, for example, fetal bovine serum), or a growth matrix (such as, for example, extracellular matrix from bovine corneal epithelium, matrigel (BD biosciences), or gelatin).
  • mechanical or enzymatic methods are used to select RPE cells from amongst clusters of non-RPE cells in a culture of embryoid body, or to facilitate sub-culture of adherent cells.
  • Exemplary mechanical methods include, but are not limited to, tituration with a pipette or cutting with a pulled needle.
  • Exemplary enzymatic methods include, but are not limited to, any enzymes appropriate for disassociating cells (e.g., trypsin, collagenase, dispase).
  • a non-enzymatic solution is used to disassociate the cells, such as a high EDTA-containing solution such as, for example, Hanks-based cell disassociation buffer.
  • the cells are cultured for between about 3 days and 45 days, such as 7 days, 7-10 days, 7-14 days, or 14-21 days.
  • the cells are cultured for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, or about 46 days.
  • the cells are cultured for less than or equal to about: 45, 40, 35, 30, 25, 21, 20, 18, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 days.
  • the cells may be cultured for the same period of time at each step or for differing periods of time at one or more of the steps.
  • the RPE cells are further cultured to produce a culture of mature RPE cells.
  • Both RPE cells and mature RPE cells are differentiated RPE cells.
  • mature RPE cells are characterized by increased level of pigment in comparison to differentiated RPE cells. The level of maturity and pigmentation can be modulated by increasing or decreasing the density of the culture of differentiated RPE cells.
  • a culture of RPE cells can be further cultured to produce mature RPE cells.
  • the density of a culture containing mature RPE cells can be decreased to decrease the percentage of mature differentiated RPE cells and increase the percentage of differentiated RPE cells.
  • the medium used to culture the RPE cells is any medium appropriate for cell culture, and can be selected by the skilled person.
  • any medium that is capable of supporting high-density cultures may be used, such as medium for viral, bacterial, or animal cell culture.
  • the cells described herein may be cultured in VP-SFM, EGM-2, and MDBK-MM.
  • said substantially purified culture of RPE cells are frozen for storage.
  • the cells may be stored by any appropriate method known in the art, e.g., cryogenically frozen and may be frozen at any temperature appropriate for storage of the cells.
  • the cells may be frozen at approximately ⁇ 20° C., ⁇ 80° C., ⁇ 120° C., or at any other temperature appropriate for storage of cells.
  • Cryogenically frozen cells are stored in appropriate containers and prepared for storage to reduce risk of cell damage and maximize the likelihood that the cells will survive thawing.
  • RPE cells are maintained at room temperature, or refrigerated at, for example, approximately 4° C.
  • the method is performed in accordance with Good Manufacturing Practices (GMP).
  • GMP Good Manufacturing Practices
  • the human embryonic stem cells from which the RPE cells are differentiated were derived in accordance with Good Manufacturing Practices (GMP).
  • the human embryonic stem cells from which the RPE cells are differentiated were derived from one or more blastomeres removed from an early stage embryo without destroying the remaining embryo.
  • the method is used to produce a preparation comprising at least 1 ⁇ 10 5 RPE cells, at least 5 ⁇ 10 5 RPE cells, at least 1 ⁇ 10 6 RPE cells, at least 5 ⁇ 10 6 RPE cells, at least 1 ⁇ 10 7 RPE cells, at least 2 ⁇ 10 7 RPE cells, at least 3 ⁇ 10 7 RPE cells, at least 4 ⁇ 10 7 RPE cells, at least 5 ⁇ 10 7 RPE cells, at least 6 ⁇ 10 7 RPE cells, at least 7 ⁇ 10 7 RPE cells, at least 8 ⁇ 10 7 RPE cells, at least 9 ⁇ 10 7 RPE cells, at least 1 ⁇ 10 8 RPE cells, at least 2 ⁇ 10 8 RPE cells, at least 5 ⁇ 10 8 RPE cells, at least 7 ⁇ 10 8 RPE cells, or at least 1 ⁇ 10 9 RPE cells.
  • the number of RPE cells in the preparation includes differentiated RPE cells, regardless of level of maturity and regardless of the relative percentages of differentiated RPE cells and mature RPE cells. In other embodiments, the number of RPE cells in the preparation refers to the number of either differentiated RPE cells or mature RPE cells.
  • the method further comprises formulating the differentiated RPE cells and/or differentiated mature RPE cells to produce a preparation of RPE cells suitable for transplantation.
  • the invention provides a method for treating or preventing a condition characterized by retinal degeneration, comprising administering to a subject in need thereof an effective amount of a preparation comprising RPE cells, which RPE cells are derived from human embryonic stem cells, iPS cells, or other pluripotent stem cells.
  • RPE cells which RPE cells are derived from human embryonic stem cells, iPS cells, or other pluripotent stem cells.
  • Conditions characterized by retinal degeneration include, for example, Stargardt's macular dystrophy, age related macular degeneration (dry or wet), diabetic retinopathy, and retinitis pigmentosa.
  • the RPE cells are derived from human pluripotent stem cells using one or more of the methods described herein.
  • the preparation was previously cryopreserved and was thawed before transplantation.
  • the method of treating further comprises administration of cyclosporin or one or more other immunosuppressants.
  • immunosuppressants When immunosuppressants are used, they may be administered systemically or locally, and they may be administered prior to, concomitantly with, or following administration of the RPE cells. In certain embodiments, immunosuppressive therapy continues for weeks, months, years, or indefinitely following administration of RPE cells.
  • the method of treatment comprises administration of a single dose of RPE cells.
  • the method of treatment comprises a course of therapy where RPE cells are administered multiple times over some period.
  • Exemplary courses of treatment may comprise weekly, biweekly, monthly, quarterly, biannually, or yearly treatments.
  • treatment may proceed in phases whereby multiple doses are required initially (e.g., daily doses for the first week), and subsequently fewer and less frequent doses are needed. Numerous treatment regimens are contemplated.
  • the administered RPE cells comprise a mixed population of differentiated RPE cells and mature RPE cells.
  • the administered RPE cells comprise a substantially purified population of either differentiated RPE cells or mature RPE cells.
  • the administered RPE cells may contain less than 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% of the other RPE cell-type.
  • the RPE cells are formulated in a pharmaceutically acceptable carrier or excipient.
  • the preparation comprising RPE cells is transplanted in a suspension, matrix or substrate.
  • the preparation is administered by injection into the subretinal space of the eye.
  • about 10 4 to about 10 6 cells are administered to the subject.
  • the method further comprises the step of monitoring the efficacy of treatment or prevention by measuring electroretinogram responses, optomotor acuity threshold, or luminance threshold in the subject.
  • the method may also comprise monitoring the efficacy of treatment or prevention by monitoring immunogenicity of the cells or migration of the cells in the eye.
  • the invention provides a pharmaceutical preparation for treating or preventing a condition characterized by retinal degeneration, comprising an effective amount of RPE cells, which RPE cells are derived from human embryonic stem cells or other pluripotent stem cells.
  • the pharmaceutical preparation may be formulated in a pharmaceutically acceptable carrier according to the route of administration.
  • the preparation may be formulated for administration to the subretinal space of the eye.
  • the composition may comprise at least 10 4 , 10 5 , 5 ⁇ 10 5 , 6 ⁇ 10 5 , 7 ⁇ 10 5 , 8 ⁇ 10 5 , 9 ⁇ 10 5 , 10 6 , 2 ⁇ 10 6 , 3 ⁇ 10 6 , 4 ⁇ 10 6 , 5 ⁇ 10 6 , 6 ⁇ 10 6 , 7 ⁇ 10 6 , 8 ⁇ 10 6 , 9 ⁇ 10 6 , or 10 7 RPE cells.
  • the composition may comprise at least 2 ⁇ 10 7 , 5 ⁇ 10 7 , 6 ⁇ 10 7 , 7 ⁇ 10 7 , 8 ⁇ 10 7 , 9 ⁇ 10 7 , 1 ⁇ 10 8 RPE cells.
  • the RPE cells may include mature RPE cells, and thus the cell number includes the total of both differentiated RPE cells and mature differentiated RPE cells.
  • the invention provides a method for screening to identify agents that modulate the survival of RPE cells.
  • RPE cells differentiated from human embryonic stem cells can be used to screen for agents that promote RPE survival.
  • Identified agents can be used, alone or in combination with RPE cells, as part of a treatment regimen.
  • identified agents can be used as part of a culture method to improve the survival of RPE cells differentiated in vitro.
  • the invention provides a method for screening to identify agents that modulate RPE cell maturity.
  • RPE cells differentiated from human ES cells can be used to screen for agents that promote RPE maturation.
  • the method is performed in accordance with Good Manufacturing Practices (GMP).
  • GMP Good Manufacturing Practices
  • the human embryonic stem cells or other pluripotent stem cells from which the RPE cells are differentiated were derived in accordance with Good Manufacturing Practices (GMP).
  • the human embryonic stem cells from which the RPE cells are differentiated were derived from one or more blastomere removed from an early stage embryo without destroying the remaining embryo.
  • the invention contemplates that, instead of human embryonic stem cells, the starting material for producing RPE cells, or preparations thereof, can be other types of human pluripotent stem cells.
  • the invention contemplates that induced pluripotent stem (iPS) cells are used as a starting material for differentiating RPE cells using the methods described herein.
  • iPS cells can be obtained from a cell bank, or otherwise previously derived.
  • iPS cells can be newly generated prior to commencing differentiation to RPE cells.
  • RPE cells or preparations differentiated from pluripotent stem cells, including iPS cells are used in a therapeutic method.
  • the present invention also provides functional human retinal pigmented epithelial cells (hRPEs) that are terminally differentiated from human embryonic stem cells (hESCs) or other human pluripotent stem cells.
  • hRPEs human retinal pigmented epithelial cells
  • hESCs human embryonic stem cells
  • hRPEs may treat retinal degeneration in the diseased animal.
  • the hRPEs of the invention are useful for treating patients afflicted by various retinal degenerative disorders.
  • the present invention therefore provides a renewable source of hRPEs that can be produced and manufactured under GLP-like and GMP-compliant conditions for the treatment of visual degenerative diseases and disorders.
  • the present invention provides a human retinal pigmented epithelial cell derived from a human embryonic stem cell, which cell is pigmented and expresses at least one gene that is not expressed in a cell that is not a human retinal pigmented epithelial cell.
  • the human retinal pigmented epithelial cell is isolated from at least one protein, molecule, or other impurity that is found in its natural environment.
  • the invention provides a cell culture comprising human RPE cells derived from human embryonic stem cells or other pluripotent stem cells, which are pigmented and express at least one gene that is not expressed in a cell that is not a human RPE.
  • pigmented refers to any level of pigmentation, for example, the pigmentation that initial occurs when RPE cells differentiate from ES cells. Pigmentation may vary with cell density and the maturity of the differentiated RPE cells. However, when cells are referred to as pigmented—the term is understood to refer to any and all levels of pigmentation.
  • the cell culture comprises a substantially purified population of human RPE cells.
  • a substantially purified population of hRPE cells is one in which the hRPE cells comprise at least about 75% of the cells in the population.
  • a substantially purified population of hRPE cells is one in which the hRPE cells comprise at least about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 97.5%, 98%, 99%, or even greater than 99% of the cells in the population.
  • the pigmentation of the hRPE cells in the cell culture is homogeneous.
  • the pigmentation of the hRPE cells in the cell culture is heterogeneous, and the culture of RPE cells comprises both differentiated RPE cells and mature RPE cells.
  • a cell culture of the invention may comprise at least about 10 1 , 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , or at least about 10 9 hRPE cells.
  • the present invention provides human retinal pigmented epithelial cells with varying degrees of pigmentation.
  • the pigmentation of a human retinal pigmented epithelial cell is the same as an average human pigmented epithelial cell after terminal differentiation of the hRPE cell.
  • the pigmentation of a human retinal pigmented epithelial cell is more pigmented than the average human retinal pigmented epithelial cell after terminal differentiation of the hRPE cell.
  • the pigmentation of a human retinal pigmented epithelial cell is less pigmented than the average human retinal pigmented epithelial cell after terminal differentiation.
  • the present invention provides human RPE cells differentiated from ES cells or other pluripotent stem cells and that express (at the mRNA and/or protein level) one or more (1, 2, 3, 4, 5, or 6) of the following: RPE-65, Bestrophin, PEDF, CRALBP, Otx2, and Mit-F.
  • gene expression is measured by mRNA expression.
  • gene expression is measured by protein expression.
  • the RPE cells do not substantially express ES-specific genes, such as Oct-4, alkaline phosphatase, nanog, and/or Rex-1.
  • the RPE cells express one or more (1, 2, or 3) of pax-2, pax-6, and/or tyrosinase. In certain embodiments, expression of pax-2, pax-6, and/or tyrosinase distinguishes differentiated RPE cells from mature differentiated RPE cells. In other embodiments, the RPE cells express one or more of the markers presented in Table 2, and the expression of the one or more markers is upregulated in RPE cells relative to expression (if any) in human ES cells. In other embodiments, the RPE cells express one or more of the markers presented in Table 3, and the expression of the one or more markers is downregulated in RPE cells relative to expression (if any) in human ES cells.
  • the invention provides a pharmaceutical preparation comprising human RPE cells derived from human embryonic stem cells or other pluripotent stem cells.
  • Pharmaceutical preparations may comprise at least about 10 1 , 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 or about 10 9 hRPE cells.
  • the invention provides a cryopreserved preparation of the RPE cells described herein.
  • the cryopreserved preparation may be frozen for storage for days or years.
  • the cells may be stored by any appropriate method known in the art, e.g., cryogenically frozen and may be frozen at any temperature appropriate for storage of the cells.
  • the cells may be frozen at approximately ⁇ 20° C., ⁇ 80° C., ⁇ 120° C., or at any other temperature appropriate for storage of cells.
  • Cryogenically frozen cells are stored in appropriate containers and prepared for storage to reduce risk of cell damage and maximize the likelihood that the cells will survive thawing.
  • RPE cells can be maintained at room temperature, or refrigerated at, for example, approximately 4° C.
  • Cryopreserved preparations of the compositions described herein may comprise at least about 10 1 , 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 or about 10 9 hRPE cells.
  • the hRPE cells of the invention are recovered from storage following cryopreservation.
  • greater than 65%, 70%, 75,%, or greater than 80% of the RPE cells retain viability following cryopreservation.
  • the invention provides substantially purified preparations of human RPE cells have any combination of the structural, molecular, and functional characteristics described herein.
  • Such preparations may be formulated as pharmaceutical preparations for administration and/or may be formulated for cryopreservation.
  • the invention provides use of the human RPE cells described herein in the manufacture of a medicament to treat a condition in a patient in need thereof.
  • the invention provides use of a cell culture comprising the human RPE cells described herein in the manufacture of a medicament to treat a condition in a patient in need thereof.
  • the invention provides the use of a pharmaceutical preparation comprising the human RPE cells described herein in the manufacture of a medicament to treat a condition in a patient in need thereof.
  • Conditions that may be treated include, without limitation, degenerative diseases of the retina, such as Stargardt's macular dystrophy, retinitis pigmentosa, macular degeneration, glaucoma, and diabetic retinopathy.
  • the invention provides methods for treating or preventing a condition characterized by retinal degeneration, comprising administering to a subject in need thereof an effective amount of a preparation comprising RPE cells, which RPE cells are derived from mammalian embryonic stem cells.
  • Conditions characterized by retinal degeneration include, for example, Stargardt's macular dystrophy, age related macular degeneration, diabetic retinopathy, and retinitis pigmentosa.
  • the invention provides a solution of human RPE cells derived from a human embryonic stem cell, or other pluripotent stem cell, which RPE cells have any combinations of the features described herein.
  • a solutions may comprise at least about 10 1 , 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 or about 10 9 hRPE cells as described herein.
  • Such solutions are suitable for injection to a subject.
  • Such solutions are suitable for cryopreservation as described herein.
  • This invention also provides a use of these solutions for the manufacture of a medicament to treat a disease that could be treated by the administration of RPE cells, such as, for example, retinal degenerative diseases of the eye.
  • the RPE cells of the invention are derived from human embryonic stem cells, or other pluripotent stem cells, previously derived under GMP conditions.
  • the human ES cells are derived from one or more blastomeres of an early cleavage stage embryo, optionally without destroying the embryo.
  • the human ES cells are from a library of human embryonic stem cells.
  • said library of human embryonic stem cells comprises stem cells, each of which is hemizygous, homozygous, or nullizygous for at least one MHC allele present in a human population, wherein each member of said library of stem cells is hemizygous, homozygous, or nullizygous for a different set of MHC alleles relative to the remaining members of the library.
  • the library of human embryonic stem cells comprises stem cells that are hemizygous, homozygous, or nullizygous for all MHC alleles present in a human population.
  • the invention provides a library of RPE cells, each of which is hemizygous, homozygous, or nullizygous for at least one MHC allele present in a human population, wherein each member of said library of RPE cells is hemizygous, homozygous, or nullizygous for a different set of MHC alleles relative to the remaining members of the library.
  • invention provides a library of human RPE cells that are hemizygous, homozygous, or nullizygous for all MHC alleles present in a human population.
  • said substantially purified culture of RPE cells are frozen for storage.
  • the cells may be stored by any appropriate method known in the art, e.g., cryogenically frozen and may be frozen at any temperature appropriate for storage of the cells.
  • the cells may be frozen at approximately ⁇ 20° C., ⁇ 80° C., ⁇ 120° C., or at any other temperature appropriate for storage of cells.
  • Cryogenically frozen cells are stored in appropriate containers and prepared for storage to reduce risk of cell damage and maximize the likelihood that the cells will survive thawing.
  • RPE cells can be maintained at room temperature, or refrigerated at, for example, approximately 4° C.
  • human RPE cells are produced in accordance with Good Manufacturing Practices (GMP).
  • the human embryonic stem cells from which the RPE cells are differentiated were derived in accordance with Good Manufacturing Practices (GMP).
  • the human embryonic stem cells from which the RPE cells are differentiated were derived from one or more blastomeres removed from an early stage embryo without destroying the remaining embryo.
  • the invention provides GMP compliant preparations of RPE cells, including substantially purified preparations of RPE cells. Such preparations are substantially free of viral, bacterial, and/or fungal contamination or infection.
  • compositions or preparations of RPE cells comprise at least 1 ⁇ 10 5 RPE cells, at least 5 ⁇ 10 5 RPE cells, at least 1 ⁇ 10 6 RPE cells, at least 5 ⁇ 10 6 RPE cells, at least 1 ⁇ 10 7 RPE cells, at least 2 ⁇ 10 7 RPE cells, at least 3 ⁇ 10 7 RPE cells, at least 4 ⁇ 10 7 RPE cells, at least 5 ⁇ 10 7 RPE cells, at least 6 ⁇ 10 7 RPE cells, at least 7 ⁇ 10 7 RPE cells, at least 8 ⁇ 10 7 RPE cells, at least 9 ⁇ 10 7 RPE cells, at least 1 ⁇ 10 8 RPE cells, at least 2 ⁇ 10 8 RPE cells, at least 5 ⁇ 10 8 RPE cells, at least 7 ⁇ 10 8 RPE cells, or at least 1 ⁇ 10 9 RPE cells.
  • the number of RPE cells in the preparation includes differentiated RPE cells, regardless of level of maturity and regardless of the relative percentages of differentiated RPE cells and mature differentiated RPE cells. In other embodiments, the number of RPE cells in the preparation refers to the number of either differentiated RPE cells or mature RPE cells.
  • the method further comprises formulating the differentiated RPE cells and/or differentiated mature RPE cells to produce a preparation of RPE cells suitable for transplantation.
  • the invention provides a method for treating or preventing a condition characterized by retinal degeneration, comprising administering to a subject in need thereof an effective amount of a preparation comprising RPE cells, which RPE cells are derived from human pluripotent stem cells.
  • RPE cells are derived using any of the methods described herein.
  • Conditions characterized by retinal degeneration include, for example, Stargardt's macular dystrophy, age related macular degeneration (dry or wet), diabetic retinopathy, and retinitis pigmentosa.
  • the preparation was previously cryopreserved and was thawed before transplantation.
  • the method of treating further comprises administration of cyclosporin or one or more other immunosuppressants.
  • immunosuppressants When immunosuppressants are used, they may be administered systemically or locally, and they may be administered prior to, concomitantly with, or following administration of the RPE cells. In certain embodiments, immunosuppressive therapy continues for weeks, months, years, or indefinitely following administration of RPE cells.
  • the method of treatment comprises administration of a single dose of RPE cells.
  • the method of treatment comprises a course of therapy where RPE cells are administered multiple times over some period.
  • Exemplary courses of treatment may comprise weekly, biweekly, monthly, quarterly, biannually, or yearly treatments.
  • treatment may proceed in phases whereby multiple doses are required initially (e.g., daily doses for the first week), and subsequently fewer and less frequent doses are needed. Numerous treatment regimens are contemplated.
  • the administered RPE cells comprise a mixed population of differentiated RPE cells and mature RPE cells.
  • the administered RPE cells comprise a substantially purified population of either differentiated RPE cells or mature RPE cells.
  • the administered RPE cells may contain less than 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% of the other RPE cell-type.
  • the RPE cells are formulated in a pharmaceutically acceptable carrier or excipient.
  • the preparation comprising RPE cells is transplanted in a suspension, matrix or substrate.
  • the preparation is administered by injection into the subretinal space of the eye.
  • the preparation is administered transcorneally.
  • about 10 4 to about 10 6 cells are administered to the subject.
  • the method further comprises the step of monitoring the efficacy of treatment or prevention by measuring electroretinogram responses, optomotor acuity threshold, or luminance threshold in the subject.
  • the method may also comprise monitoring the efficacy of treatment or prevention by monitoring immunogenicity of the cells or migration of the cells in the eye.
  • the invention provides a pharmaceutical preparation for treating or preventing a condition characterized by retinal degeneration, comprising an effective amount of RPE cells, which RPE cells are derived from human embryonic stem cells.
  • the pharmaceutical preparation may be formulated in a pharmaceutically acceptable carrier according to the route of administration.
  • the preparation may be formulated for administration to the subretinal space or cornea of the eye.
  • the composition may comprise at least 10 4 , 10 5 , 5 ⁇ 10 5 , 6 ⁇ 10 5 , 7 ⁇ 10 5 , 8 ⁇ 10 5 , 9 ⁇ 10 5 , 10 6 , 2 ⁇ 10 6 , 3 ⁇ 10 6 , 4 ⁇ 10 6 , 5 ⁇ 10 6 , 6 ⁇ 10 6 , 7 ⁇ 10 6 , 8 ⁇ 10 6 , 9 ⁇ 10 6 , or 10 7 RPE cells.
  • the composition may comprise at least 2 ⁇ 10 7 , 5 ⁇ 10 7 , 6 ⁇ 10 7 , 7 ⁇ 10 7 , 8 ⁇ 10 7 , 9 ⁇ 10 7 , 1 ⁇ 10 8 RPE cells.
  • the RPE cells may include mature RPE cells, and thus the cell number includes the total of both differentiated RPE cells and mature differentiated RPE cells.
  • the invention provides a method for screening to identify agents that modulate the survival of RPE cells.
  • RPE cells differentiated from human embryonic stem cells can be used to screen for agents that promote RPE survival.
  • Identified agents can be used, alone or in combination with RPE cells, as part of a treatment regimen.
  • identified agents can be used as part of a culture method to improve the survival of RPE cells differentiated in vitro.
  • the invention provides a method for screening to identify agents that modulate RPE cell maturity.
  • RPE cells differentiated from human ES cells can be used to screen for agents that promote RPE maturation.
  • the method is performed in accordance with Good Manufacturing Practices (GMP).
  • GMP Good Manufacturing Practices
  • the human embryonic stem cells from which the RPE cells are differentiated were derived in accordance with Good Manufacturing Practices (GMP).
  • the human embryonic stem cells from which the RPE cells are differentiated were derived from one or more blastomere removed from an early stage embryo without destroying the remaining embryo.
  • the invention contemplates that, instead of human embryonic stem cells, the starting material for producing RPE cells, or preparations thereof, can be other types of human pluripotent stem cells.
  • the invention contemplates that induced pluripotent stem (iPS) cells, which have the characteristic of hES, can similarly be used as a starting material for differentiating RPE cells using the methods described herein.
  • iPS cells can be obtained from a cell bank, or otherwise previously derived.
  • iPS cells can be newly generated prior to commencing differentiation to RPE cells.
  • RPE cells or preparations differentiated from pluripotent stem cells, including iPS cells are used in a therapeutic method.
  • preparations of RPE cells comprising any combination of differentiated RPE cells and mature RPE cells can be used in the treatment of any of the diseases and conditions described herein.
  • methods described herein for producing RPE cells using human embryonic stem cells as a starting material may be similarly performed using any human pluripotent stem cells as a starting material.
  • FIG. 1 is a schematic model showing the developmental ontogeny of human RPE cells derived from human embryonic stem cells.
  • FIG. 2 is a graph showing gene expression comparison of hES cells and human embryonic stem cell-derived RPE cells by quantitative, Real-Time, Reverse Transcription PCR (qPCR).
  • FIG. 3 is a graph showing gene expression comparison of ARPE-19 cells (a previously derived RPE cell line) and human embryonic stem cell-derived RPE cells by quantitative, Real-Time, Reverse Transcription PCR (qPCR).
  • FIG. 4 is a graph showing gene expression comparison of fetal RPE cells and human embryonic stem cell-derived RPE cells by quantitative, Real-Time, Reverse Transcription PCR (qPCR).
  • FIG. 5 is a graph showing gene expression comparison of mature RPE cells and hES cells by quantitative, Real-Time, Reverse Transcription PCR (qPCR).
  • FIG. 6 is a photomicrograph showing Western Blot analysis of hES-specific and RPE-specific markers.
  • Embryonic stem cell-derived RPE cells (lane 1) derived from hES cells (lane 2) did not express the hES-specific proteins Oct-4, Nanog, and Rex-1.
  • embryonic stem cell-derived RPE cells express RPE-specific proteins included RPE65, CRALBP, PEDF, Bestrophin, PAX6, and Otx2. Actin was used as protein loading control.
  • FIG. 7 is a graph showing the principal components analysis plot of microarray gene expressions.
  • Component 1 representing 69% of the variability represents the cell type, whereas Component 2, represents the cell line (i.e., genetic variability).
  • a near-linear scatter of gene expression profiles characterizes the developmental ontogeny of hRPE derived from hES cells.
  • embryo or “embryonic” is meant a developing cell mass that has not implanted into the uterine membrane of a maternal host.
  • An “embryonic cell” is a cell isolated from or contained in an embryo. This also includes blastomeres, obtained as early as the two-cell stage, and aggregated blastomeres.
  • embryo-derived stem cells refers to embryo-derived cells. More specifically it refers to cells isolated from the inner cell mass of blastocysts or morulae and that have been serially passaged as cell lines. The term also includes cells isolated from one or more blastomeres of an embryo, preferably without destroying the remainder of the embryo. The term also includes cells produced by somatic cell nuclear transfer, even when non-embryonic cells are used in the process.
  • hES cells human embryonic stem cells
  • This term includes cells derived from the inner cell mass of human blastocysts or morulae that have been serially passaged as cell lines.
  • the hES cells may be derived from fertilization of an egg cell with sperm or DNA, nuclear transfer, parthenogenesis, or by means to generate hES cells with homozygosity in the HLA region.
  • Human ES cells are also cells derived from a zygote, blastomeres, or blastocyst-staged mammalian embryo produced by the fusion of a sperm and egg cell, nuclear transfer, parthenogenesis, or the reprogramming of chromatin and subsequent incorporation of the reprogrammed chromatin into a plasma membrane to produce a cell.
  • Human embryonic stem cells of the present invention may include, but are not limited to, MAO1, MAO9, ACT-4, No. 3, H1, H7, H9, H14 and ACT30 embryonic stem cells.
  • human ES cells used to produce RPE cells are derived and maintained in accordance with GMP standards.
  • Human embryonic stem cells regardless of their source or the particular method use to produce them, can be identified based on (i) the ability to differentiate into cells of all three germ layers, (ii) expression of at least Oct-4 and alkaline phosphatase, and (iii) ability to produce teratomas when transplanted into immunocompromised animals.
  • human embryo-derived cells refers to morula-derived cells, blastocyst-derived cells including those of the inner cell mass, embryonic shield, or epiblast, or other totipotent or pluripotent stem cells of the early embryo, including primitive endoderm, ectoderm, and mesoderm and their derivatives, also including blastomeres and cell masses from aggregated single blastomeres or embryos from varying stages of development, but excluding human embryonic stem cells that have been passaged as cell lines.
  • pluripotent stem cells includes embryonic stem cells, embryo-derived stem cells, and induced pluripotent stem cells, regardless of the method by which the pluripotent stem cells are derived.
  • Pluripotent stem cells are defined functionally as stem cells that: (a) are capable of inducing teratomas when transplanted in immunodeficient (SCID) mice; (b) are capable of differentiating to cell types of all three germ layers (e.g., can differentiate to ectodermal, mesodermal, and endodermal cell types); and (c) express one or more markers of embryonic stem cells (e.g., express Oct 4, alkaline phosphatase, SSEA-3 surface antigen, SSEA-4 surface antigen, nanog, TRA-1-60, TRA-1-81, SOX2, REX1, etc).
  • Exemplary pluripotent stem cells can be generated using, for example, methods known in the art.
  • Exemplary pluripotent stem cells include embryonic stem cells derived from the ICM of blastocyst stage embryos, as well as embryonic stem cells derived from one or more blastomeres of a cleavage stage or morula stage embryo (optionally without destroying the remainder of the embryo). Such embryonic stem cells can be generated from embryonic material produced by fertilization or by asexual means, including somatic cell nuclear transfer (SCNT), parthenogenesis, and androgenesis. Further exemplary pluripotent stem cells include induced pluripotent stem cells (iPS cells) generated by reprogramming a somatic cell by expressing or inducing expression of a combination of factors (herein referred to as reprogramming factors).
  • iPS cells induced pluripotent stem cells
  • iPS cells 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, a combination of Oct4 (sometimes referred to as Oct 3/4), Sox2, c-Myc, and Klf4.
  • factors that can be used to reprogram somatic cells to pluripotent stem cells include, for example, a combination of Oct 4, Sox2, Nanog, and Lin28.
  • somatic cells are reprogrammed by expressing at least 2 reprogramming factors, at least three reprogramming factors, or four reprogramming factors.
  • additional reprogramming factors are identified and used alone or in combination with one or more known reprogramming factors to reprogram a somatic cell to a pluripotent stem cell.
  • RPE cell and “differentiated RPE cell” and “ES-derived RPE cell” and “human RPE cell” are used interchangeably throughout to refer to an RPE cell differentiated from a human embryonic stem cell using a method of the invention.
  • the term is used generically to refer to differentiated RPE cells, regardless of the level of maturity of the cells, and thus may encompass RPE cells of various levels of maturity.
  • Differentiated RPE cells can be visually recognized by their cobblestone morphology and the initial appearance of pigment. Differentiated RPE cells can also be identified molecularly based on substantial lack of expression of embryonic stem cell markers such as Oct-4 and nanog, as well as based on the expression of RPE markers such as RPE-65, PEDF, CRALBP, and bestrophin.
  • RPE-like cells are generally referred to specifically as adult, fetal or APRE19 cells.
  • RPE cells refers to RPE cells differentiated from human embryonic stem cells.
  • mature RPE cell and “mature differentiated RPE cell” are used interchangeably throughout to refer to changes that occur following initial differentiating of RPE cells.
  • RPE cells can be recognized, in part, based on initial appearance of pigment
  • mature RPE cells can be recognized based on enhanced pigmentation.
  • Pigmentation post-differentiation is not indicative of a change in the RPE state of the cells (e.g., the cells are still differentiated RPE cells). Rather, the changes in pigment post-differentiation correspond to the density at which the RPE cells are cultured and maintained.
  • mature RPE cells have increased pigmentation that accumulates after initial differentiation. Mature RPE cells are more pigmented than RPE cells—although RPE cells do have some level of pigmentation.
  • Mature RPE cells can be subcultured at a lower density, such that the pigmentation decreases.
  • mature RPE cells can be cultured to produce RPE cells.
  • Such RPE cells are still differentiated RPE cells that express markers of RPE differentiation.
  • pigmentation changes post-differentiation are phenomenological and do not reflect dedifferentiation of the cells away from an RPE fate.
  • changes in pigmentation post-differentiation also correlate with changes in pax-2 expression.
  • RPE-like cells when other RPE-like cells are referred to, they are generally referred to specifically as adult, fetal or APRE19 cells.
  • RPE cells refers to RPE cells differentiated from human embryonic stem cells.
  • APRE-19 refers to cells of a previously derived, human RPE cell line. APRE-19 cells arose spontaneously and are not derived from human embryos or embryonic stem cells.
  • This invention provides preparations and compositions comprising human retinal pigmented epithelium (RPE) cells derived from human embryonic stem cells or other human pluripotent stem cells.
  • the RPE cells are pigmented, to at least some extent.
  • the RPE cells do not express (at any appreciable level) the embryonic stem cell markers Oct-4, nanog, or Rex-1. Specifically, expression of these ES genes is approximately 100-1000 fold lower in RPE cells than in ES cells, when assessed by quantitative RT-PCR.
  • the RPE cells do express, both at the mRNA and protein level, one or more of the following: RPE65, CRALBP, PEDF, Bestrophin, MitF and/or Otx2.
  • the RPE cells express, both at the mRNA and protein level, one or more of Pax-2, Pax-6, MitF, and tyrosinase. In certain embodiments of any of the foregoing, the RPE cells are mature RPE cells with increased pigmentation in comparison to differentiated RPE cells.
  • the invention provides for human RPE cells, cell cultures comprising a substantially purified population of human RPE cells, pharmaceutical preparations comprising human retinal pigmented epithelial cells and cryopreserved preparations of the human RPE cells.
  • the invention provides for the use of the human RPE cells in the manufacture of a medicament to treat a condition in a patient in need thereof.
  • the invention provides the use of the cell cultures in the manufacture of a medicament to treat a condition in a patient in need thereof.
  • the invention also provides the use of the pharmaceutical preparations in the manufacture of a medicament to treat a condition in a patient in need thereof.
  • preparations comprising RPE cells may include differentiated RPE cells of varying levels of maturity, or may be substantially pure with respect to differentiated RPE cells of a particular level of maturity.
  • the preparations comprising RPE cells are prepared in accordance with Good Manufacturing Practices (GMP) (e.g., the preparations are GMP-compliant).
  • GMP Good Manufacturing Practices
  • the preparations comprising RPE cells are substantially free of bacterial, viral, or fungal contamination or infection.
  • the human RPE cells can be identified and characterized based on their structural properties. Specifically, and in certain embodiments, these cells are unique in that they can be identified or characterized based on the expression or lack of expression (as assessed at the level of the gene or the level of the protein) of one or more markers.
  • differentiated ES-derived RPE cells can be identified or characterized based on expression of one or more (e.g., the cells can be characterized based on expression of at least one, at least two, at least three, at least four, at least five, or at least six) of the following markers: RPE-65, Bestrophin, PEDF, CRALBP, Otx2, and Mit-F. Additionally or alternatively, ES-derived RPE cells can be identified or characterized based on expression of PAX2, tyrosinase, and/or PAX6.
  • hRPE cells can be identified or characterized based on expression or lack of expression (as assessed at the level of the gene or the level of the protein) of one or more (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10) markers analyzed in any of Tables 1-3.
  • ES-derived RPE cells can also be identified and characterized based on the degree of pigmentation of the cell. Differentiated hRPE cells that are rapidly dividing are lightly pigmented. However, when cell density reaches maximal capacity, or when hRPE cells are specifically matured, hRPE take on their characteristic phenotypic hexagonal shape and increase pigmentation level by accumulating melanin and lipofuscin. As such, initial accumulation of pigmentation serves as an indicator of RPE differentiation and increased pigmentation associated with cell density serves as an indicator of RPE maturity.
  • Preparations comprising RPE cells include preparations that are substantially pure, with respect to non-RPE cell types, but which contain a mixture of differentiated RPE cells and mature differentiated RPE cells. Preparations comprising RPE cells also include preparations that are substantially pure both respect to non-RPE cell types and with respect to RPE cells of other levels of maturity.
  • the invention contemplates that the RPE cells (characterized as described above) may be derived from human pluripotent stem cells, for example iPS cells and embryonic stem cells.
  • the RPE cells are derived from human pluripotent stem cells using any of the methods described herein.
  • Embryonic stem cells can be indefinitely maintained in vitro in an undifferentiated state and yet are capable of differentiating into virtually any cell type, providing a limitless supply of rejuvenated and histocompatible cells for transplantation therapy.
  • the problem of immune rejection can be overcome with nuclear transfer and parthenogenetic technology.
  • human embryonic stem (hES) cells are useful for studies on the differentiation of human cells and can be considered as a potential source for transplantation therapies.
  • adipocytes (Bost et al., 2002, Aubert et al., 1999), hepatocyte-like cells (Rambhatla et al., 2003), hematopoetic cells (Chadwick et al., 2003).
  • oocytes Hubner et all., 2003
  • thymocyte-like cells Libner et all., 2003
  • pancreatic islet cells Kahan, 2003
  • osteoblasts Zur Nieden et al., 2003).
  • the present invention provides for the differentiation of human ES cells into a specialized cell in the neuronal lineage, the retinal pigment epithelium (RPE).
  • RPE is a densely pigmented epithelial monolayer between the choroid and neural retina. It serves as a part of a barrier between the bloodstream and retina. Its functions include phagocytosis of shed rod and cone outer segments, absorption of stray light, vitamin A metabolism, regeneration of retinoids, and tissue repair (Grierson et al., 1994, Fisher and Reh, 2001, Marmorstein et al., 1998).
  • the RPE can be recognized by its cobblestone cellular morphology of black pigmented cells.
  • RPE retinaldehyde-binding protein
  • CRALBP retinaldehyde-binding protein
  • RPE65 a cytoplasmic protein involved in retinoid metabolism (Ma et al., 2001, Redmond et al., 1998); bestrophin, the product of the Best vitelliform macular dystrophy gene (VMD2, Marmorstein et al., 2000), and pigment epithelium derived factor (PEDF), a 48 kD secreted protein with angiostatic properties (Karakousis et al., 2001, Jablonski et al., 2000).
  • CRALBP retinaldehyde-binding protein
  • VMD2 Best vitelliform macular dystrophy gene
  • PEDF pigment epithelium derived factor
  • RPE plays an important role in photoreceptor maintenance, and various RPE malfunctions in vivo are associated with a number of vision-altering ailments, such as RPE detachment, displasia, atrophy, retinopathy, retinitis pigmentosa, macular dystrophy or degeneration, including age-related macular degeneration, which can result in photoreceptor damage and blindness. Because of its wound healing abilities, RPE has been extensively studied in application to transplantation therapy.
  • hES cells As a source of immune compatible tissues, hES cells hold a promise for transplantation therapy, as the problem of immune rejection can be overcome with nuclear transfer technology.
  • the use of the new differentiation derivatives of human ES cells, including retinal pigment epithelium-like cells and neuronal precursor cells, and the use of the differentiation system for producing the same offers an attractive potential supply of RPE and neuronal precursor cells for transplantation.
  • one aspect of the present invention is to provide an improved method of generating RPE cells derived from human embryonic stem cells, which may be purified and/or isolated. Such cells are useful for therapy for retinal degeneration diseases such as, for example, retinitis pigmentosa, macular degeneration and other eye disorders.
  • the cell types that can be produced using this invention include, but are not limited to, RPE cells and RPE progenitor cells.
  • Cells that may also be produced include iris pigmented epithelial (IPE) cells and other vision associated neural cells, such as internuncial neurons (e.g. “relay” neurons of the inner nuclear layer (INL)) and amacrine cells.
  • IPE iris pigmented epithelial
  • other vision associated neural cells such as internuncial neurons (e.g. “relay” neurons of the inner nuclear layer (INL)) and amacrine cells.
  • retinal cells, rods, cones, and corneal cells can be produced.
  • the human embryonic stem cells are the starting material of this method.
  • the embryonic stem cells may be cultured in any way known in the art, such as in the presence or absence of feeder cells.
  • human ES cells produced using any method can be used as the starting material to produce RPE cells.
  • the human ES cells may be derived from blastocyst stage embryos that were the product of in vitro fertilization of egg and sperm.
  • the human ES cells may be derived from one or more blastomeres removed from an early cleavage stage embryo, optionally, without destroying the remainder of the embryo.
  • the human ES cells may be produced using nuclear transfer.
  • previously cryopreserved human ES cells can be used.
  • human embryonic stem cells are cultured as embryoid bodies.
  • Embryonic stem cells may be pelleted, resuspended in culture medium, and plated on culture dishes (e.g., low attachment culture dishes).
  • Cells may be cultured in any medium that is sufficient for growth of cells at high-density, such as, commercially available medium for viral, bacterial, insect, or animal cell culture.
  • nutrient rich, low protein medium is used (e.g., MDBK-GM medium, containing about 150 mg/mL (0.015%) animal-derived protein).
  • the medium When low protein medium is used, the medium contains, for example, less than or equal to about 5%, 4%, 3%, 2.5%, 2%, 1.5%, 1%, 0.75%, 0.5%, 0.25%, 0.2%, 0.1%, 0.05%, 0.02%, 0.016%, 0.015%, or even less than or equal to 0.010% animal-derived protein.
  • reference to the percentage of protein present in low protein medium refers to the medium alone and does not account for protein present in, for example, B-27 supplement. Thus, it is understood that when cells are cultured in low protein medium and B-27 supplement, the percentage of protein present in the medium may be higher.
  • nutrient rich, protein-free medium is used (e.g., MDBK-MM medium).
  • culture media include, for example, OptiPro SFM, VP-SFM, and EGM-2.
  • Such media may include nutrient components such as insulin, transferrin, sodium selenite, glutamine, albumin, ethanolamine, fetuin, peptone, purified lipoprotein material, vitamin A, vitamin C, and vitamin E.
  • cell cultures in either low protein or protein free medium are supplemented with serum free B-27 supplement (Brewer et al., Journal of Neuroscience Research 35:567-576 (1993)).
  • Nutrient components of B27 supplement may include biotin, L-carnitine, corticosterone, ethanolamine, D+-galactose, reduced glutathione, lineleic acid, linolenic acid, progesterone, putrescine, retinyl acetate, selenium, triodo-1-thyronine (T3), DL-alpha-tocopherol (vitamin E), DL-alpha-tocopherol acedate, bovine serum albumin, catalase, insulin, superoxide dismutase, and transferrin.
  • protein free refers to the medium prior to addition of B-27.
  • the medium may also contain supplements such as heparin, hydrocortisone, ascorbic acid, serum (such as, for example, fetal bovine serum), or a growth matrix (such as, for example, extracellular matrix from bovine corneal epithelium, matrigel (BD biosciences), or gelatin).
  • supplements such as heparin, hydrocortisone, ascorbic acid, serum (such as, for example, fetal bovine serum), or a growth matrix (such as, for example, extracellular matrix from bovine corneal epithelium, matrigel (BD biosciences), or gelatin).
  • RPE cells differentiate from the embryoid bodies. Isolating RPE cells from the EBs allows for the expansion of the RPE cells in an enriched culture in vitro.
  • RPE cells may be obtained form EBs grown for less than 90 days.
  • RPE cells arise in human EBs grown for 7-14 days.
  • RPE cells arise in human EBs grown for 14-28 days.
  • RPE cells are identified and may be isolated from human EBs grown for 28-45 days.
  • RPE cells arise in human EBs grown for 45-90 days.
  • the medium used to culture embryonic stem cells, embryoid bodies, and RPE cells may be removed and/or replaced with the same or different media at any interval.
  • the medium may be removed and/or replaced after 0-7 days, 7-10 days, 10-14 days, 14-28 days, or 28-90 days.
  • the medium is replaced at least daily, every other day, or at least every three days.
  • the RPE cells that differentiate from the EBs are washed and dissociated (e.g., by Trypsin/EDTA, collegenase B, collegenase IV, or dispase).
  • a non-enzymatic solution is used to disassociate the cells, such as a high EDTA-containing solution such as, for example, Hanks-based cell disassociation buffer.
  • RPE cells are selected from the dissociated cells and cultured separately to produce a substantially purified culture of RPE cells.
  • RPE cells are selected based on characteristics associated with RPE cells. For example, RPE cells can be recognized by cobblestone cellular morphology and pigmentation.
  • RPE retinaldehyde-binding protein
  • CRALBP retinaldehyde-binding protein
  • RPE65 a cytoplasmic protein involved in retinoid metabolism (Ma et al., 2001, Redmond et al., 1998); bestrophin, the product of the Best vitelliform macular dystrophy gene (VMD2, Marmorstein et al., 2000), and pigment epithelium derived factor (PEDF), a 481(D secreted protein with angiostatic properties (Karakousis et al., 2001, Jablonski et al., 2000).
  • CRALBP retinaldehyde-binding protein
  • VMD2 Best vitelliform macular dystrophy gene
  • PEDF pigment epithelium derived factor
  • RPE cells can be selected based on cell function, such as by phagocytosis of shed rod and cone outer segments, absorption of stray light, vitamin A metabolism, regeneration of retinoids, and tissue repair (Grierson et al., 1994, Fisher and Reh, 2001, Marmorstein et al., 1998). Evaluation may also be performed using behavioral tests, fluorescent angiography, histology, tight junctions conductivity, or evaluation using electron microscopy.
  • Another embodiment of the present invention is a method of identifying RPE cells by comparing the messenger RNA transcripts of such cells with cells derived in-vivo. In certain embodiments, an aliquot of cells is taken at various intervals during the differentiation of embryonic stem cells to RPE cells and assayed for the expression of any of the markers described above. These characteristic distinguish differentiated RPE cells.
  • RPE cell culture media may be supplemented with one or more growth factors or agents.
  • Growth factors that may be used include, for example, EGF, FGF, VEGF, and recombinant insulin-like growth factor.
  • Other growth factors that may be used in the present invention include 6Ckine (recombinant), activin A, AlphaA-interferon, alpha-interferon, amphiregulin, angiogenin, B-endothelial cell growth factor, beta cellulin, B-interferon, brain derived neurotrophic factor, C10 (recombinant), cardiotrophin-1, ciliary neurotrophic factor, cytokine-induced neutrophil chemoattractant-1, endothelial cell growth supplement, eotaxin, epidermal growth factor, epithelial neutrophil activating peptide-78, erythropoiten, estrogen receptor-alpha, estrogen receptor-B, fibroblast growth factor (acidic/basic, heparin stabilized, recombinant), FL
  • Agents that can be used according to the present invention include cytokines such as interferon-alpha A, interferon-alpha A/D, interferon-.beta., interferon-gamma, interferon-gamma-inducible protein-10, interleukin-1, interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-1, interleukin-12, interleukin-13, interleukin-15, interleukin-17, keratinocyte growth factor, leptin, leukemia inhibitory factor, macrophage colony-stimulating factor, and macrophage inflammatory protein-1 alpha.
  • cytokines such as interferon-alpha A, interferon-alpha A/D, interferon-.beta., interferon-gamma, interferon-gamma-induc
  • Agents according to the invention also include hormones and hormone antagonists, such as 17B-estradiol, adrenocorticotropic hormone, adrenomedullin, alpha-melanocyte stimulating hormone, chorionic gonadotropin, corticosteroid-binding globulin, corticosterone, dexamethasone, estriol, follicle stimulating hormone, gastrin 1, glucagon, gonadotropin, hydrocortisone, insulin, insulin-like growth factor binding protein, L-3,3′,5′-triiodothyronine, L-3,3′,5-triiodothyronine, leptin, leutinizing hormone, L-thyroxine, melatonin, MZ-4, oxytocin, parathyroid hormone, PEC-60, pituitary growth hormone, progesterone, prolactin, secretin, sex hormone binding globulin, thyroid stimulating hormone, thyrotropin releasing
  • agents according to the invention include extracellular matrix components such as fibronectin, proteolytic fragments of fibronectin, laminin, thrombospondin, aggrecan, and syndezan.
  • Agents according to the invention also include antibodies to various factors, such as anti-low density lipoprotein receptor antibody, anti-progesterone receptor, internal antibody, anti-alpha interferon receptor chain 2 antibody, anti-c-c chemokine receptor 1 antibody, anti-CD 118 antibody, anti-CD 119 antibody, anti-colony stimulating factor-1 antibody, anti-CSF-1 receptor/c-fins antibody, anti-epidermal growth factor (AB-3) antibody, anti-epidermal growth factor receptor antibody, anti-epidermal growth factor receptor, phospho-specific antibody, anti-epidermal growth factor (AB-1) antibody, anti-erythropoietin receptor antibody, anti-estrogen receptor antibody, anti-estrogen receptor, C-terminal antibody, anti-estrogen receptor-B antibody, anti-fibroblast growth factor receptor antibody, anti-fibroblast growth factor, basic antibody, anti-gamma-interferon receptor chain antibody, anti-gamma-interferon human recombinant antibody, anti-GFR alpha-1
  • Growth factors, agents, and other supplements described herein may be used alone or in combination with other factors, agents, or supplements. Factors, agents, and supplements may be added to the media immediately or any time after cell culture.
  • the RPE cells are further cultured to produce a culture of mature RPE cells.
  • the medium used to culture the RPE cells can be any medium appropriate for high-density cell culture growth, such as described herein.
  • the cells described herein may be cultured in VP-SFM, EGM-2, and MDBK-MM.
  • a previously derived culture of human embryonic stem cells is provided.
  • the hES cells can be, for example, previously derived from a blastocyst (produced by fertilization or nuclear transfer) or from one or more blastomeres from an early cleavage stage embryo (optionally without destroying the remainder of the embryo).
  • the human ES cells are cultured as a suspension culture to produce embryoid bodies (EBs).
  • the embryoid bodies are cultured in suspension for approximately 7-14 days.
  • the EBs can be cultured in suspension for fewer than 7 days (less than 7, 6, 5, 4, 3, 2, or less than 1 day) or greater than 14 days.
  • the EBs can be cultured in medium optionally supplemented with B-27 supplement.
  • the EBs can transferred to produce an adherent culture.
  • the EBs can be plated in medium onto gelatin coated plates.
  • the media is not supplemented with B-27 supplement when the cells are cultured as an adherent culture.
  • the medium is supplemented with B-27 initially (e.g., for less than or equal to about 7 days), but then subsequently cultured in the absence of B-27 for the remainder of the period as an adherent culture.
  • the EBs can be cultured as an adherent culture for approximately 14-28. However, in certain embodiments, the EBs can be cultured for fewer than 14 days (less than 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or less than 1 day) or greater than 28 days.
  • RPE cells begin to differentiate from amongst cells in the adherent culture of EBs. RPE cells can be visually recognized based on their cobblestone morphology and the initial appearance of pigmentation. As RPE cells continue to differentiate, clusters of RPE cells can be observed.
  • RPE cells are dissociated from each other and from non-RPE cells using mechanical and/or chemical methods. A suspension of RPE cells can then be transferred to fresh medium and a fresh culture vessel to provide an enriched population of RPE cells.
  • Enriched cultures of RPE cells can be cultured in appropriate medium, for example, EGM-2 medium.
  • EGM-2 medium This, or a functionally equivalent or similar medium, may be supplemented with one or more growth factors or agents (e.g., bFGF, heparin, hydrocortisone, vascular endothelial growth factor, recombinant insulin-like growth factor, ascorbic acid, human epidermal growth factor).
  • growth factors or agents e.g., bFGF, heparin, hydrocortisone, vascular endothelial growth factor, recombinant insulin-like growth factor, ascorbic acid, human epidermal growth factor.
  • the RPE cells can be further cultured in, for example MDBK-MM medium until the desired level of maturation is obtained. This can be determined by monitoring the increase in pigmentation level during maturation.
  • MDBK-MM medium a functionally equivalent or similar medium, may be used. Regardless of the particular medium used to mature the RPE cells, the medium may optionally be supplemented with one or more growth factors or agents.
  • the culture of RPE cells can be substantially pure RPE cells containing less than 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% non-RPE cells.
  • the substantially purified (with respect to non-RPE cells) cultures contain RPE cells of varying levels of maturity. In other embodiments, the cultures are substantially pure both with respect to non-RPE cells and with respect to RPE cells of differing level of maturity.
  • the invention contemplates that the RPE cells (characterized as described above) may be derived from human pluripotent stem cells, for example iPS cells and embryonic stem cells.
  • the RPE cells are derived from human pluripotent stem cells using any of the methods described herein.
  • the present invention provides preparations of human pluripotent stem cell-derived RPE cells.
  • the preparation is a preparation of human embryonic stem cell-derived RPE cells.
  • the preparation is a preparation of human iPS cell-derived RPE cells.
  • the preparations are substantially purified (with respect to non-RPE cells) preparations comprising differentiated ES-derived RPE cells.
  • substantially purified with respect to non-RPE cells, is meant that the preparation comprises at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or even greater than 99% RPE cells.
  • the substantially purified preparation of RPE cells contains less than 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% non-RPE cell type.
  • the RPE cells in such a substantially purified preparation contain RPE cells of varying levels of maturity/pigmentation.
  • the RPE cells are substantially pure, both with respect to non-RPE cells and with respect to RPE cells of other levels of maturity.
  • the preparations are substantially purified, with respect to non-RPE cells, and enriched for mature RPE cells.
  • enriched for mature RPE cells it is meant that at least 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or even greater than 99% of the RPE cells are mature RPE cells.
  • the preparations are substantially purified, with respect to non-RPE cells, and enriched for differentiated RPE cells rather than mature RPE cells.
  • RPE cells are differentiated RPE cells rather than mature RPE cells.
  • mature RPE cells are distinguished from RPE cells by one or more of: the level of pigmentation, level of expression of Pax-2, Pax-6, and/or tyrosinase.
  • the preparations include at least 1 ⁇ 10 3 RPE cells, 5 ⁇ 10 3 RPE cells, 1 ⁇ 10 4 RPE cells, 5 ⁇ 10 4 RPE cells, 1 ⁇ 10 5 RPE cells, 2 ⁇ 10 5 RPE cells, 3 ⁇ 10 5 RPE cells, 4 ⁇ 10 5 RPE cells, 5 ⁇ 10 5 RPE cells, 6 ⁇ 10 5 RPE cells, 7 ⁇ 10 5 RPE cells, 8 ⁇ 10 5 RPE cells, 9 ⁇ 10 5 RPE cells, 1 ⁇ 10 6 RPE cells, 5 ⁇ 10 6 RPE cells, 6 ⁇ 10 6 RPE cells, 7 ⁇ 10 6 RPE cells, 8 ⁇ 10 6 RPE cells, 9 ⁇ 10 6 RPE cells, 1 ⁇ 10 7 RPE cells, 5 ⁇ 10 7 RPE cells, 1 ⁇ 10 8 RPE cells, 1 ⁇ 10 9 RPE cells, or even more than 1 ⁇ 10 9 RPE cells.
  • the ES-derived RPE cells do not express ES cell markers.
  • expression of the ES cell genes Oct-4, nanog, and/or Rex-1 is approximately 100-1000 fold lower in RPE cells than in ES cells, as assessed by quantitative RT-PCR.
  • RPE cells are substantially negative for Oct-4, nanog, and/or Rex-1 gene expression.
  • the ES-derived RPE cells express, at the mRNA and protein level, one or more of the following: RPE65, bestrophin, PEDF, CRALBP, Otx2, and MitF. In certain embodiments, RPE cells express two or more, three or more, four or more, five or more, or six of these markers. In certain embodiments, the RPE cells additionally or alternatively express, at the mRNA and protein level, one or more (1, 2, or 3) of the following: pax-2, pax6, and tyrosinase. In other embodiments, the level of maturity of the RPE cells is assessed by expression of one or more (1, 2, or 3) of pax-2, pax6, and tyrosinase.
  • the ES-derived RPE cells express, at the mRNA and/or protein level, one or more (1, 2, 3, 4, 5, 6, 7, 8, or 9) of the RPE-specific genes listed in Table 1 (pax-6, pax-2, RPE65, PEDF, CRALBP, bestrophin, mitF, Otx-2, and tyrosinase, as well as one or more (1, 2, 3, or 4) of the neuroretina genes listed in Table 1 (CHX10, NCAM, nestin, beta-tubulin).
  • the RPE cells do not substantially express the ES cell specific genes Oct-4, nanog, and/or Rex-1 (e.g., expression of the ES cell specific genes is 100-1000 fold less in RPE cells, as determined by quantitative RT-PCR).
  • the ES-derived RPE cells express, at the mRNA and/or protein level, one or more (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or more than 48) of the genes listed in Table 2, and the expression of the one or more genes is increased in RPE cells relative to the level of expression (if any) in human ES cells.
  • the ES-derived RPE cells express, at the mRNA and/or protein level one or more (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or more than 25) of the genes listed in Table 3, but the expression of the one or more genes is decreased (including decreased to nearly undetectable levels) in RPE cells relative to the level of expression in human ES cells.
  • the substantially purified preparation of RPE cells comprises RPE cells of differing levels of maturity (e.g., differentiated RPE cells and mature differentiated RPE cells).
  • RPE cells of differing levels of maturity e.g., differentiated RPE cells and mature differentiated RPE cells.
  • RPE cells may have substantially the same expression of RPE65, PEDF, CRALBP, and bestrophin.
  • the RPE cells may vary, depending on level of maturity, with respect to expression of one or more of pax-2, pax-6, mitF, and/or tyrosinase.
  • the ES-derived RPE cells are stable, terminally differentiated RPE cells that do not de-differentiate to a non-RPE cell type. In certain embodiments, the ES-derived RPE cells are functional RPE cells.
  • the ES-derived RPE cells are characterized by the ability to integrate into the retina upon corneal, sub-retinal, or other transplantation or administration into an animal.
  • the preparations are produced in compliance with GMP standards. As such, in certain embodiments, the preparations are GMP compliant preparations. In other embodiments, the preparations are substantially free of viral, bacterial, and/or fungal infection and contamination.
  • the preparations are cryopreserved for storage and future use.
  • the invention provides cryopreserved preparations comprising substantially purified RPE cells.
  • Cryopreserved preparations are formulated in excipients suitable to maintain cell viability during and following cryopreservation.
  • the cryopreserved preparation comprises at least 1 ⁇ 10 3 RPE cells, 5 ⁇ 10 3 RPE cells, 1 ⁇ 10 4 RPE cells, 5 ⁇ 10 4 RPE cells, 1 ⁇ 10 5 RPE cells, 2 ⁇ 10 5 RPE cells, 3 ⁇ 10 5 RPE cells, 4 ⁇ 10 5 RPE cells, 5 ⁇ 10 5 RPE cells, 6 ⁇ 10 5 RPE cells, 7 ⁇ 10 5 RPE cells, 8 ⁇ 10 5 RPE cells, 9 ⁇ 10 5 RPE cells, 1 ⁇ 10 6 RPE cells, 5 ⁇ 10 6 RPE cells, 6 ⁇ 10 6 RPE cells, 7 ⁇ 10 6 RPE cells, 8 ⁇ 10 6 RPE cells, 9 ⁇ 10 6 RPE cells, 1 ⁇ 10 7 RPE cells, 5 ⁇ 10 7 RPE cells, 1 ⁇ 10 8 RPE cells, 1 ⁇ 10 9 RPE cells, or even more than 1 ⁇ 10 9 RPE cells.
  • Cryopreserved preparations may have the same levels of purity with respect to non-RPE cells and/or with respect to RPE cells of varying levels of maturity as detailed above.
  • at least 65% of the RPE cells in a cryopreserved preparation of RPE cells retain viability following thawing.
  • at least 70%, 75%, 80%, 85%, 90%, 81%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or greater than 99% of the RPE cells in a cryopreserved preparation of RPE cells retain viability following thawing.
  • the RPE cells provided herein are human cells. Note, however, that the human cells may be used in human patients, as well as in animal models or animal patients. For example, the human cells may be tested in rat, dog, or non-human primate models of retinal degeneration. Additionally, the human cells may be used therapeutically to treat animals in need thereof, such as in a veterinary medical setting.
  • Preparations may be formulated as pharmaceutical preparations prepared in a pharmaceutically acceptable carrier or excipient.
  • Preferred preparations are specifically formulated for administration to the eye (e.g., sub-retinal, corneal, ocular, etc.)
  • the RPE cells are derived from human pluripotent stem cells, such as human embryonic stem cells or human iPS cells.
  • human pluripotent stem cells such as human embryonic stem cells or human iPS cells.
  • the invention contemplates that any of the preparations described herein may be derived from an appropriate human pluripotent stem cell.
  • the invention contemplates that any of the foregoing preparations of RPE cells, including substantially purified preparations and preparations have a particular minimal number of RPE cells, may be used in the treatment of any of the indications described herein. Further, RPE cells differentiated using any of the methods described herein may be used in the treatment of any of the indications described herein.
  • RPE cells and pharmaceutically preparations comprising RPE cells produced by the methods described herein and/or having the characteristics of RPE cell preparations described herein may be used for cell-based treatments in which RPE cells are needed or would improve treatment.
  • the following section describes methods of using RPE cells provided by the present invention for treating various conditions that may benefit from RPE cell-based therapies.
  • the particular treatment regimen, route of administration, and any adjuvant therapy will be tailored based on the particular condition, the severity of the condition, and the patient's overall health.
  • administration of RPE cells may be effective to fully restore any vision loss or other symptoms.
  • administration of RPE cells may be effective to reduce the severity of the symptoms and/or to prevent further degeneration in the patient's condition.
  • the invention contemplates that administration of a preparation comprising RPE cells can be used to treat (including reducing the severity of the symptoms, in whole or in part) any of the foregoing or following conditions. Additionally, RPE cell administration may be used to help treat the symptoms of any injury to the endogenous RPE layer.
  • RPE cells including preparations comprising RPE cells, derived using any of the methods described herein can be used in the treatment of any of the indications described herein. Further, the invention contemplates that any of the preparations comprising RPE cells described herein can be used in the treatment of any of the indications described herein.
  • Retinitis pigmentosa is a hereditary condition in which the vision receptors are gradually destroyed through abnormal genetic programming. Some forms cause total blindness at relatively young ages, where other forms demonstrate characteristic “bone spicule” retinal changes with little vision destruction. This disease affects some 1.5 million people worldwide. Two gene defects that cause autosomal recessive retinitis pigmentosa have been found in genes expressed exclusively in RPE. One is due to an RPE protein involved in vitamin A metabolism (cis retinaldehyde binding protein). The second involves another protein unique to RPE, RPE65. This invention provides methods and compositions for treating both of these forms of retinitis pigmentosa by administration of RPE cells.
  • the present invention provides methods and compositions for treating disorders associated with retinal degeneration, including macular degeneration.
  • a further aspect of the present invention is the use of RPE cells for the therapy of eye diseases, including hereditary and acquired eye diseases.
  • eye diseases including hereditary and acquired eye diseases.
  • acquired or hereditary eye diseases are age-related macular degeneration, glaucoma and diabetic retinopathy.
  • Age-related macular degeneration is the most common reason for legal blindness in western countries. Atrophy of the submacular retinal pigment epithelium and the development of choroidal neovascularizations (CNV) results secondarily in loss of central visual acuity. For the majority of patients with subfoveal CNV and geographic atrophy there is at present no treatment available to prevent loss of central visual acuity. Early signs of AMD are deposits (druses) between retinal pigment epithelium and Bruch's membrane. During the disease there is sprouting of choroid vessels into the subretinal space of the macula. This leads to loss of central vision and reading ability.
  • AMD Age-related macular degeneration
  • Glaucoma is the name given to a group of diseases in which the pressure in the eye increases abnormally. This leads to restrictions of the visual field and to the general diminution in the ability to see.
  • the most common form is primary glaucoma; two forms of this are distinguished: chronic obtuse-angle glaucoma and acute angle closure. Secondary glaucoma may be caused by infections, tumors or injuries.
  • a third type, hereditary glaucoma is usually derived from developmental disturbances during pregnancy.
  • the aqueous humor in the eyeball is under a certain pressure which is necessary for the optical properties of the eye. This intraocular pressure is normally 15 to 20 millimeters of mercury and is controlled by the equilibrium between aqueous production and aqueous outflow.
  • Glaucoma In glaucoma, the outflow of the aqueous humor in the angle of the anterior chamber is blocked so that the pressure inside the eye rises. Glaucoma usually develops in middle or advanced age, but hereditary forms and diseases are not uncommon in children and adolescents. Although the intraocular pressure is only slightly raised and there are moreover no evident symptoms, gradual damage occurs, especially restriction of the visual field. Acute angle closure by contrast causes pain, redness, dilation of the pupils and severe disturbances of vision. The cornea becomes cloudy, and the intraocular pressure is greatly increased. As the disease progresses, the visual field becomes increasingly narrower, which can easily be detected using a perimeter, an ophthalmologic instrument. Chronic glaucoma generally responds well to locally administered medicaments which enhance aqueous outflow.
  • Diabetic retinopathy arises in cases of diabetes mellitus. It can lead to thickening of the basal membrane of the vascular endothelial cells as a result of glycosilation of proteins. It is the cause of early vascular sclerosis and the formation of capillary aneurysms. These vascular changes lead over the course of years to diabetic retinopathy. The vascular changes cause hypoperfusion of capillary regions. This leads to lipid deposits (hard exudates) and to vasoproliferation. The clinical course is variable in patients with diabetes mellitus. In age-related diabetes (type II diabetes), capillary aneurysms appear first.
  • the fatty deposits are arranged like a corona around the macula (retinitis circinata). These changes are frequently accompanied by edema at the posterior pole of the eye. If the edema involves the macula there is an acute serious deterioration in vision.
  • the main problem in type I diabetes is the vascular proliferation in the region of the fundus of the eye.
  • the standard therapy is laser coagulation of the affected regions of the fundus of the eye. The laser coagulation is initially performed focally in the affected areas of the retina.
  • the RPE cells of the invention may be used to treat disorders of the central nervous system.
  • RPE cells may be transplanted into the CNS.
  • CNS central nervous system
  • fetal cells obtained from brains of human fetuses.
  • Fetal cells from the ventral midbrain or dopaminergic neurons have already been transplanted in clinical studies on more than 300 patients with Parkinson's disease (for review, see Alexi T, Borlongan C V, Faull R L, Williams C E, Clark R G, Gluckman P D, Hughes P E (2000) (Neuroprotective strategies for basal ganglia degeneration: Parkinson's and Huntington's diseases.
  • Prog Neurobiol 60: 409 470 A number of different cell types, including non-neuronal cells, e.g. cells from the adrenal cortex, Sertoli cells on the gonads or glomus cells from the carotid bodies, fibroblasts or astrocytes, have been used in patients with Parkinson's disease or in animal models with the aim of replacing dopamine spontaneously or after gene transfer (Alexi et al. 2000, supra).
  • the survival rate of transplanted fetal dopaminergic neurons is 5 8%, which was enough to cause a slight improvement in the signs and symptoms (Alexi et al. 2000, supra).
  • neuronal stem cells from brains of adult vertebrates have been isolated, expanded in vitro and reimplanted into the CNS, after which they differentiated into pure neurons. Their function in the CNS remains uncertain, however. Neuronal precursor cells have also been used for gene transfer (Raymon H K, Thode S, Zhou J, Friedman G C, Pardinas J R, Barrere C, Johnson R M, Sah D W (1999) Immortalized human dorsal root ganglion cells differentiate into neurons with nociceptive properties. J Neurosci 19: 5420 5428).
  • Schwann cells which overexpressed NGF and GDNF had neuroprotective effects in models of Parkinsonism (Wilby M J, Sinclair S R, Muir E M, Zietlow R, Adcock K H, Horellou P, Rogers J H, Dunnett S B, Fawcett J W (1999)
  • Another aspect of the present invention is therefore the use of pigment epithelial cells for the therapy of nerve diseases, in particular a disease of the nervous system, preferably of the CNS, especially of Parkinson's disease.
  • Parkinson's disease is a chronic degenerative disease of the brain.
  • the disease is caused by degeneration of specialized neuronal cells in the region of the basal ganglia.
  • the death of dopaminergic neurons results in reduced synthesis of dopamine, an important neurotransmitter, in patients with Parkinson's disease.
  • the standard therapy is medical therapy with L-dopa.
  • L-Dopa is metabolized in the basal ganglia to dopamine and there takes over the function of the missing endogenous neurotransmitter.
  • L-dopa therapy loses its activity after some years.
  • Animal models of retinitis pigmentosa that may be treated or used to test the efficacy of the RPE cells produced using the methods described herein include rodents (rd mouse, RPE-65 knockout mouse, tubby-like mouse, RCS rat), cats (Abyssinian cat), and dogs (cone degeneration “cd” dog, progressive rod-cone degeneration “prcd” dog, early retinal degeneration “erd” dog, rod-cone dysplasia 1, 2 & 3 “rcd1, rcd2 & rcd3” dogs, photoreceptor dysplasia “pd” dog, and Briard “RPE-65” (dog)).
  • rodents rd mouse, RPE-65 knockout mouse, tubby-like mouse, RCS rat
  • cats Abyssinian cat
  • dogs cone degeneration “cd” dog, progressive rod-cone degeneration “prcd” dog, early retinal degeneration “erd” dog, rod-con
  • Another embodiment of the present invention is a method for the derivation of RPE lines or precursors to RPE cells that have an increased ability to prevent neovascularization.
  • Such cells can be produced by aging a somatic cell from a patient such that telomerase is shortened where at least 10% of the normal replicative lifespan of the cell has been passed, then the use of said somatic cell as a nuclear transfer donor cell to create cells that overexpress angiogenesis inhibitors such as Pigment Epithelium Derived Factor (PEDF/EPC-1).
  • PEDF/EPC-1 Pigment Epithelium Derived Factor
  • Such cells may be genetically modified with exogenous genes that inhibit neovascularization.
  • the invention contemplates that preparations of RPE cells differentiated from human pluripotent stem cells (e.g., human embryonic stem cells, iPS cells, or other pluripotent stem cells) can be used to treat any of the foregoing diseases or conditions, as well as injuries of the endogenous RPE layer. These diseases can be treated with preparations of RPE cells comprising a mixture of differentiated RPE cells of varying levels of maturity, as well as with preparations of differentiated RPE cells that are enriched for mature differentiated RPE cells or differentiated RPE cells.
  • human pluripotent stem cells e.g., human embryonic stem cells, iPS cells, or other pluripotent stem cells
  • RPE cells of the invention may be administered topically, systemically, or locally, such as by injection (e.g., intravitreal injection), or as part of a device or implant (e.g., a sustained release implant).
  • the cells of the present invention may be transplanted into the subretinal space by using vitrectomy surgery.
  • RPE cells can be added to buffered and electrolyte balanced aqueous solutions, buffered and electrolyte balanced aqueous solutions with a lubricating polymer, mineral oil or petrolatum-based ointment, other oils, liposomes, cylcodextrins, sustained release polymers or gels. These preparations can be administered topically to the eye 1 to 6 times per day for a period up to the lifetime of the patient.
  • methods of treating a patient suffering from a condition associated with retinal degeneration comprise administering a composition of the invention locally (e.g., by intraocular injection or insertion of a sustained release device that releases a composition of the invention), by topical means or by systemic administration (e.g., by routes of administration that allow in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body, including, without limitation, by intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular routes).
  • a composition of the invention locally (e.g., by intraocular injection or insertion of a sustained release device that releases a composition of the invention), by topical means or by systemic administration (e.g., by routes of administration that allow in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body, including, without limitation, by intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular routes).
  • Intraocular administration of compositions of the invention includes, for example, delivery into the vitreous body, transcorneally, sub-conjunctival, juxtascleral, posterior scleral, and sub-tenon portions of the eye. See, for example, U.S. Pat. Nos. 6,943,145; 6,943,153; and 6,945,971, the contents of which are hereby incorporated by reference.
  • RPE cells of the invention may be delivered in a pharmaceutically acceptable ophthalmic formulation by intraocular injection.
  • the solution should be concentrated so that minimized volumes may be delivered. Concentrations for injections may be at any amount that is effective and non-toxic, depending upon the factors described herein.
  • RPE cells for treatment of a patient are formulated at doses of about 10 4 cells/mL. In other embodiments, RPE cells for treatment of a patient are formulated at doses of about 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , or 10 10 cells/mL.
  • RPE cells may be formulated for delivery in a pharmaceutically acceptable ophthalmic vehicle, such that the composition is maintained in contact with the ocular surface for a sufficient time period to allow the cells to penetrate the affected regions of the eye, as for example, the anterior chamber, posterior chamber, vitreous body, aqueous humor, vitreous humor, cornea, iris/ciliary, lens, choroid, retina, sclera, suprachoridal space, conjunctiva, subconjunctival space, episcleral space, intracorneal space, epicorneal space, pars plana, surgically-induced avascular regions, or the macula.
  • a therapeutic method of the invention includes the step of administering RPE cells of the invention as an implant or device.
  • the device is bioerodible implant for treating a medical condition of the eye comprising an active agent dispersed within a biodegradable polymer matrix, wherein at least about 75% of the particles of the active agent have a diameter of less than about 10 ⁇ m.
  • the bioerodible implant is sized for implantation in an ocular region.
  • the ocular region can be any one or more of the anterior chamber, the posterior chamber, the vitreous cavity, the choroid, the suprachoroidal space, the conjunctiva, the subconjunctival space, the episcleral space, the intracorneal space, the epicorneal space, the sclera, the pars plana, surgically-induced avascular regions, the macula, and the retina.
  • the biodegradable polymer can be, for example, a poly(lactic-co-glycolic)acid (PLGA) copolymer.
  • the ratio of lactic to glycolic acid monomers in the polymer is about 25/75, 40/60, 50/50, 60/40, 75/25 weight percentage, more preferably about 50/50.
  • the PLGA copolymer can be about 20, 30, 40, 50, 60, 70, 80 to about 90 percent by weight of the bioerodible implant. In certain preferred embodiments, the PLGA copolymer can be from about 30 to about 50 percent by weight, preferably about 40 percent by weight of the bioerodible implant.
  • the volume of composition administered according to the methods described herein is also dependent on factors such as the mode of administration, number of RPE cells, age and weight of the patient, and type and severity of the disease being treated.
  • the liquid volume comprising a composition of the invention may be from about 0.5 milliliters to about 2.0 milliliters, from about 2.0 milliliters to about 5.0 milliliters, from about 5.0 milliliters to about 10.0 milliliters, or from about 10.0 milliliters to about 50.0 milliliters.
  • the liquid volume comprising a composition of the invention may be from about 5.0 microliters to about 50 microliters, from about 50 microliters to about 250 microliters, from about 250 microliters to about 1 milliliter, from about 1 milliliter to about 5 milliliters, from about 5 milliliters to about 25 milliliters, from about 25 milliliters to about 100 milliliters, or from about 100 milliliters to about 1 liter.
  • RPE cells can be delivered one or more times periodically throughout the life of a patient. For example RPE cells can be delivered once per year, once every 6-12 months, once every 3-6 months, once every 1-3 months, or once every 1-4 weeks. Alternatively, more frequent administration may be desirable for certain conditions or disorders. If administered by an implant or device, RPE cells can be administered one time, or one or more times periodically throughout the lifetime of the patient, as necessary for the particular patient and disorder or condition being treated. Similarly contemplated is a therapeutic regimen that changes over time. For example, more frequent treatment may be needed at the outset (e.g., daily or weekly treatment). Over time, as the patient's condition improves, less frequent treatment or even no further treatment may be needed.
  • outset e.g., daily or weekly treatment
  • patients are also administered immunosuppressive therapy, either before, concurrently with, or after administration of the RPE cells.
  • Immunosuppressive therapy may be necessary throughout the life of the patient, or for a shorter period of time.
  • RPE cells of the present invention are formulated with a pharmaceutically acceptable carrier.
  • RPE cells may be administered alone or as a component of a pharmaceutical formulation.
  • the subject compounds may be formulated for administration in any convenient way for use in human medicine.
  • pharmaceutical compositions suitable for parenteral administration may comprise the RPE cells, in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions of the invention may also contain adjuvants, such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like in the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of one or more agents that delay absorption, such as, e.g., aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include is
  • the therapeutic composition for use in this invention is, of course, in a pyrogen-free, physiologically acceptable form.
  • the composition may desirably be encapsulated or injected in a viscous form into the vitreous humor for delivery to the site of retinal or choroidal damage.
  • the human embryonic stem cells used as the starting point for the method of producing RPE cells of this invention may also be derived from a library of human embryonic stem cells, each of which is hemizygous or homozygous for at least one MHC allele present in a human population.
  • each member of said library of stem cells is hemizygous or homozygous for a different set of MHC alleles relative to the remaining members of the library.
  • the library of stem cells is hemizygous or homozygous for all MHC alleles that are present in a human population.
  • stem cells that are homozygous for one or more histocompatibility antigen genes include cells that are nullizygous for one or more (and in some embodiments, all) such genes.
  • Nullizygous for a genetic locus means that the gene is null at that locus, i.e., both alleles of that gene are deleted or inactivated.
  • Stem cells that are nullizygous for all MHC genes may be produced by standard methods known in the art, such as, for example, gene targeting and/or loss of heterozygosity (LOH). See, for example, United States patent publications US 20040091936, US 20030217374 and US 20030232430, and U.S. provisional application No. 60/729,173, the disclosures of all of which are hereby incorporated by reference herein.
  • the present invention relates to methods of obtaining RPE cells, including a library of RPE cells, with reduced MHC complexity.
  • RPE cells with reduced MHC complexity will increase the supply of available cells for therapeutic applications as it will eliminate the difficulties associated with patient matching.
  • Such cells may be derived from stem cells that are engineered to be hemizygous or homozygous for genes of the MHC complex.
  • a human ES cell may comprise modifications to one of the alleles of sister chromosomes in the cell's MHC complex.
  • a variety of methods for generating gene modifications such as gene targeting, may be used to modify the genes in the MHC complex.
  • the modified alleles of the MHC complex in the cells may be subsequently engineered to be homozygous so that identical alleles are present on sister chromosomes. Methods such as loss of heterozygosity (LOH) may be utilized to engineer cells to have homozygous alleles in the MHC complex.
  • LHO loss of heterozygosity
  • one or more genes in a set of MHC genes from a parental allele can be targeted to generate hemizygous cells.
  • the other set of MHC genes can be removed by gene targeting or LOH to make a null line.
  • This null line can be used further as the embryonic cell line in which to drop arrays of the HLA genes, or individual genes, to make a hemizygous or homozygous bank with an otherwise uniform genetic background.
  • a library of ES cell lines wherein each member of the library is homozygous for at least one HLA gene, is used to derive RPE cells according to the methods of the present invention.
  • the invention provides a library of RPE cells (and/or RPE lineage cells), wherein several lines of ES cells are selected and differentiated into RPE cells. These RPE cells and/or RPE lineage cells may be used for a patient in need of a cell-based therapy.
  • certain embodiments of this invention pertain to a method of administering human RPE cells that have been derived from reduced-complexity embryonic stem cells to a patient in need thereof.
  • this method comprises the steps of: (a) identifying a patient that needs treatment involving administering human RPE cells to him or her; (b) identifying MHC proteins expressed on the surface of the patient's cells; (c) providing a library of human RPE cells of reduced MHC complexity made by the method for producing RPE cells of the present invention; (d) selecting the RPE cells from the library that match this patient's MHC proteins on his or her cells; (e) administering any of the cells from step (d) to said patient.
  • This method may be performed in a regional center, such as, for example, a hospital, a clinic, a physician's office, and other health care facilities. Further, the RPE cells selected as a match for the patient, if stored in small cell numbers, may be expanded prior to patient treatment.
  • Certain aspects of the present invention pertain to the production of RPE cells to reach commercial quantities.
  • RPE cells are produced on a large scale, stored if necessary, and supplied to hospitals, clinicians or other healthcare facilities. Once a patient presents with an indication such as, for example, Stargardt's macular dystrophy, age related macular degeneration, or retinitis pigmentosa, RPE cells can be ordered and provided in a timely manner.
  • the present invention relates to methods of producing RPE cells to attain cells on a commercial scale, cell preparations comprising RPE cells derived from said methods, as well as methods of providing (i.e., producing, optionally storing, and selling) RPE cells to hospitals and clinicians.
  • RPE cells may be harvested, purified and optionally stored prior to a patient's treatment.
  • RPE cells may optionally be patient specific or specifically selected based on HLA or other immunologic profile.
  • the present invention provides methods of supplying RPE cells to hospitals, healthcare centers, and clinicians, whereby RPE cells produced by the methods disclosed herein are stored, ordered on demand by a hospital, healthcare center, or clinician, and administered to a patient in need of RPE cell therapy.
  • a hospital, healthcare center, or clinician orders RPE cells based on patient specific data, RPE cells are produced according to the patient's specifications and subsequently supplied to the hospital or clinician placing the order.
  • the method of differentiating RPE cells from human embryonic stem cells is conducted in accordance with Good Manufacturing Practices (GMP).
  • GMP Good Manufacturing Practices
  • the initial derivation or production of human embryonic stem cells is also conducted in accordance with Good Manufacturing Practices (GMP).
  • the cells may be tested at one or more points throughout the differentiation protocol to ensure, for example, that there is no viral, bacterial, or fungal infection or contamination in the cells or culture medium.
  • the human embryonic stem cells used as starting material may be tested to ensure that there is no viral, bacterial, or fungal infection or contamination.
  • the production of differentiated RPE cells or mature differentiated RPE cells is scaled up for commercial use.
  • the method can be used to produce at least 1 ⁇ 10 5 , 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , or 1 ⁇ 10 10 RPE cells.
  • the invention provides a method of conducting a pharmaceutical business, comprising the step of providing RPE cell preparations that are homozygous for at least one histocompatibility antigen, wherein cells are chosen from a bank of such cells comprising a library of RPE cells that can be expanded by the methods disclosed herein, wherein each RPE cell preparation is hemizygous or homozygous for at least one MHC allele present in the human population, and wherein said bank of RPE cells comprises cells that are each hemizygous or homozygous for a different set of MHC alleles relative to the other members in the bank of cells.
  • Methods of conducting a pharmaceutical business may also comprise establishing a distribution system for distributing the preparation for sale or may include establishing a sales group for marketing the pharmaceutical preparation.
  • RPE cells of the present invention relate to the use of the RPE cells of the present invention as a research tool in settings such as a pharmaceutical, chemical, or biotechnology company, a hospital, or an academic or research institution.
  • uses include the use of RPE cells differentiated from embryonic stem cells in screening assays to identify, for example, agents that can be used to promote RPE survival in vitro or in vivo, or that can be used to promote RPE maturation.
  • Identified agents can be studied in vitro or in animal models to evaluate, for example, their potential use alone or in combination with RPE cells.
  • the present invention also includes methods of obtaining human ES cells from a patient and then generating and expanding RPE cells derived from the ES cells. These RPE cells may be stored. In addition, these RPE cells may be used to treat the patient from which the ES were obtained or a relative of that patient.
  • the present invention also relates to solutions of RPE cells that are suitable for such applications.
  • the present invention accordingly relates to solutions of RPE cells that are suitable for injection into a patient.
  • Such solutions may comprise cells formulated in a physiologically acceptable liquid (e.g., normal saline, buffered saline, or a balanced salt solution).
  • the number of cells in the solution may be at least about 10 2 and less than about 10 9 cells.
  • the number of cells in the solution may range from about 10 1 , 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , or 10 8 to about 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , or 10 9 , where the upper and lower limits are selected independently, except that the lower limit is always less than the upper limit.
  • the cells may be administered in a single or in multiple administrations.
  • the present invention provides a cryopreserved preparation of RPE cells, wherein said cryopreserved preparation comprises at least about 10 1 , 10 2 , 5 ⁇ 10 2 , 10 3 , 5 ⁇ 10 3 , 10 4 , 5 ⁇ 10 4 , 10 5 , 5 ⁇ 10 5 , or 10 6 .
  • Cryopreserved preparations may further comprise at least about 5 ⁇ 10 6 , 10 7 , 5 ⁇ 10 7 , 10 8 , 15 ⁇ 0 8 , 10 9 , 5 ⁇ 10 9 , or 10 10 cells.
  • methods of cryopreserving RPE cells RPE cells may be cryopreserved immediately following differentiation, following in vitro maturation, or after some period of time in culture.
  • the RPE cells in the preparations may comprise a mixture of differentiated RPE cells and mature RPE cells.
  • pluripotent stem cells includes embryonic stem cells, embryo-derived stem cells, and induced pluripotent stem cells, regardless of the method by which the pluripotent stem cells are derived.
  • Pluripotent stem cells are defined functionally as stem cells that: (a) are capable of inducing teratomas when transplanted in immunodeficient (SCID) mice; (b) are capable of differentiating to cell types of all three germ layers (e.g., can differentiate to ectodermal, mesodermal, and endodermal cell types); and (c) express one or more markers of embryonic stem cells (e.g., express Oct 4, alkaline phosphatase, SSEA-3 surface antigen, SSEA-4 surface antigen, nanog, TRA-1-60, TRA-1-81, SOX2, REX1, etc).
  • SCID immunodeficient
  • Exemplary pluripotent stem cells can be generated using, for example, methods known in the art.
  • Exemplary pluripotent stem cells include embryonic stem cells derived from the ICM of blastocyst stage embryos, as well as embryonic stem cells derived from one or more blastomeres of a cleavage stage or morula stage embryo (optionally without destroying the remainder of the embryo).
  • embryonic stem cells can be generated from embryonic material produced by fertilization or by asexual means, including somatic cell nuclear transfer (SCNT), parthenogenesis, cellular reprogramming, and androgenesis.
  • SCNT somatic cell nuclear transfer
  • pluripotent stem cells include induced pluripotent stem cells (iPS cells) generated by reprogramming a somatic cell by expressing or inducing the expression of a combination of factors (herein referred to as reprogramming factors).
  • iPS cells 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, a combination of Oct4 (sometimes referred to as Oct 3/4), Sox2, c-Myc, and Klf4.
  • factors that can be used to reprogram somatic cells to pluripotent stem cells include, for example, a combination of Oct 4, Sox2, Nanog, and Lin28.
  • somatic cells are reprogrammed by expressing at least 2 reprogramming factors, at least three reprogramming factors, or four reprogramming factors.
  • additional reprogramming factors are identified and used alone or in combination with one or more known reprogramming factors to reprogram a somatic cell to a pluripotent stem cell.
  • Embryonic stem cells are one example of pluripotent stem cells.
  • Another example are induced pluripotent stem (iPS) cells.
  • the pluripotent stem cell is an embryonic stem cell or embryo-derived cell. In other embodiments, the pluripotent stem cell is an induced pluripotent stem cell. In certain embodiments, the pluripotent stem cell is an induced pluripotent stem cell produced by expressing or inducing the expression of one or more reprogramming factors in a somatic cell. In certain embodiments, the somatic cell is a fibroblast, such as a dermal fibroblast, synovial fibroblast, or lung fibroblast. In other embodiments, the somatic cell is not a fibroblast, but rather is a non-fibroblastic somatic cell.
  • the somatic cell is reprogrammed by expressing at least two reprogramming factors, at least three reprogramming factors, or four reprogramming factors. In other embodiments, the somatic cell is reprogrammed by expressing at least four, at least five, or at least six reprogramming factors. In certain embodiments, the reprogramming factors are selected from Oct 3/4, Sox2, Nanog, Lin28, c-Myc, and Klf4. In other embodiments, the set of reprogramming factors expressed includes at least one, at least two, at least three, or at least four of the foregoing list of reprogramming factors, and optionally includes one or more other reprogramming factors.
  • expression of at least one, at least two, at least three, or at least four of the foregoing or other reprogramming factors is induced by contacting the somatic cells with one or more agents, such as a small organic molecule agents, that induce expression of one or more reprogramming factors.
  • the somatic cell is reprogramming using a combinatorial approach wherein one or more reprogramming factor is expressed (e.g., using a viral vector, plasmid, and the like) and the expression of one or more reprogramming factor is induced (e.g., using a small organic molecule.).
  • reprogramming factors are expressed in the somatic cell by infection using a viral vector, such as a retroviral vector or a lentiviral vector.
  • reprogramming factors are expressed in the somatic cell using a non-integrative vector, such as an episomal plasmid.
  • the factors can be expressed in the cells using electroporation, transfection, or transformation of the somatic cells with the vectors.
  • the pluripotent stem cells are generated from somatic cells, and the somatic cells are selected from embryonic, fetal, neonatal, juvenile, or adult cells.
  • somatic cells are infected, transfected, or otherwise transduced with expression vectors expressing reprogramming factors.
  • expression vectors expressing reprogramming factors.
  • expression of four factors Oct3/4, Sox2, c-myc, and Klf4 using integrative viral vectors was sufficient to reprogram a somatic cell.
  • expression of four factors Oct3/4, Sox2, Nanog, and Lin28 using integrative viral vectors was sufficient to reprogram a somatic cell.
  • expression (or induction of expression) of fewer factors or other reprogramming factors may also be sufficient.
  • the use of integrative vectors is only one mechanism for expressing reprogramming factors in the cells. Other methods including, for example, the use of non-integrative vectors can be used.
  • expression of at least one, at least two, at least three, or at least four of the foregoing or other reprogramming factors is induced by contacting the somatic cells with one or more agents, such as a small organic molecule agents, that induce expression of one or more reprogramming factors.
  • the somatic cell is reprogramming using a combinatorial approach wherein one or more reprogramming factor is expressed (e.g., using a viral vector, plasmid, and the like) and the expression of one or more reprogramming factor is induced (e.g., using a small organic molecule.).
  • the cells are cultured. Over time, cells with ES characteristics appear in the culture dish. The cells can be picked and subcultured based on, for example, ES morphology, or based on expression of a selectable or detectable marker. The cells are cultured to produce a culture of cells that look like ES cells. These cells are putative iPS cells.
  • the cells can be tested in one or more assays of pluripotency.
  • the cells can be tested for expression of ES cell markers; the cells can be evaluated for ability to produce teratomas when transplanted into SCID mice; the cells can be evaluated for ability to differentiate to produce cell types of all three germ layers.
  • pluripotent iPS cells Once pluripotent iPS cells are obtained (either freshly derived or from a bank or stock of previously derived cells), such cells can be used to make RPE cells.
  • the making of iPS cells is an initial step in the production of RPE cells. In other embodiments, previously derived iPS cells are used. In certain embodiments, iPS cells are specifically generated using material from a particular patient or matched donor with the goal of generating tissue-matched RPE cells. In certain embodiments, the iPS cells are universal donor cells that are not substantially immunogenic.
  • the retinal pigmented epithelium develops from the neuroectoderm and is located adjacent to the neural retina and choroid, providing a barrier between the vascular system and the retina.
  • the data provided herein indicates that RPE cells are genetically and functionally distinguished from surrounding photoreceptors after terminal differentiation, although the cells may share a common progenitor.
  • PAX6 Paired-box 6
  • PAX6 acts synergistically with PAX2 to terminally differentiate mature RPE via the coordination of Mit-F and Otx2 to transcribe RPE-specific genes such as Tyrosinase (Tyr), and downstream targets such as RPE-65, Bestrophin, CRALBP, and PEDF.
  • the RPE-specific signature of mRNA and protein expression was not only unique from hES cells, but also from fetal RPE and ARPE-19 cells.
  • the RPE cells described herein expressed multiple genes that were not expressed in hES cells, fetal RPE cells, or ARPE-19 cells ( FIGS. 3 , 4 , and 6 ).
  • the unique expression of mRNA and proteins in the RPE cells of the invention constitutes a set of markers that make these RPE cells distinct from cells in the art, such as hES cells, ARPE-19 cells, and fetal RPE cells.
  • hES cells were thawed and placed into suspension culture on Lo-bind Nunclon Petri dishes in MDBK-Growth Medium (Sigma—SAFC Biosciences) or OptimPro SFM (Invitrogen) supplemented with L-Glutamine, Penicillin/Streptomycin, and B-27 supplement.
  • the hES cells had been previously derived from single blastomeres biopsied from early cleavage stage human embryos. The remainder of the human embryo was not destroyed. Two hES cell line derived from single blastomeres were used—MAO1 and MAO9. The cells were cultured for 7-14 days as embryoid bodies (EBs).
  • EBs embryoid bodies
  • the EBs were plated onto tissue culture plates coated with gelatin from porcine skin.
  • the EBs were grown as adherent cultures for an additional 14-28 days in MDBK-Growth Medium or OptimPro SFM supplemented with L-Glutamine, and Penicillin/Streptomycin, without B-27 supplement.
  • RPE cells From amongst the cells in the adherent culture of EBs, RPE cells become visible and are recognized by their cobblestone cellular morphology and emergence of pigmentation.
  • Pigmented clumps are transferred with a stem cell cutting tool (Swemed-Vitrolife) to a well of a 6-well plate containing 3 ml of MEF media. After all clumps have been picked up, the suspension of pigmented cells is transferred to a 15 ml conical tube containing 7 ml of MEF medium and centrifuged at 1000 rpm for five minutes. The supernatant is removed. 5 ml of a 1:1 mixture of 0.25% trypsin and cell dissociation buffer is added to the cells. The cells are incubated for 10 minutes at 37° C. The cells are dispersed by pipetting in a 5 ml pipette until few clumps are remaining.
  • a stem cell cutting tool Stemed-Vitrolife
  • the culture of RPE cells was expanded by continued culture in EGM-2 medium.
  • the cells were passaged, as necessary, at a 1:3 to 1:6 ratio using a 1:1 mixture of 0.25% trypsin EDTA and Cell Dissociation Buffer.
  • the cells were grown to near confluence in EGM-2. The medium was then changed to MDBK-MM (SAFC Biosciences) to help further promote maturation of the RPE cells.
  • MDBK-MM SAFC Biosciences
  • qPCR was developed to provide a quantitative and relative measurement of the abundance of cell type-specific mRNA transcripts of interest in the RPE differentiation process. qPCR was used to determine genes that are uniquely expressed in human embryonic stem cells, in neuroretinal cells during eye development, and in RPE cells differentiated from human embryonic stem cells. The genes for each cell type are listed below in Table 1.
  • hES-specific genes included Oct-4 (POU5F1), Nanog, Rex-1, TDGF-1, SOX-2, and DPPA-2.
  • Genes specific to neural ectoderm/neural retina include CHX10, NCAM, Nestin, and Beta-Tubulin.
  • RPE cells differentiated from human embryonic stem cells were found to uniquely express PAX-6, PAX-2, RPE-65, PEDF, CRALBP, Bestrophin, MitF, Otx-2, and Tyr by qPCR measurement.
  • hES-specific genes are grossly downregulated (near 1000-fold) in RPE cells derived from hES, whereas genes specific for RPE and neuroectoderm are vastly upregulated (approximately 100-fold) in RPE cells derived from hES.
  • RPE cells derived from hES cells did not express the hES-specific proteins Oct-4, Nanog, and Rex-1, whereas they expressed RPE65, CRALBP, PEDF, Bestrophin, PAX6, and Otx2. These proteins are therefore prominent markers of RPE cells differentiated from hES cells. By contrast, APRE-19 cells showed an inconclusive pattern of proteomic marker expression.
  • hES cell-differentiated hRPE in vitro undergo significant morphological events in culture during the expansion phase.
  • Single-cell suspensions plated in thin cultures depigment and cells increase in surface area.
  • hRPE cells maintain this morphology during expansion when the cells are rapidly dividing.
  • RPE take on their characteristic phenotypic hexagonal shape and increase pigmentation level by accumulating melanin and lipofuscin.
  • FIG. 7 depicts a principle components analysis plot scattering of each sample based upon the minimal number of genes accounting for variability amongst each sample.
  • Component 1 representing 69% of the variability represents the cell type, whereas Component 2, represents the cell line (i.e., genetic variability).
  • Component 2 represents the cell line (i.e., genetic variability).
  • a near-linear scatter of gene expression profiles characterizes the developmental ontogeny of hRPE derived from hES cells.
  • VMD2 is characterized by typical “egg- yolk” macular lesions due to abnormal accumulation of lipofuscin within and beneath the retinal pigmented epithelium cells. Progression of the disease leads to destruction of the retinal pigmented epithelium and vision loss. Defects in BEST1 are a cause of adult-onset vitelliform macular dystrophy (AVMD). AVMD is a rare autosomal dominant disorder with incomplete penetrance and highly variable expression. Patients usually become symptomatic in the fourth or fifth decade of life with a protracted disease of decreased visual acuity. CLUL1 clusterin-like 1 retinal Associated strongly with cone photoreceptors and (retinal) (retinal) development appears in different tissues throughout retinal development.
  • CRYAA crystallin, eye Crystallins are the dominant structural components of the alpha A development vertebrate eye lens. May contribute to the transparency and refractive index of the lens. Defects in CRYAA are the cause of zonular central nuclear cataract one of a considerable number of phenotypically and genotypically distinct forms of autosomal dominant cataract.
  • This congenital cataract is a common major abnormality of the eye that frequently causes blindness in infants. Crystallins do not turn over as the lens ages, providing ample opportunity for post-translational modifications or oxidations. These modifications may change crystallin solubility properties and favor senile cataract.
  • CRYBA1 crystallin, eye Crystallins are the dominant structural components of the beta A1 development vertebrate eye lens. Crystallins do not turn over as the lens ages, providing ample opportunity for post- translational modifications or oxidations. These modifications may change crystallin solubility properties and favor senile cataract.
  • CRYBA2 crystallin, eye Crystallins are the dominant structural components of the beta A2 development vertebrate eye lens.
  • Crystallins do not turn over as the lens ages, providing ample opportunity for post- translational modifications or oxidations. These modifications may change crystallin solubility properties and favor senile cataract.
  • CRYBA4 crystallin, eye Crystallins are the dominant structural components of the beta A4 development vertebrate eye lens. Defects in CRYBA4 are the cause of lamellar cataract 2. Cataracts are a leading cause of blindness worldwide, affecting all societies. A significant proportion of cases are genetically determined. More than 15 genes for cataracts have been identified, of which the crystallin genes are the most commonly mutated. Lamellar cataract 2 is an autosomal dominant congenital cataract.
  • CRYBA4 Defects in CRYBA4 are a cause of isolated microphthalmia with cataract 4 (MCOPCT4).
  • Microphtalmia consists of a development defect causing moderate or severe reduction in size of the eye. Opacities of the cornea and lens, scaring of the retina and choroid, and other abnormalities like cataract may also be present Crystallins do not turn over as the lens ages, providing ample opportunity for post-translational modifications or oxidations. These modifications may change crystallin solubility properties and favor senile cataract.
  • CRYBB1 crystallin, eye Crystallins are the dominant structural components of the beta B1 development vertebrate eye lens.
  • CRYBB2 crystallin, eye Crystallins are the dominant structural components of the beta B2 development vertebrate eye lens.
  • CCA2 congenital cerulean cataract 2
  • CCA2 congenital cerulean cataract blue dot type 2.
  • CCA2 is a form of autosomal dominant congenital cataract (ADCC). Cerulean cataracts have peripheral bluish and white opacifications in concentric layers with occasional central lesions arranged radially. Although the opacities may be observed during fetal development and childhood, usually visual acuity is only mildly reduced until adulthood, when lens extraction is generally necessary.
  • Defects in CRYBB2 are the cause of sutural cataract with punctate and cerulean opacities (CSPC). The phenotype associated with this form of autosomal dominant congenital cataract differed from all other forms of cataract reported.
  • CRYBB2 Coppock-like cataract
  • Crystallins do not turn over as the lens ages, providing ample opportunity for post-translational modifications or oxidations.
  • CRYBB3 crystallin, eye Crystallins are the dominant structural components of the beta B3 development vertebrate eye lens.
  • Defects in CRYBB3 are the cause of autosomal recessive congenital nuclear cataract 2(CATCN2); a form of nonsyndromic congenital cataract.
  • Non-syndromic congenital cataracts vary markedly in severity and morphology, affecting the nuclear, cortical, polar, or subcapsular parts of the lens or, in severe cases, the entire lens, with a variety of types of opacity.
  • DCT/TYRP2 dopachrome pigmented Tyrosine metabolism and Melanin biosynthesis.
  • tautomerase cells dopachrome delta- isomerase, tyrosine- related protein 2
  • LHX2 LIM development/ Transcriptional regulatory protein involved in the control homeobox 2 differentiation of cell differentiation in developing lymphoid and neural cell types.
  • LIM2 lens intrinsic eye Present in the thicker 16-17 nm junctions of mammalian membrane development lens fiber cells, where it may contribute to cell junctional protein 2, organization. Acts as a receptor for calmodulin. May play 19 kDa an important role in both lens development and cataractogenesis.
  • MITF microphthalmia- RPE Transcription factor for tyrosinase and tyrosinase- related associated development protein 1. Binds to a symmetrical DNA sequence (E- transcription boxes) (5'-CACGTG-3') found in the tyrosinase factor promoter.
  • OCA2 oculocutaneous albinism type II
  • OCA2 is an autosomal recessive form of albinism, a disorder of pigmentation in the skin, hair, and eyes.
  • the phenotype of patients with OCA2 is typically somewhat less severe than in those with tyrosinase- deficient OCA1.
  • AROA autosomal recessive ocular albinism
  • OCA2 is localized to chromosome 15 at 15q11.2- q12 OPN3 opsin 3 eye May play a role in encephalic photoreception. Strongly development expressed in brain. Highly expressed in the preoptic area and paraventricular nucleus of the hypothalamus. Shows highly patterned expression in other regions of the brain, being enriched in selected regions of the cerebral cortex, cerebellar Purkinje cells, a subset of striatal neurons, selected thalamic nuclei, and a subset of interneurons in the ventral horn of the spinal cord. OPN5 opsin 5 eye Associated with visual perception and phototransduction.
  • OTX2 orthodenticle retinal Probably plays a role in the development of the brain and homolog 2 development the sense organs.
  • Defects in OTX2 are the cause of ( Drosophila ) syndromic microphthalmia 5 (MCOPS5).
  • MCOPS5 syndromic microphthalmia 5
  • Microphthalmia is a clinically heterogeneous disorder of eye formation, ranging from small size of a single eye to complete bilateral absence of ocular tissues. Up to 80% of cases of microphthalia occur in association with syndromes that include non-ocular abnormalities such as cardiac defects, facial clefts, microcephaly and hydrocephaly.
  • MCOPS5 patients manifest unilateral or bilateral microphthalmia/clinical anophthalmia and variable additional features including coloboma, microcomea, cataract, retinal dystrophy, hypoplasia or agenesis of the optic nerve, agenesis of the corpus callosum, developmental delay, joint laxity, hypotonia, and seizures.
  • PAX6 paired box RPE Transcription factor with important functions in the gene 6 development development of the eye, nose, central nervous system and (aniridia, pancreas. Required for the differentiation of pancreatic keratitis) islet alpha cells (By similarity). Competes with PAX4 in binding to a common element in the glucagon, insulin and somatostatin promoters (By similarity).
  • Isoform 5a appears to function as a molecular switch that specifies target genes. Defects in Pax6 results in a number of eye defects and malformations.
  • PHC2 polyhomeotic- development/ Component of the Polycomb group (PcG) multiprotein like 2 differentiation PRC1 complex a complex required to maintain the ( Drosophila ) transcriptionally repressive state of many genes, including Hox genes, throughout development.
  • PcG PRC1 complex acts via chromatin remodeling and modification of histones; it mediates monoubiquitination of histone H2A ‘Lys-119’, rendering chromatin heritably changed in its expressibility.
  • PKNOX2 PBX/knotted 1 development/ Known to be involved in development and may, along homeobox 2 differentiation with MEIS, control Pax6.
  • PRKCA protein kinase cellular Very important for cellular signaling pathways such as C, alpha signalling the MAPK, Wnt, PI3, VEGF and Calcium pathways.
  • PROX1 prospero- eye May play a fundamental role in early development of related development CNS. May regulate gene expression and development of homeobox 1 postmitotic undifferentiated young neurons. Highly expressed in lens, retina, and pancreas.
  • PRRX1 paired related development/ Necessary for development. Transcription coactivator, homeobox 1 differentiation enhancing the DNA-binding activity of serum response factor.
  • RAI1 retinoic acid development/ May function as a transcriptional regulator. Regulates induced 1 differentiation transcription through chromatin remodeling by interacting with other proteins in chromatin as well as proteins in the basic transcriptional machinery. May be important for embryonic and postnatal development. May be involved in neuronal differentiation.
  • RARA retinoic acid development/ This is a receptor for retinoic acid. This metabolite has receptor, alpha differentiation profound effects on vertebrate development. This receptor controls cell function by directly regulating gene expression.
  • RARRES1 retinoic acid development/ Associated with differentiation and control of cell receptor differentiation proliferation May be a growth regulator that mediates responder some of the growth suppressive effects of (tazarotene retinoids. induced) 1 RAX retina and eye Plays a critical role in eye formation by regulating the anterior neural development initial specification of retinal cells and/or their fold homeobox subsequent proliferation. Binds to the photoreceptor conserved element-I (PCE-1/Ret 1) in the photoreceptor cell-specific arrestin promoter. RB1 retinoblastoma development/ An important regulator of other genes and cell growth. 1 (including differentiation Defects in RB1 are the cause of childhood cancer osteosarcoma) retinoblastoma (RB).
  • RDH5 retinol RPE retinol dehydrogenase 5,11-cis expressed in retinal dehydrogenase development pigmented epithelium, formerly RDH1.
  • Stereospecific 5(11-cis/9-cis) 11-cis retinol dehydrogenase which catalyzes the final step in the biosynthesis of 11-cis retinaldehyde, the universal chromophore of visual pigments.
  • Abundant in the retinal pigmented epithelium. Defects in RDH5 are a cause of fundus albipunctatus (FA).
  • FA is a rare form of stationary night blindness characterized by a delay in the regeneration of cone and rod photopigments.
  • RGR retinal G RPE Preferentially expressed at high levels in the retinal protein development pigmented epithelium (RPE) and Mueller cells of the coupled neural retina.
  • RLBP1/ retinaldehyde RPE Carries 11-cis-retinol and 11-cis-retinaldehyde as CRALBP1 binding development endogenous ligands and may be a functional component protein 1 of the visual cycle.
  • Defects in RLBP1 are a cause of autosomal recessive retinitis pigmentosa (arRP).
  • Retinitis pigmentosa (RP) leads to degeneration of retinal photoreceptor cells.
  • Defects in RLBP1 are the cause of Bothnia retinal dystrophy, also known as Vasterbotten dystrophy. It is another form of autosomal recessive retinitis pigmentosa.
  • NFRCD Newfoundland rod- cone dystrophy
  • RRH retinal pigment RPE Found only in the eye, where it is localized to the retinal epithelium- development pigment epithelium (RPE). In the RPE, it is localized to derived the microvilli that surround the photoreceptor outer rhodopsin segments. May play a role in rpe physiology either by homolog detecting light directly or by monitoring the concentration of retinoids or other photoreceptor-derived compounds.
  • RXRB retinoid X development/ Nuclear hormone receptor RXRB retinoid X development/ Nuclear hormone receptor.
  • Neurotrophic protein induces extensive inhibitor, clade neuronal differentiation in retinoblastoma cells.
  • F alpha-2 antiplasmin, pigment epithelium derived factor
  • SIX3 sine oculis eye Expressed during eye development in midline forebrain homeobox development and in anterior region of the neural plate especially inner homolog 3 retina and later in ganglion cells and in cells of the inner ( Drosophila ) nuclear layer, involved in regulation of eye development.
  • SOX10 SRY sex development/ Transcription factor that seems to function synergistically determining differentiation with other development associated proteins.
  • Can region Y confer cell specificity to the function of other 10 transcription factors in developing and mature glia.
  • SOX5 SRY (sex development/ Expression is associated with craniofacial, skeletal and determining differentiation cartilage development and is highly expressed in brain, region Y)-box testis and various tissues.
  • 5 SOX6 SRY (sex development/ Expression is associated with craniofacial, skeletal and determining differentiation cartilage development and is highly expressed in brain, region Y)-box testis and various tissues.
  • 6 SOX8 SRY (sex development/ May play a role in central nervous system, limb and determining differentiation facial development. region Y)-box 8 SOX9 SRY (sex development/ Plays an important role in the normal development.
  • TIMP3 Sorsby fundus dystrophy
  • SFD Sorsby fundus dystrophy
  • SFD is a rare autosomal dominant macular disorder with an age of onset in the fourth decade. It is characterized by loss of central vision from subretinal neovascularization and atrophy of the ocular tissues.
  • TTR transthyretin prealbumin, Thyroid hormone-binding protein. Probably transports amyloidosis type I) thyroxine from the bloodstream to the brain.
  • Defects in TTR are the cause of amyloidosis VII; also known as leptomeningeal amyloidosis or meningocerebrovascular amyloidosis.
  • Leptomeningeal amyloidosis is distinct from other forms of transthyretin amyloidosis in that it exhibits primary involvement of the central nervous system. Neuropathologic examination shows amyloid in the walls of leptomeningeal vessels, in pia arachnoid, and subpial deposits. Some patients also develop vitreous amyloid deposition that leads to visual impairment (oculoleptomeningeal amyloidosis).
  • TYR tyrosinase pigmented This is a copper-containing oxidase that functions in the (oculocutaneous cells formation of pigments such as melanins and other albinism polyphenolic compounds.
  • OCA-IA oculocutaneous albinism type IA
  • OCA-IA also known as tyrosinase negative oculocutaneous albinism
  • OCA-I is an autosomal recessive disorder characterized by absence of pigment in hair, skin and eyes.
  • OCA-I is divided into 2 types: type IA, characterized by complete lack of tyrosinase activity due to production of an inactive enzyme, and type IB characterized by reduced activity of tyrosinase.
  • OCA-IA patients presents with the life-long absence of melanin pigment after birth and manifest increased sensitivity to ultraviolet radiation and to predisposition to skin cancer defects in TYR are the cause of oculocutaneous albinism type IB (OCA-IB); also known as albinism yellow mutant type.
  • OCA-IB patients have white hair at birth that rapidly turns yellow or blond.
  • TYRP1 tyrosinase- pigmented Specific expression in Pigment cells. Oxidation of 5,6- related protein cells dihydroxyindole-2-carboxylic acid (DHICA) into indole- 1 5,6-quinone-2-carboxylic acid. May regulate or influence the type of melanin synthesized.
  • DHICA dihydroxyindole-2-carboxylic acid
  • Defects in TYRP1 are the cause of rufous oculocutaneous albinism (ROCA).
  • ROCA occurs in blacks and is characterized by bright copper-red coloration of the skin and hair and dilution of the color of the iris.
  • Defects in TYRP1 are the cause of oculocutaneous albinism type III (OCA-III); also known as OCA3.
  • OCA-III is a form of albinism with only moderate reduction of pigment. Individuals with OCA-III are recognized by their reddish skin and hair color.
  • CECR2 cat eye Part of the CERF (CECR2-containing-remodeling syndrome factor) complex which facilitates the perturbation of chromosome chromatin structure in an ATP-dependent manner. May region, be involved through its interaction with LRPPRC in the candidate 2 integration of cytoskeletal network with vesicular trafficking, nucleocytosolic shuttling, transcription, chromosome remodeling and cytokinesis. Developmental disorders are associated with the duplication of the gene.
  • DCAMKL1 doublecortin Embryonic Probable kinase that may be involved in a calcium- and CaM development signaling pathway controlling neuronal migration in kinase-like 1 the developing brain.
  • DPPA2 developmental ES cells May play a role in maintaining cell pluripotentiality.
  • pluripotency associated 2 DPPA3 developmental ES cells May play a role in maintaining cell pluripotentiality.
  • DPPA4 developmental ES cells May indicate cell pluripotentiality.
  • pluripotency associated 4 DPPA5/Esg1 developmental ES cells Embryonic stem cell marker.
  • pluripotency associated 5/Embryonic stem cellspecific gene 1 FOXD3 forkhead box Pluripotence Required for maintenance of pluripotent cells in the D3 pre-implantation and peri-implantation stages of embryogenesis.
  • L1TD1ECAT11 LINE-1 type ES cells Embryonic stem cell marker.
  • transposase domain containing 1/ES cell associated transcript 11 NANOG Nanog ES cells Embryonic stem cell marker.
  • Transcription regulator homeobox involved in inner cell mass and embryonic stem (ES) cells proliferation and self-renewal. Imposes pluripotency on ES cells and prevents their differentiation towards extraembryonic endoderm and trophectoderm lineages.
  • NCAM1 neural cell neuroprogenitors This protein is a cell adhesion molecule involved in adhesion neuron-neuron adhesion, neurite fasciculation, molecule 1 outgrowth of neurites. etc.
  • NES/Nestin nestin ES cells Neuralprogenitor cells. NODAL nodal Embryonic Essential for mesoderm formation and axial patterning development during embryonic development.
  • NR5A2/FTF nuclear Embryonic May contribute to the development and regulation of receptor development liver and pancreas-specific genes and play important subfamily 5, roles in embryonic development. group A, member 2 POU5F1/ POU domain, ES cells Embryonic stem cell marker. Indicator of “Stemness”.
  • Oct-3/4 class 5 Transcription factor that binds to the octamer motif (5′′- transcription ATTTGCAT-3′′). Prime candidate for an early factor 1 developmental control gene.
  • SOX17 SRY (sex Inhibitor of Negative regulator of the Wnt signalling pathway.
  • SOX2 SRY (sex ES cells Indicator of “Stemness”. Expressed in inner cell mass, determining primitive ectoderm and developing CNS. region Y)-box 2 TBX3 T-box 3 (ulnar Embryonic Transcriptional represser involved in developmental mammary development processes.
  • Murine T-box gene Tbx3 syndrome (T, brachyury)homolog, putative transcription factor, pairing with TBX5, homolog to Drosophila optomotor-blind gene (omb), involved in optic lobe and wing development, involved in developmental regulation, expressed in anterior and posterior mouse limb buds, widely expressed in adults TDGF1/ teratocarcinoma- ES cells Indicator of “Stemness”.
  • Tbx3 syndrome T, brachyury
  • TBX5 homolog to Drosophila optomotor-blind gene (omb)
  • omb Drosophila optomotor-blind gene
  • This protein is a protein tyrosine-kinase kinase, Endothelial transmembrane receptor for angiopoietin 1. It may endothelial progenitors constitute the earliest mammalian endothelial cell (venous lineage marker. Probably regulates endothelial cell malformations, proliferation, differentiation and guides the proper multiple patterning of endothelial cells during blood vessel cutaneous and formation mucosal) TUBB2A, tubulin, beta neuroprogenitors Tubulin is the major constituent of microtubules.
  • TUBB2B 2A tubulin
  • Tubulin binds two moles of GTP, one at an exchangeable site beta 2B on the beta chain and one at a non-exchangeable site on the alpha-chain.
  • TUBB2A, tubulin, beta neuroprogenitors Tubulin is the major constituent of microtubules.
  • tubulin is the major constituent of microtubules. It binds two moles of GTP, one at an exchangeable site on the beta chain and one at a non-exchangeable site on the alpha-chain. Often associated with the formation of gap junctions in neural cells. TWIST1 twist homolog Inhibitor of Probable transcription factor, which negatively 1 differentiation regulates cellular determination and differentiation. UTF1 undifferentiated ES cells Embryonic stem cell marker. Acts as a transcriptional embryonic cell coactivator of ATF2.
  • transcription factor 1 VSNL1 visinin-like 1 Inhibitor of Regulates the inhibition of rhodopsin phosphorylation.
  • rhodopsin ZFP42/Rex-1 zinc finger ES cells Embryonic Stem cell marker. protein 42
  • human RPE cells can be reliably differentiated and expanded from human ES cells under well-defined and reproducible conditions—representing an inexhaustible source of cells for patients with retinal degenerative disorders.
  • concentration of these cells would not be limited by availability, but rather could be titrated to the precise clinical requirements of the individual. Repeated infusion or transplantation of the same cell population over the lifetime of the patient would also be possible if deemed necessary by the physician.
  • the ability to create banks of matching or reduced-complexity HLA hES lines from which RPE cells could be produced could potentially reduce or eliminate the need for immunosuppressive drugs and/or immunomodulatory protocols altogether.
  • RPE cells differentiated by the methods described herein express multiple genes that are not expressed by hES cells, fetal RPE cells, or ARPE-19 cells.
  • the unique molecular fingerprint of mRNA and protein expression in the ES-cell derived RPE cells of the invention constitutes a set of markers, such as RPE-65, Bestrophin, PEDF, CRABLP, Otx2, Mit-F, PAX6 and PAX2, that make these RPE cells distinct from cells in the art, such as hES cells, ARPE-19 cells, and fetal RPE cells.
  • Certain retinal diseases are characterized by degeneration of the retinal pigment epithelium (RPE) which in turn results in photoreceptor loss.
  • RPE retinal pigment epithelium
  • Examples include Stargardt's macular dystrophy in humans and the genetically-determined dystrophy in the Royal College of Surgeons (RCS) rat.
  • Such a process may also play a role in macular degeneration, affecting more than 10 million people in the US alone.
  • Superior colliculus recordings at P94 also showed much lower luminance threshold responses in RPE cell-injected eyes with some individual recordings within the normal range. Histological studies showed donor cells disposed as a semi-continuous, pigmented cell layer immediately internal to endogenous, host RPE. The donor RPE cells were positive for RPE65 and bestrophin, indicating that the transplanted cells were RPE cells and that the cell maintain their cell fate following transplantation.
  • transplanted animals maintained photoreceptor thickness in comparison to control animals.
  • the photoreceptors in RPE treatment animals were 4-5 cells thick in the rescued area compared with only a single layer in sham and untreated controls.
  • RPE cells derived from embryonic stem cells and manufactured under GMP-compliant conditions survive after transplantation to the subretinal space of RCS rats, do not migrate into the retina and continue to express molecules characteristic of RPE. Most importantly, they achieve significant rescue of visual function in a dose dependent fashion in an animal model of photoreceptor degeneration.
  • the data further suggest that these cells may be effective in limiting and/or reversing the deterioration of vision that accompanies RPE-driven photoreceptor degeneration in human disease.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Ophthalmology & Optometry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Psychology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Materials For Medical Uses (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US12/682,712 2007-10-12 2008-10-10 Methods of Producing RPE Cells and Compositions of RPE Cells Abandoned US20110274662A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/682,712 US20110274662A1 (en) 2007-10-12 2008-10-10 Methods of Producing RPE Cells and Compositions of RPE Cells

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US99876607P 2007-10-12 2007-10-12
US99866807P 2007-10-12 2007-10-12
US991108P 2008-01-02 2008-01-02
US990808P 2008-01-02 2008-01-02
US12/682,712 US20110274662A1 (en) 2007-10-12 2008-10-10 Methods of Producing RPE Cells and Compositions of RPE Cells
PCT/US2008/011669 WO2009051671A1 (en) 2007-10-12 2008-10-10 Improved methods of producing rpe cells and compositions of rpe cells

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US61009911 Continuation-In-Part 2008-01-02
PCT/US2008/011669 A-371-Of-International WO2009051671A1 (en) 2007-10-12 2008-10-10 Improved methods of producing rpe cells and compositions of rpe cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/254,833 Continuation US10077424B2 (en) 2007-10-12 2014-04-16 Methods of producing RPE cells and compositions of RPE cells

Publications (1)

Publication Number Publication Date
US20110274662A1 true US20110274662A1 (en) 2011-11-10

Family

ID=40567677

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/682,712 Abandoned US20110274662A1 (en) 2007-10-12 2008-10-10 Methods of Producing RPE Cells and Compositions of RPE Cells
US14/254,833 Active US10077424B2 (en) 2007-10-12 2014-04-16 Methods of producing RPE cells and compositions of RPE cells
US16/113,717 Abandoned US20190062703A1 (en) 2007-10-12 2018-08-27 Methods of producing rpe cells and compositions of rpe cells
US17/216,172 Pending US20220049217A1 (en) 2007-10-12 2021-03-29 Methods of producing rpe cells and compositions of rpe cells

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/254,833 Active US10077424B2 (en) 2007-10-12 2014-04-16 Methods of producing RPE cells and compositions of RPE cells
US16/113,717 Abandoned US20190062703A1 (en) 2007-10-12 2018-08-27 Methods of producing rpe cells and compositions of rpe cells
US17/216,172 Pending US20220049217A1 (en) 2007-10-12 2021-03-29 Methods of producing rpe cells and compositions of rpe cells

Country Status (13)

Country Link
US (4) US20110274662A1 (he)
EP (2) EP2209888B1 (he)
JP (5) JP2011500024A (he)
KR (2) KR101849336B1 (he)
CN (2) CN104946591A (he)
AU (1) AU2008312007A1 (he)
CA (3) CA3177952A1 (he)
DK (1) DK2209888T3 (he)
ES (1) ES2764473T3 (he)
IL (3) IL292561A (he)
PL (1) PL2209888T3 (he)
PT (1) PT2209888T (he)
WO (1) WO2009051671A1 (he)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100299765A1 (en) * 2004-01-23 2010-11-25 Irina Klimanskaya Modalities for the treatment of degenerative diseases of the retina
US20110117063A1 (en) * 2004-01-23 2011-05-19 Advanced Cell Technology, Inc. Methods For Producing Enriched Populations of Human Retinal Pigment Epithelium Cells For Treatment of Retinal Degeneration
US20120322670A1 (en) * 2009-08-28 2012-12-20 Cellular Dynamics International, Inc. Identifying genetic variation in affected tissues
US20130330302A1 (en) * 2008-04-22 2013-12-12 Regenerative Research Foundation Retinal pigment epithelial stem cells
US10077424B2 (en) 2007-10-12 2018-09-18 Astellas Institute For Regenerative Medicine Methods of producing RPE cells and compositions of RPE cells
US10480031B2 (en) 2013-02-01 2019-11-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Method for generating Retinal Pigment Epithelium (RPE) cells from Induced Pluripotent Stem Cells (IPSCs)
US10485829B2 (en) 2009-11-17 2019-11-26 Astellas Institute For Regenerative Medicine Methods of producing human RPE cells and pharmaceutical preparations of human RPE cells
US10501724B2 (en) 2013-08-23 2019-12-10 Sumitomo Chemical Company, Limited Method for producing retinal tissue and retina-related cells
IL273107A (he) * 2011-11-14 2020-04-30 Astellas Inst For Regenerative Medicine תכשירים רוקחיים של תאי rpe הומאנים ושימושם
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US10793830B2 (en) * 2009-11-05 2020-10-06 Riken Method for producing retinal tissue in vitro
US10934523B2 (en) 2008-06-06 2021-03-02 Riken Method for culture of stem cell
US11013808B2 (en) 2015-08-18 2021-05-25 Astellas Institute For Regenerative Medicine Clinical formulations
US11214771B2 (en) 2014-10-24 2022-01-04 Sumitomo Dainippon Pharma Co., Ltd. Production method for nerve tissue
US11214772B2 (en) 2014-10-24 2022-01-04 Sumitomo Dainippon Pharma Co., Ltd. Production method for retinal tissue
US11371016B2 (en) 2016-04-22 2022-06-28 Sumitomo Pharma Co., Ltd. Method for producing retinal tissue
CN114717188A (zh) * 2015-09-08 2022-07-08 富士胶片细胞动力公司 干细胞来源的视网膜色素上皮的基于macs的纯化
US11422125B2 (en) 2015-03-23 2022-08-23 Astellas Institute For Regenerative Medicine Assays for potency of human retinal pigment epithelium (RPE) cells and photoreceptor progenitors
US11524035B2 (en) 2016-06-30 2022-12-13 Healios K.K. Transplantation medium
US11739366B2 (en) 2010-07-23 2023-08-29 Astellas Institute For Regenerative Medicine Methods for detection of rare subpopulations of cells and highly purified compositions of cells

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2128244A4 (en) 2007-01-18 2012-05-09 Riken INDUCTION / DIFFERENTIATION METHOD IN PHOTORECEPTOR CELL
EP2509596B1 (en) 2009-12-08 2019-08-28 Case Western Reserve University Gamma aminoacids for treating ocular disorders
FI20106354A0 (fi) * 2010-12-20 2010-12-20 Suomen Punainen Risti Veripalvelu Menetelmä ihmisen retinan pigmentti-epiteelisolujen valmistamiseksi
CN111265551A (zh) 2011-05-18 2020-06-12 加利福尼亚大学董事会 用于治疗视网膜疾病的组合物及方法
JP5985209B2 (ja) * 2011-11-25 2016-09-06 住友化学株式会社 眼杯様構造体の製造方法
JP5985208B2 (ja) * 2011-11-25 2016-09-06 住友化学株式会社 網膜組織の製造方法
JP5985207B2 (ja) * 2011-11-25 2016-09-06 住友化学株式会社 網膜色素上皮細胞の製造方法
JP6012164B2 (ja) 2011-11-25 2016-10-25 住友化学株式会社 多能性幹細胞由来の組織の凍結保存方法
CN113817679A (zh) * 2011-11-25 2021-12-21 住友化学株式会社 制备视网膜组织和视网膜相关细胞的方法
JP6067232B2 (ja) * 2011-11-25 2017-01-25 住友化学株式会社 網膜層特異的神経細胞を製造する方法
CN108478599B (zh) 2011-11-30 2022-08-02 安斯泰来再生医药协会 间充质基质细胞及其相关用途
US8961956B2 (en) 2011-11-30 2015-02-24 Ocata Therapeutics, Inc. Mesenchymal stromal cells and uses related thereto
US10519422B2 (en) * 2012-02-29 2019-12-31 Riken Method of producing human retinal pigment epithelial cells
CA3177929A1 (en) * 2012-12-21 2014-06-26 Astellas Institute For Regenerative Medicine Methods for production of platelets from pluripotent stem cells and compositions thereof
EP2828380B1 (en) * 2013-04-03 2016-09-21 Tampereen Yliopisto Methods and media for differentiating eye cells
WO2015087231A1 (en) 2013-12-11 2015-06-18 Pfizer Limited Method for producing retinal pigment epithelial cells
KR20170095365A (ko) * 2014-12-18 2017-08-22 유니베르시테 피에르 에 마리에 쿠리에 (파리 6) 망막 변성의 치료를 위한 otx2를 과발현하는 형질전환 rpe 세포
IL273405B2 (he) 2014-12-30 2024-01-01 Cell Cure Neurosciences Ltd אוכלוסיות תאי rpe ושיטות לייצורן
MX2017008818A (es) * 2014-12-30 2018-03-14 Cell Cure Neurosciences Ltd Metodos para tratar enfermedades de la retina.
JP7061961B2 (ja) 2015-10-20 2022-05-02 フジフィルム セルラー ダイナミクス,インコーポレイテッド 多能性幹細胞の免疫細胞への分化を誘導する方法
AU2016347652A1 (en) * 2015-10-26 2018-05-24 Cell Cure Neurosciences Ltd. Preparation of retinal pigment epithelium cells
CN107760649B (zh) * 2017-11-02 2021-03-30 北京全式金生物技术有限公司 一种用于人神经系统细胞培养和人多能性干细胞向神经系统细胞分化的营养添加剂
KR102198942B1 (ko) * 2019-07-02 2021-01-05 의료법인 성광의료재단 Pedf를 발현하거나 모세포에 비하여 pedf를 과발현하도록 유전적으로 조작된 중간엽 줄기세포
WO2021014515A1 (ja) * 2019-07-19 2021-01-28 東京エレクトロン株式会社 細胞の分化状態の評価方法
CN114981417A (zh) * 2019-10-30 2022-08-30 安斯泰来再生医药协会 生产视网膜色素上皮细胞的方法
TW202302839A (zh) 2021-03-09 2023-01-16 國立研究開發法人理化學研究所 低免疫原性視網膜色素上皮細胞之製造方法
US20230377685A1 (en) 2022-04-15 2023-11-23 Aspen Neuroscience, Inc. Methods of classifying the differentiation state of cells and related compositions of differentiated cells

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040018617A1 (en) * 2002-07-26 2004-01-29 Shiaw-Min Hwang Somatic pluripotent cells
US20070031386A1 (en) * 2004-01-23 2007-02-08 Advanced Cell Technology, Inc. Modalities for the treatment of degenerative diseases of the retina
US20110027333A1 (en) * 2007-04-18 2011-02-03 Hadasit Medical Research Services And Development Ltd. Stem cell-derived retin retinal pigment epithelial cells

Family Cites Families (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5610753A (en) 1991-12-12 1997-03-11 Eastman Kodak Company Optical design of laser scanner to reduce thermal sensitivity
WO1994025569A1 (en) 1993-04-30 1994-11-10 Photogenesis, Incorporated Retinal pigment epithelium transplantation
DE19609838A1 (de) 1996-03-13 1997-09-18 Boehringer Mannheim Gmbh Verfahren und Teststreifen zur Bestimmung eines Analyten
US6878544B2 (en) 1996-04-19 2005-04-12 Neurotech Sa Retinal cell lines with extended life-span and their applications
US20040086494A1 (en) 1996-10-07 2004-05-06 John Constance Mary Immune privileged cells for delivery of proteins and peptides
AU5734998A (en) * 1997-01-10 1998-08-03 Life Technologies, Inc. Embryonic stem cell serum replacement
US6331406B1 (en) 1997-03-31 2001-12-18 The John Hopkins University School Of Medicine Human enbryonic germ cell and methods of use
US6440933B1 (en) 1997-09-10 2002-08-27 University Of Florida Compounds and method for the prevention and treatment of diabetic retinopathy
WO1999034834A1 (en) 1998-01-02 1999-07-15 Titan Pharmaceuticals, Inc. Use of pigmented retinal epithelial cells for creation of an immune privilege site
WO1999045094A1 (en) 1998-03-02 1999-09-10 Compucyte Corp. Selective cell analysis
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
US6943153B1 (en) 1999-03-15 2005-09-13 The Regents Of The University Of California Use of recombinant gene delivery vectors for treating or preventing diseases of the eye
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
CN100497598C (zh) 1999-10-28 2009-06-10 马萨诸塞大学 多能细胞和细胞系的单雌生殖或单雄生殖产生,及其产生分化细胞和组织的用途
WO2001050848A2 (en) 2000-01-07 2001-07-19 Oregon Health And Science University Clonal propagation of primate offspring by embryo splitting
US6602711B1 (en) 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells
US20030084471A1 (en) 2000-03-16 2003-05-01 David Beach Methods and compositions for RNA interference
US6458589B1 (en) 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
JP2004522414A (ja) 2000-08-19 2004-07-29 アクソーディア・リミテッド 幹細胞分化
US6576464B2 (en) 2000-11-27 2003-06-10 Geron Corporation Methods for providing differentiated stem cells
AU2002236495B2 (en) 2000-11-29 2006-05-11 Allergan, Inc. Intraocular implants for preventing transplant rejection in the eye
DE10108412B4 (de) 2001-02-21 2006-03-09 Cevec Pharmaceuticals Gmbh Pigmentepithelzelle des Auges, ihre Herstellung und Verwendung in der Therapie einer Augen- oder Nervenerkrankung
CA2447015A1 (en) 2001-05-15 2002-11-21 Rappaport Family Institute For Research In The Medical Sciences Insulin producing cells derived from human embryonic stem cells
KR101073411B1 (ko) 2001-07-12 2011-10-17 제론 코포레이션 인간 다분화능 줄기 세포로부터 제조된 심근 세포 계통의 세포
EP1456374A4 (en) 2001-11-26 2005-08-17 Advanced Cell Tech Inc METHODS FOR THE PRODUCTION AND USE OF REPROGRAMME HUMAN SOMATIC CELL CORES AND AUTOLOGOUS AND ISOGENIC HUMAN STEM CELLS
IL162131A0 (en) 2001-12-07 2005-11-20 Geron Corp Islet cells from human embryonic stem cells
CN1602207A (zh) 2001-12-11 2005-03-30 法布罗根股份有限公司 抑制眼病理过程的方法
AU2003235652A1 (en) 2002-01-15 2003-07-30 Advanced Cell Technology, Inc. Cloning b and t lymphocytes
EP1495120B1 (en) 2002-04-18 2012-10-10 Acuity Pharmaceuticals, Inc Means and methods for the specific modulation of target genes in the eye
US20040091936A1 (en) 2002-05-24 2004-05-13 Michael West Bank of stem cells for producing cells for transplantation having HLA antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
EP1556063A2 (en) 2002-10-04 2005-07-27 TissueTech, Inc. Retinal pigment epithelial cell cultures on amniotic membrane and transplantation
AU2003302020B2 (en) 2002-11-14 2008-01-31 Ethicon Endo-Surgery, Inc. Methods and devices for detecting tissue cells
US20040156878A1 (en) 2003-02-11 2004-08-12 Alireza Rezania Implantable medical device seeded with mammalian cells and methods of treatment
CN101044404A (zh) 2003-03-03 2007-09-26 长冈实业株式会社 用于不同类型细胞检测和定量的方法和装置以及生物光盘在该检测和定量中的应用
WO2004098285A2 (en) 2003-05-08 2004-11-18 Cellartis Ab Cryopreservation of human blastocyst-derived stem cells by use of a closed straw vitrification method
KR100569168B1 (ko) 2003-08-08 2006-04-07 (주)아비코아생명공학연구소 조류 정원줄기세포의 배양방법 및 이에 의해 수득한 조류정원줄기세포
US7794704B2 (en) 2004-01-23 2010-09-14 Advanced Cell Technology, Inc. Methods for producing enriched populations of human retinal pigment epithelium cells for treatment of retinal degeneration
WO2005111197A1 (en) 2004-05-07 2005-11-24 Wisconsin Alumni Research Foundation Method of forming mesenchymal stem cells from embryonic stem cells
US6945971B1 (en) 2004-07-19 2005-09-20 Gwon Arlene E Controlled ocular lens regeneration
EP1799810A2 (en) 2004-10-12 2007-06-27 Technion Research & Development Foundation Ltd. Isolated primate embryonic cells and methods of generating and using same
AU2005304978A1 (en) 2004-11-04 2006-05-18 Advanced Cell Technology, Inc. Derivation of embryonic stem cells
US7893315B2 (en) 2004-11-04 2011-02-22 Advanced Cell Technology, Inc. Derivation of embryonic stem cells and embryo-derived cells
SG148171A1 (en) 2005-02-11 2008-12-31 Agency Science Tech & Res Methods of proliferating stem cells
JP2009517001A (ja) 2005-05-17 2009-04-30 リライアンス ライフ サイエンシーズ プライベイト リミテッド 哺乳動物細胞を使用するヒト胚性幹細胞株の構築
JP5006312B2 (ja) 2005-05-27 2012-08-22 エイジェンシー フォー サイエンス, テクノロジー アンド リサーチ バキュロウイルスベクターを使用して核酸分子を胚性幹細胞に送達する方法
WO2007015252A2 (en) 2005-08-03 2007-02-08 I.M.T. Interface Multigrad Technology Ltd. Somatic cells for use in cell therapy
JP4926615B2 (ja) 2006-08-29 2012-05-09 太陽化学株式会社 ガラクトマンナン酵素分解物の製造方法
KR100832592B1 (ko) 2006-08-17 2008-05-27 박현숙 폴리머 막을 이용한 줄기세포와 피더세포의 공배양방법
ZA200901914B (en) 2006-09-22 2010-06-30 Riken Stem cell culture medium and method
JP2008307007A (ja) 2007-06-15 2008-12-25 Bayer Schering Pharma Ag 出生後のヒト組織由来未分化幹細胞から誘導したヒト多能性幹細胞
AU2008312007A1 (en) 2007-10-12 2009-04-23 Advanced Cell Technology, Inc. Improved methods of producing RPE cells and compositions of RPE cells
HU0700675D0 (en) 2007-10-15 2007-12-28 Mta Tamogatott Kutatohelyek Ir Method for monitoring stem cell differentiation
US20090226955A1 (en) 2007-12-21 2009-09-10 University Of Miami Immortalized retinal pigmented epithelial cells
US20090233324A1 (en) 2008-03-11 2009-09-17 Kopf-Sill Anne R Methods for Diagnosing Cancer Using Samples Collected From A Central Vein Location or an Arterial Location
WO2009132156A1 (en) * 2008-04-22 2009-10-29 Regenerative Research Foundation Retinal pigment epithelial stem cells
US8652123B2 (en) 2008-09-02 2014-02-18 Geoffrey C. GURTNER Methods and devices for improving the appearance of tissue
KR102073730B1 (ko) 2009-11-17 2020-02-05 아스텔라스 인스티튜트 포 리제너러티브 메디슨 인간 rpe 세포의 생산 방법 및 인간 rpe 세포의 제약 제제
US11739366B2 (en) 2010-07-23 2023-08-29 Astellas Institute For Regenerative Medicine Methods for detection of rare subpopulations of cells and highly purified compositions of cells
WO2012149484A2 (en) 2011-04-29 2012-11-01 University Of Southern California Method of cryopreservation of stem cell-derived retinal pigment epithelial cells on polymeric substrate
CA2855941A1 (en) 2011-11-14 2013-05-23 Advanced Cell Technology, Inc. Pharmaceutical preparations of human rpe cells and uses thereof
US9850463B2 (en) 2012-02-01 2017-12-26 The Regents Of The University Of California Methods of culturing retinal pigmented epithelium cells, including xeno-free production, RPE enrichment, and cryopreservation
US9458428B2 (en) 2012-06-05 2016-10-04 The Regents Of The University Of California Methods and compositions for the rapid production of retinal pigmented epithelial cells from pluripotent cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040018617A1 (en) * 2002-07-26 2004-01-29 Shiaw-Min Hwang Somatic pluripotent cells
US20070031386A1 (en) * 2004-01-23 2007-02-08 Advanced Cell Technology, Inc. Modalities for the treatment of degenerative diseases of the retina
US20110027333A1 (en) * 2007-04-18 2011-02-03 Hadasit Medical Research Services And Development Ltd. Stem cell-derived retin retinal pigment epithelial cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Chaudhry et al. Basal medium composition and serum or serum replacement concentration influences on the maintenance of murine embryonic stem cells. Cytotechnology (2008) 58:173-179 *

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9193950B2 (en) 2004-01-23 2015-11-24 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9040038B2 (en) 2004-01-23 2015-05-26 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9562217B2 (en) 2004-01-23 2017-02-07 Astellas Institute For Regenerative Medicine Modalities for the treatment of degenerative diseases of the retina
US8268303B2 (en) 2004-01-23 2012-09-18 Advanced Cell Technology, Inc. Methods for producing enriched populations of human retinal pigment epithelium cells for treatment of retinal degeneration
US20100299765A1 (en) * 2004-01-23 2010-11-25 Irina Klimanskaya Modalities for the treatment of degenerative diseases of the retina
US20110117062A1 (en) * 2004-01-23 2011-05-19 Advanced Cell Technology, Inc. Methods For Producing Enriched Populations of Human Retinal Pigment Epithelium Cells
US9040039B2 (en) 2004-01-23 2015-05-26 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9649340B2 (en) 2004-01-23 2017-05-16 Astellas Institute For Regenerative Medicine Methods for producing enriched populations of human retinal pigment epithelium cells
US9040770B2 (en) 2004-01-23 2015-05-26 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9045732B2 (en) 2004-01-23 2015-06-02 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9080150B2 (en) 2004-01-23 2015-07-14 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9181524B2 (en) 2004-01-23 2015-11-10 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US20110117063A1 (en) * 2004-01-23 2011-05-19 Advanced Cell Technology, Inc. Methods For Producing Enriched Populations of Human Retinal Pigment Epithelium Cells For Treatment of Retinal Degeneration
US9730962B2 (en) 2004-01-23 2017-08-15 Astellas Institute For Regenerative Medicine Modalities for the treatment of degenerative diseases of the retina
US9650607B2 (en) 2004-01-23 2017-05-16 Astellas Institute For Regenerative Medicine Modalities for the treatment of degenerative diseases of the retina
US10077424B2 (en) 2007-10-12 2018-09-18 Astellas Institute For Regenerative Medicine Methods of producing RPE cells and compositions of RPE cells
US10034916B2 (en) * 2008-04-22 2018-07-31 Regenerative Research Foundation Methods of treating a retinal disease by retinal pigment epithelial stem cells
US20130330302A1 (en) * 2008-04-22 2013-12-12 Regenerative Research Foundation Retinal pigment epithelial stem cells
US10934523B2 (en) 2008-06-06 2021-03-02 Riken Method for culture of stem cell
US20120322670A1 (en) * 2009-08-28 2012-12-20 Cellular Dynamics International, Inc. Identifying genetic variation in affected tissues
US9506114B2 (en) * 2009-08-28 2016-11-29 Cellular Dynamics International, Inc. Identifying genetic variation in affected tissues
US10793830B2 (en) * 2009-11-05 2020-10-06 Riken Method for producing retinal tissue in vitro
US10485829B2 (en) 2009-11-17 2019-11-26 Astellas Institute For Regenerative Medicine Methods of producing human RPE cells and pharmaceutical preparations of human RPE cells
US11850261B2 (en) 2009-11-17 2023-12-26 Astellas Institute For Regenerative Medicine Methods of producing human RPE cells and pharmaceutical preparations of human RPE cells
US11739366B2 (en) 2010-07-23 2023-08-29 Astellas Institute For Regenerative Medicine Methods for detection of rare subpopulations of cells and highly purified compositions of cells
CN113786417A (zh) * 2011-11-14 2021-12-14 安斯泰来再生医药协会 人rpe细胞的药物制剂及其用途
IL273107A (he) * 2011-11-14 2020-04-30 Astellas Inst For Regenerative Medicine תכשירים רוקחיים של תאי rpe הומאנים ושימושם
US10480031B2 (en) 2013-02-01 2019-11-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Method for generating Retinal Pigment Epithelium (RPE) cells from Induced Pluripotent Stem Cells (IPSCs)
US11441184B2 (en) 2013-02-01 2022-09-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Method for generating retinal pigment epithelium (RPE) cells from induced pluripotent stem cells (IPSCs)
US10501724B2 (en) 2013-08-23 2019-12-10 Sumitomo Chemical Company, Limited Method for producing retinal tissue and retina-related cells
US11473056B2 (en) 2013-08-23 2022-10-18 Sumitomo Chemical Company, Limited Method for producing retinal tissue and retina-related cells
US11214771B2 (en) 2014-10-24 2022-01-04 Sumitomo Dainippon Pharma Co., Ltd. Production method for nerve tissue
US11214772B2 (en) 2014-10-24 2022-01-04 Sumitomo Dainippon Pharma Co., Ltd. Production method for retinal tissue
US11680941B2 (en) 2015-03-23 2023-06-20 Astellas Institute For Regenerative Medicine Assays for potency of human retinal pigment epithelium (RPE) cells and photoreceptor progenitors
US11422125B2 (en) 2015-03-23 2022-08-23 Astellas Institute For Regenerative Medicine Assays for potency of human retinal pigment epithelium (RPE) cells and photoreceptor progenitors
US11013808B2 (en) 2015-08-18 2021-05-25 Astellas Institute For Regenerative Medicine Clinical formulations
US11957754B2 (en) 2015-08-18 2024-04-16 Astellas Institute For Regenerative Medicine Clinical formulations
CN114717188A (zh) * 2015-09-08 2022-07-08 富士胶片细胞动力公司 干细胞来源的视网膜色素上皮的基于macs的纯化
US11371016B2 (en) 2016-04-22 2022-06-28 Sumitomo Pharma Co., Ltd. Method for producing retinal tissue
US11524035B2 (en) 2016-06-30 2022-12-13 Healios K.K. Transplantation medium
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11987813B2 (en) 2017-03-30 2024-05-21 The Research Foundation for The Sate University of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation

Also Published As

Publication number Publication date
EP2209888B1 (en) 2019-12-04
PT2209888T (pt) 2020-01-17
CA2702386A1 (en) 2009-04-23
CA3006687C (en) 2024-01-09
US20150086512A1 (en) 2015-03-26
JP6692768B2 (ja) 2020-05-13
KR20160045933A (ko) 2016-04-27
EP3636748A1 (en) 2020-04-15
JP2022079484A (ja) 2022-05-26
IL292561A (he) 2022-06-01
US20190062703A1 (en) 2019-02-28
KR20100065373A (ko) 2010-06-16
JP2017136088A (ja) 2017-08-10
CA3177952A1 (en) 2009-04-23
CA3006687A1 (en) 2009-04-23
AU2008312007A1 (en) 2009-04-23
CN104946591A (zh) 2015-09-30
KR101849336B1 (ko) 2018-04-16
IL246181A0 (he) 2016-07-31
CN101878295A (zh) 2010-11-03
JP2011500024A (ja) 2011-01-06
JP6137626B2 (ja) 2017-05-31
JP2020115882A (ja) 2020-08-06
US10077424B2 (en) 2018-09-18
EP2209888A4 (en) 2011-02-09
WO2009051671A1 (en) 2009-04-23
ES2764473T3 (es) 2020-06-03
PL2209888T3 (pl) 2020-05-18
CA2702386C (en) 2018-07-24
EP2209888A1 (en) 2010-07-28
IL204956A (he) 2016-06-30
DK2209888T3 (da) 2020-01-20
IL204956A0 (en) 2010-11-30
US20220049217A1 (en) 2022-02-17
JP2014168482A (ja) 2014-09-18
KR101849329B1 (ko) 2018-04-16

Similar Documents

Publication Publication Date Title
US20220049217A1 (en) Methods of producing rpe cells and compositions of rpe cells
US11850261B2 (en) Methods of producing human RPE cells and pharmaceutical preparations of human RPE cells
AU2022201847A1 (en) Improved methods of producing RPE cells and compositions of RPE cells
JP2012024100A (ja) 網膜の変性疾患の処置のための改良されたモーダリティー
CN107148276A (zh) 视网膜神经节细胞及其祖细胞
US20230042830A1 (en) Compositions and methods for generation of retinal ganglion cells from inducible pluripotent stem cells for the treatment of progressive optic neuropathies, including glaucoma
WO2022009969A1 (ja) 網膜色素上皮細胞の移植用生着促進剤
Embryonic EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM (IPS) CELLS

Legal Events

Date Code Title Description
AS Assignment

Owner name: ADVANCED CELL TECHNOLOGY, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MALCUIT, CHRISTOPHER;LEMIEUX, LINDA;HOLMES, WILLIAM;AND OTHERS;SIGNING DATES FROM 20101110 TO 20101202;REEL/FRAME:025495/0443

AS Assignment

Owner name: ADVANCED CELL TECHNOLOGY, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HUERTAS, PEDRO;REEL/FRAME:027964/0401

Effective date: 20120324

AS Assignment

Owner name: CAMHZN MASTER LDC, NEW YORK

Free format text: SECURITY AGREEMENT;ASSIGNORS:ADVANCED CELL TECHNOLOGY, INC.;MYTOGEN, INC.;REEL/FRAME:029698/0001

Effective date: 20121231

Owner name: CAMOFI MASTER LDC, NEW YORK

Free format text: SECURITY AGREEMENT;ASSIGNORS:ADVANCED CELL TECHNOLOGY, INC.;MYTOGEN, INC.;REEL/FRAME:029698/0001

Effective date: 20121231

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ADVANCED CELL TECHNOLOGY, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MALCUIT, CHRISTOPHER;LEMIEUX, LINDA;HOLMES, WILLIAM;AND OTHERS;SIGNING DATES FROM 20090318 TO 20090320;REEL/FRAME:034122/0403

AS Assignment

Owner name: MYTOGEN, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNORS:CAMOFI MASTER LDC;CAMHZN MASTER LDC;REEL/FRAME:034421/0863

Effective date: 20141003

Owner name: ADVANCED CELL TECHNOLOGY, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNORS:CAMOFI MASTER LDC;CAMHZN MASTER LDC;REEL/FRAME:034421/0863

Effective date: 20141003

AS Assignment

Owner name: ADVANCED CELL TECHNOLOGY, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HUERTAS, PEDRO;REEL/FRAME:034273/0668

Effective date: 20120324