WO2005070011A2 - Improved modalities for the treatment of degenerative diseases of the retina - Google Patents

Improved modalities for the treatment of degenerative diseases of the retina Download PDF

Info

Publication number
WO2005070011A2
WO2005070011A2 PCT/US2005/002273 US2005002273W WO2005070011A2 WO 2005070011 A2 WO2005070011 A2 WO 2005070011A2 US 2005002273 W US2005002273 W US 2005002273W WO 2005070011 A2 WO2005070011 A2 WO 2005070011A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
rpe
cell
hes
derived
Prior art date
Application number
PCT/US2005/002273
Other languages
French (fr)
Other versions
WO2005070011A3 (en
Inventor
Irina V. Klimanskaya
Robert Lanza
Original Assignee
Advanced Cell Technology, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2005207042A priority Critical patent/AU2005207042B2/en
Application filed by Advanced Cell Technology, Inc. filed Critical Advanced Cell Technology, Inc.
Priority to KR1020067016837A priority patent/KR101258292B1/en
Priority to CN2005800073590A priority patent/CN1968608B/en
Priority to EP05711960A priority patent/EP1708575A4/en
Priority to KR1020127008183A priority patent/KR101398356B1/en
Priority to JP2006551392A priority patent/JP2007522131A/en
Priority to KR1020137002345A priority patent/KR20130025953A/en
Priority to CA2555370A priority patent/CA2555370C/en
Priority to NZ548929A priority patent/NZ548929A/en
Priority to BRPI0507074-0A priority patent/BRPI0507074A/en
Priority to EP11183613.6A priority patent/EP2438816B1/en
Publication of WO2005070011A2 publication Critical patent/WO2005070011A2/en
Publication of WO2005070011A3 publication Critical patent/WO2005070011A3/en
Priority to IL177015A priority patent/IL177015B/en
Priority to US11/490,953 priority patent/US7795025B2/en
Priority to US12/857,866 priority patent/US9649340B2/en
Priority to AU2010249263A priority patent/AU2010249263B2/en
Priority to IL267126A priority patent/IL267126B/en
Priority to IL291162A priority patent/IL291162A/en

Links

Classifications

    • 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
    • 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
    • A61K35/44Vessels; Vascular smooth muscle cells; Endothelial cells; Endothelial progenitor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • 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
    • 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
    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • 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
    • C12N5/062Sensory transducers, e.g. photoreceptors; Sensory neurons, e.g. for hearing, taste, smell, pH, touch, temperature, pain
    • 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
    • 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
    • C12N2501/01Modulators of cAMP or cGMP, e.g. non-hydrolysable analogs, phosphodiesterase inhibitors, cholera toxin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • 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

Definitions

  • This invention relates generally to methods for improved cell-based therapies for retinal degeneration and other visual disorders as well as treatment of Parkinson's disease and for differentiating mammalian embryonic stem cells and mammalian embryo-derived cells into retinal pigment epithelium (RPE) cells and other eye tissue including, but not limited to) rods, cones, bipolar, comeal, neural, iris epithelium, and progenitor cells.
  • RPE retinal pigment epithelium
  • CNS central nervous system
  • ASD age-related macular degeneration
  • RPE retinal pigment epithelium
  • INL intemuncial
  • Restoration of moderate-to-high acuity vision therefore, requires the functional replacement of some or all of the damaged cellular layers.
  • retinitis pigmentosa a family of inherited retinal degenerations
  • RP retinitis pigmentosa
  • the underlying cellular mechanisms are diverse and can result from various mutations in many genes. Most involve mutations that alter the expression of photoreceptor-cell-specific genes, with mutations in the rhodopsin gene accounting for approximately 10% of these.
  • the regulatory genes of apoptosis are altered (for example, Box and Pax2). AMD is a clinical diagnosis encompassing a range of degenerative conditions that likely differ in etiology at the molecular level.
  • the retinal pigment epithelium is one of the most important cell types in the eye, as it is crucial to the support of the photoreceptor function. It performs several complex tasks, including phagocytosis of shed outer segments of rods and cones, vitamin A metabolism, synthesis of mucoploysacharides involved in the metabolite exchange in the subretinal space, transport of metabolites, regulation of angiogenesis, absorption of light, enhancement of resolution of images, and the regulation of many other functions in the retina through secreted proteins such as proteases and protease inhibitors..
  • An additional feature present in some cases of AMD is the presence of aberrant blood vessels, which result in a condition known as choroidal neovascularization (CNN).
  • CNN choroidal neovascularization
  • This neovascular (“wet”) form of AMD is particularly destructive and seems to result from a loss of proper regulation of angiogenesis. Breaks in Bruch's membrane as a result of RPE dysfunction allows new vessels from the choroidal circulation access to the subretinal space, where they can physically disrupt outer-segment organization and cause vascular leakage or hemorrhage leading to additional photoreceptor loss. C ⁇ N can be targeted by laser treatment. Thus, laser treatment for the "wet" form of AMD is in general use in the United States. There are often undesirable side effects, however, and therefore patient dissatisfaction with treatment outcome.
  • RPE plays an important role in photoreceptor maintenance, and regulation of angiogenesis
  • various RPE malfunctions in vivo are associated with vision-altering ailments, such as retinitis pigmentosa, RPE detachment, displasia, atlrrophy, retinopathy, macular dystrophy or degeneration, including age-related macular degeneration, which can result in photoreceptor damage and blindness.
  • the variety of other degenerative conditions affecting the macula include, but are not limited to, cone dystrophy, cone-rod dystrophy, malattia leventinese, Doyne honeycomb dystrophy, Sorsby's dystrophy, Stargardt disease, pattern/butterfly dystrophies, Best vitelliform dystrophy, North Carolina dystrophy, central areolar choroidal dystrophy, angioid streaks, and toxic maculopathies.
  • General retinal diseases that can secondarily effect the macula include retinal detachment, pathologic myopia, retinitis pigmentosa, diabetic retinopathy, CMV retinitis, occlusive retinal vascular disease, retinopathy of prematurity (ROP), choroidal rupture, ocular histoplasmosis syndrome (POHS), toxoplasmosis, and Leber's congenital amaurosis. None of the above lists is exhaustive. All of the above conditions involve loss of photoreceptors and, therefore, treatment options are few and insufficient. Because of its wound healing abilities, RPE has been extensively studied in application to transplantation therapy. In 2002, one year into the trial, patients were showing a 30-50% improvement.
  • RPE cells derived from fetal tissue are another problem, as these cells have shown a very low proliferative potential.
  • Emory University researchers performed a trial where they cultured RPE cells from a human eye donor in vitro and transplanted them into six patients with advanced Parkinson's Disease. Although a 30-50% decrease in symptoms was found one year after transplantation, there is a shortage of eye donors, this is not yet FDA approved, and there would still exist a need beyond what could be met by donated eye tissue.
  • therapies using ectopic RPE cells have been shown to behave like fibroblasts and have been associated with a number of destructive retinal complications including axonal loss (Villegas-Perez, et.
  • RPE delivered as a loose sheet tends to scroll up. This results in poor effective coverage of photoreceptors as well as a multilayered RPE with incorrect polarity, possibly resulting in cyst formation or macular edema.
  • Delivery of neural retinal grafts to the subretinal (submacular) space of the diseased human eye has been described in Kaplan et. al. (1997), Humayun et. al. (2000), and del Cerro et. al. (2000). A serious problem exists in that the neural retinal grafts typically do not functionally integrate with the host retina.
  • the purpose of the present invention is to provide improved methods for the derivation of eye cells including, but not limited to, neural cells, including horizontal cells and amacrine cells, retinal cells such as rods and cones, corneal cells, vascular cells, and RPE and RPE-like cells from stem cells and to provide improved methods and therapies for the treatment of retinal degeneration, h particular, these methods involve the use of RPE and RPE-like cells derived from human embryonic stem cells.
  • One embodiment of the present invention provides an improved method of generating cells for therapy for retinal degeneration using RPE cells, RPE-like cells, the progenitors of these cells or a combination of two or three of any of the preceding derived from mammalian embryonic stem cells in order to treat various conditions including but not limited to retinitis pigmentosa and macular degeneration and associated conditions.
  • the cell types which can be produced using this invention include, but are not limited to, RPE, RPE-like cells, and RPE progenitors. Cells which may also be produced include iris pigmented epithelial (IPE) cells.
  • Vision associated neural cells including intemuncial neurons (e.g.
  • retinal neurons of the inner nuclear layer (INL)) and amacrine cells interneurons that interact at the second synaptic level of the vertically direct pathways consisting of the photoreceptor-bipolar- ganglion cell chain - they are synaptically active in the inner plexiform layer (IPL) and serve to integrate, modulate and interpose a temporal domain to the visual message presented to the ganglion cell
  • IPL inner plexiform layer
  • retinal cells, rods, cones, and comeal cells can be produced.
  • cells providing the vasculature of the eye can also be produced.
  • the cells of the present invention may be transplanted into the subretinal space by using vitrectomy surgery.
  • Non-limiting examples include the transplantation of these cells in a suspension, matrix, or substrate.
  • Animal models of retinitis pigmentosa that may be treated 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 "rcdl, rcd2 & rcd3" dogs, photoreceptor dysplasia "pd” dog, and Briard "RPE-65” (dog).
  • Evaluation is performed using behavioral tests, fluorescent angiography, histology, or functional testing such as measuring the ability of the cells to perform phagocytosis (photoreceptor fragments), vitamin A metabolism, tight junctions conductivity, or evaluation using electron microscopy.
  • phagocytosis photoreceptor fragments
  • vitamin A metabolism vitamin A metabolism
  • tight junctions conductivity or evaluation using electron microscopy.
  • One of the many advantages to the methods presented here is the ability to produce and treat many more patients than it would be possible to treat if one were limited to using eye donor tissue.
  • a further embodiment of the present invention provides methods for the spontaneous differentiation of hES cells into cells with numerous characteristics of RPE. These RPE preparations are capable of phenotypic changes in culture and maintaining RPE characteristics through multiple passages.
  • the present invention also provides for methods of differentiation of established RPE cell lines into alternate neuronal lineages, comeal cells, retinal cells as a non-limiting example through the use of bFGF or FGF.
  • Another embodiment of the present invention is a method for the derivation of new RPE lines and progenitor cells from existing and new ES cell lines.
  • properties such as growth rate, expression of pigment, or de- differentiation and re-differentiation in culture, of RPE-like cells when they are derived from different ES cell lines.
  • Cells which may also be derived from existing and new ES cell lines include iris pigmented epithelial (IPE) cells.
  • vision associated neural cells including intemuncial neurons (e.g. "relay" neurons of the inner nuclear layer (INL)) and amacrine cells can also be produced using this invention. Additionally, retinal cells, rods, cones, and comeal cells can be produced.
  • cells providing the vasculature of the eye can also be produced.
  • 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).
  • 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.
  • an additional embodiment of the present invention includes the characterization of ES-derived RPE-like cells.
  • the ES-derived pigmented epithelial cells strongly resemble RPE by their morphology, behavior and molecular markers, their therapeutic value will depend on their ability to perform RPE functions and to remain non-carcinogenic. Therefore, the ES-derived RPE cells are characterized using one or more of the following techniques: (i) assessment of their functionality, i.e.
  • RPE-like ES cells derivatives through animal model transplantations, (as a non-limiting example this can include SCID mice);
  • phenoytping and karyotyping of RPE-like cells (iv) evaluation of ES cells-derived RPE-like cells and RPE tissue by gene expression profiling, (v) evaluation of the expression of molecular markers of RPE at the protein level, including bestrophin, CRALBP, RPE-65, PEDF.
  • the cells can also be evaluated based on their expression of transcriptional activators normally required for the eye development, including rx/rax, chxlO/vsx-2/alx, ots-1, otx-2, six3/optx, six6/optx2, mitf, pax6/mitf, and pax6/pax2 (Fischer and Reh, 2001, Baumer et. al., 2003).
  • An additional embodiment of the present invention is a method for the characterization of ES-derived RPE-like cells using at least one of the techniques selected from the group consisting of (i) assessment of the ES-derived RPE-like cells functionality; (ii) evaluation of the pluripotency of RPE-like ES cell derivatives through animal model transplantations; (iii) phenoytping and karyotyping of RPE- like cells; (iv) evaluation of gene expression profiling, (v) evaluation of the expression of molecular markers of RPE at the protein level; and (vi) the expression of transcriptional activators normally required for the eye development.
  • these techniques may be used for the assessment of multiple hES cell- derived cell types.
  • Another embodiment of the present invention is a method for the derivation of RPE cells and RPE precursor cells directly from human and non-human animal morula or blastocyst-staged embryos (EDCs) without the generation of ES cell lines.
  • Embryonic stem cells ES
  • hES human embryonic stem cells
  • hES human embryonic stem cells
  • Another embodiment of the present invention is a method of identifying cells such as RPE cells, hematopoietic cells, muscle cells, liver cells, pancreatic beta cells, neurons, endothelium, progenitor cells or other cells useful in cell therapy or research, derived from embryos, embryonic stem cell lines, or other embryonic cells with the capacity to differentiate into useful cell types by comparing the messenger RNA transcripts of such cells with cells derived in- vivo.
  • This method facilitates the identification of cells with a normal phenotype and for deriving cells optimized for cell therapy for research.
  • 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 retinal pigment epithelium
  • 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, and it's 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 is easily 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 48kD secreted protein with angiostatic properties (Karakousis et. al., 2001, Jablonski et.
  • CRALBP retinaldehyde-binding protein
  • VMD2 Best vitelliform macular dystrophy gene
  • PEDF pigment epithelium derived factor
  • RPE cells are normally mitotically quiescent, but can begin to divide in response to injury or photocoagulation. RPE cells adjacent to the injury flatten and proliferate forming a new monolayer (Zhao et. al, 1997). Several studies have indicated that the RPE monolayer can produce cells of fibroblast appearance that can later revert to their original RPE morphology (Grierson et. al., 1994, Kirchhof et. al., 1988, Lee et. al, 2001). It is unclear whether the dividing cells and pigmented epithelial layer are from the same lineage as two populations of RPE cells have been isolated: epithelial and fusiforms.
  • RPE can be maintained as an epithelium or rapidly dedifferentiate and become proliferative (Zhao 1997, Opas and Dziak, 1994). Interestingly, the epithelial phenotype can be reestablished in long-term quiescent cultures (Griersion et. al., 1994). In mammalian development, RPE shares the same progenitor with neural retina, the neuroepithelium of the optic vesicle. Under certain conditions, it has been suggested that RPE can transdifferentiate into neuronal progenitors (Opas and Dziak, 1994), neurons (Chen et.
  • bFGF a process associated with the expression of transcriptional activators normally required for the eye development, including rx/rax, chxlO/vsx-2/alx, ots-1, otx-2, six3/optx, six6/optx2, mitf, and pax6/pax2 (Fischer and Reh, 2001, Baumer et. al, 2003).
  • the present invention provides for the derivation of trabecular meshwork cells from hES and also for genetically modified trabecular meshwork cells for the treatment of glaucoma.
  • the present invention also provides for the derivation of trabecular meshwork cells from RPE progenitors and RPE-like cells and also for genetically modified trabecular meshwork cells for the treatment of glaucoma.
  • the present invention includes methods for the derivation of RPE cells and RPE precursor cells directly from human and non-human animal morula or blastocyst-staged embryos (EDCs) without the generation of ES cell lines, comprising a) maintaining ES cells in vitro in an undifferentiated state; b) differentiating the ES cells into RPE and RPE precursor cells; and, c) identifying cells the RPE cells by comparing the messenger RNA transcripts of such cells with cells derived in- vivo.
  • EDCs blastocyst-staged embryos
  • telomere lines or precursors to RPE cells that have an increased ability to prevent neovascularization
  • said methods comprising: a) aging a somatic cell from an animal such that telomerase is shortened wherein at least 10% of the nonnal replicative lifespan of the cell has been passed; and, b) using the somatic cell as a nuclear transfer donor cell to create cells that overexpress angiogenesis inhibitors, wherein the angiogenesis inhibitors can be Pigment Epithelium Derived Factor (PEDF/EPC-1).
  • PEDF/EPC-1 Pigment Epithelium Derived Factor
  • Figure 1A is a photograph of pigmented regions in a 2.5 month old adherent culture, a well of a 6-well plate, scanned;
  • Figure IB is a photograph of pigmented regions in a 2.5 month old cultured grown in EB, at 45x magnification;
  • Figure IC is a photograph of a pigmented area of an adherent culture;
  • Figure ID is a photograph of a pigmented region of an EB grown culture;
  • Figure IE is a photograph of the boundary between pigmented region and the rest of the culture, x200;
  • Figure F same as Figure E but at x400 magnification. Arrows in A and B point to pigmented regions Figure 2A-F.
  • FIG. 2A is a photograph showing primary EB outgrowth, 1 week
  • Figure 2B is a photograph showing the primary culture of cells, isolated by trypsin, 1 week
  • Figure 2C is a photograph showing epithelial islet surrounded by proliferating cells
  • Figure 2D is a photograph showing the regain of pigmentation and epithelial morphology in 1 month old culture
  • Figure 2E is a photograph showing the culture after 3 passages, x200 magnification
  • Figure 2F shows the same culture as in E, x400 magnification, Hoffman microscopy. Black arrows point to pigmented cells, white arrows show outgrowing cells with no pigment.
  • Figure 3 Left Panel (A-D) and Right Panel is a series of photographs and one graph - these show markers of RPE in hES cells-derived pigmented epithelial cells.
  • Figures 3 A and 3B are photographs showing immunolocalization of RPE marker, bestrophin and corresponding phase microscopy field, x200 magnification;
  • Figures 3C and 3D are photographs showing CRALBP and corresponding phase contrast microscopy field, x400 magnification.
  • FIG. 1 Shows the colocalization of bestrophin (A) and CRALBP (C) to pigmented cells (C,D); arrowheads point to the absence of staining for these proteins (A,B) in non-pigmented regions (C,D)
  • Figure 3 shows a photograph and graph of western blot of cell lysates (line hES #36) with antibodies to bestrophin (a) and CRALBP (b); c,d - undifferentiated hES cells , c ⁇ control to anti-CRALBP antibody, d — control to anti- bestrophin antibody
  • Figure 4 shows photographs which demonstrate the expression of markers of
  • Pax6 (Figure 4A), Pax2 (Figure 4E) and mitf (Figure 4B, Figure 4F) in RPE-like cells in long-term quiescent cultures.
  • Figure 4C Figure 4G - phase contrast
  • Figure 4D Figure 4H - merged images of Pax6/mitf phase contrast
  • Figure 4 A Figure 4B, Figure 4C
  • Figure 4E Figure 4F, Figure 4G
  • Figure 5A-B show photographs of RPE differentiation in the culture of human embryo-derived cells: bypassing the stage of derivation of ES cell lines.
  • Figure 6 shows the transcriptional comparison of RPE preparations.
  • FIG. 6A-F Based on the Ontological annotation, this table represents the expression patterns of RPE related genes for hES cell-derived retinal pigment epithelium (hES- RPE), hES cell derived transdifferentiated (hES-RPE-TD), ARPE- 19 and D407, and freshly isolated human RPE (fe-RPE).
  • Figure 6G Further data mining revealed known RPE specific ontologies, such as melanin biosynthesis, vision, retinol- binding, only in fetal RPE and ES-RPE but not ARPE- 19.
  • 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 cells refers to embryo-derived cells.
  • hES cells human embryonic stem cells
  • hES human embryo-derived cells
  • hES refers to cells isolated from the inner cell mass of human blastocysts or morulae and that have been serially passaged as cell lines and can also include blastomeres and aggregated blastomeres.
  • 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.
  • Embryonic stem (ES) cells which have the ability to differentiate into virtually any tissue of a human body can provide a limitless supply of rejuvenated and histocompatible cells for transplantation therapy, as the problem of immune rejection can be overcome with nuclear transfer and partheno genetic technology.
  • the recent findings of Hirano et. al. (2003) have shown that mouse ES cells can produce eye-like structures in differentiation experiments in vitro. Among those, pigmented epithelial cells were described, resembling retinal pigment epithelium. Preliminary experiments carried out at Advanced Cell Technology with primate and human ES cell lines show that a in a specialized culture system these cells differentiate into RPE-like cells that can be isolated and passaged.
  • RPE Human and mouse NT, Cyno parthenote ES cell derivatives have multiple features of RPE: these pigmented epithelial cells express four molecular markers of RPE - bestrophin, CRALBP, PEDF, and RPE65; like RPE, their proliferation in culture is accompanied by dedifferentiation - loss of pigment and epithelial morphology, both of which are restored after the cells form a monolayer and become quiescent.
  • RPE-like cells can be easily passaged, frozen and thawed, thus allowing their expansion.
  • the inventors have further shown that human ES cells also produce multiple eye (vitreous body)-like structures in differentiation experiments in vitro.
  • RPE Transplantation At present, chronic, slow rejection of the RPE allo grafts prevents scientists from determining the therapeutic efficacy of this RPE transplantation.
  • Several methods are being considered to overcome this obstacle. The easiest way is to use systemic immunosuppression, which is associated with serious side-effects such as cancer and infection.
  • a second approach is to transplant the patient's own RPE, i.e. homografts, but this has the drawback of using old, diseased RPE to replace even more diseased RPE.
  • a third approach is to use iris epithelium (IPE) from the same patient but this has the drawback that IPE may not perform all the vision related functions of RPE.
  • IPE iris epithelium
  • Nuclear transfer and parthenogenesis facilitate histocompatibility of grated RPE cells and progenitors.
  • RPE defects in Retinitis Pigmentosa 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.
  • One of the patients improved from barely seeing light to being able to count fingers held at a distance of about six feet from the patient's face.
  • vision improved to ability to see letters through tunnel vision.
  • the transplants in these studies were performed by injection, introducing the new retinal cells underneath the existing neural retina. Not all of the cells survived since the transplanted fetal cells were allogeneic (i.e. not genetically- matched), although those that did survive formed comiections with other neurons and begin to function like the photoreceptors around them. Approximately a year after the first eight people received the transplants, four have recovered some visual function and a fifth shows signs of doing so.
  • Three newly derived human embryonic stem cell lines are similar in properties to those described earlier (Thomson et. al. 1998, Reibunoff et. al., 2000, Richards et. al., 2000, Lanzendorf et. al., 2001): they maintain undifferentiated phenotype and express known markers of undifferentiated hES cells, Oct-4, alkaline phosphatase, SSEA-3, SSEA-4, TRA-I-60, TRA-I-81 through 45 passages in culture or over 130 population doublings. All hES cell lines differentiate into derivatives of three germ layers in EB or long term adherent cultures and in teratomas.
  • hES cells are similar to retinal pigment epithelium by the following criteria: morphologically, they have a typical epithelial cobblestone monolayer appearance and contain dark brown pigment in their cytoplasm, which is known to be present in the human body only in melanocytes, keratinocytes, retinal and iris pigment epithelium (IPE). Melanocytes, however, are non-epithelial cells, and keratynocytes don't secrete but only accumulate melanin. The set of RPE- specific proteins — bestrophin, CRALBP, PEDF - present in these cells indicates that they are likely to be similar to RPE and not IPE.
  • the inventors have observed the same pattern of tubulin beta III localization in primary and passaged cultures of RPE and RPE-like cells which can reflect a dedifferentiation of such cells in culture or indicate a separate population of cells committed to a neuronal fate, that were originally located next to pigmented cells through differentiation of hES cells in long-term cultures and could have been co- isolated with RPE-like cells.
  • RPE and the neural retina share the same bipotential neuroepithelial progenitor, and their fate was shown to be determined by Pax2, Pax6, and Mitf (Baumer et. al, 2003), the latter being a target of the first two.
  • Pax6 at earlier stages acts as an activator of proneural genes and is downregulated in the RPE in further development, remaining in amacrine and ganglion cells in mature retina (reviewed by Ashery -Padan and Grass, 2001). In goldfish, it is also found in mitotically active progenitors of regenerating neurons (Hitchcock et. al., 1996). The inventors have found that many of the RPE-like cells expressed mitf and Pax6 in a pattern similar to tubulin beta III and were found only in non-pigmented cells of non-epithelial morphology that surround pigmented epithelial islands in long term cultures or in cells with a "partial" RPE phenotype (lightly pigmented and loosely packed).
  • progenitors can in turn coexist with mature RPE-like cells in culture or appear as a result of dedifferentiation of RPE-like cells.
  • cells of neural retina can transdifferentiate into RPE in vitro (Opas et.
  • tubulin beta III and Pax6 positive cells could represent a transient stage of such transdifferentiation of co-isolated neural cells or neural progenitors into RPE-like cells.
  • Differentiation of hES cells into RPE-like cells happened spontaneously when using methods described in the Examples below, and the inventors noticed that pigmented epithelial cells reliably appeared in cultures older than 6-8 weeks and their number progressed overtime — in 3-5 months cultures nearly every EB had a large pigmented region.
  • six more newly derived hES lines turned into RPE-like cells, which suggests that since neural fate is usually chosen by ES cells spontaneously, RPE-like cells can arise by default as an advanced stage of such pathway.
  • permissive and/or instructive differentiation signals come from extracellular matrix and growth factors produced by differentiating derivatives of hES cells.
  • the model of differentiation of hES cells into RPE-like cells could be a useful tool to study how such microenvironment orchestrates RPE differentiation and transdifferentiation.
  • 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, athrophy, 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. It has been shown in several animal models and in humans (Gouras et. al., 2002, Stanga et. al., 2002, Binder et. al., 2002, Schraermeyer et. al., 2001, reviewed by Lund et.
  • Example 1 Spontaneous differentiation into pigmented epithelial cells in long term cultures When hES cell cultures are allowed to overgrow on MEF in the absence of
  • LIF, FGF and Plasmanate they form a thick multilayer of cells.
  • dark islands of cells appear within the larger clusters (Figl). These dark cells are easily seen with the naked eye and looked like "freckles" in a plate of cells as shown in Fig 1 A. At higher magnification these islands appear as tightly packed polygonal cells in a cobblestone monolayer, typical of epithelial cells, with brown pigment in the cytoplasm (Fig. IC).
  • Fig 1, E,F There are differences in the amount of pigment in the cells with cells in the central part of the islands having the most pigment and those near the edges the least.
  • EB embryoid bodies
  • fig IB pigmented epithelial cells
  • Morphology of the cells in the pigmented regions of EBs was very similar to that of adherent cultures (fig ID).
  • Example 2 Isolation and culture of pigmented epithelial cells The inventors isolated pigmented epithelial cells from both adherent hES cell cultures and from EBs.
  • Pigmented polygonal cells were digested with enzymes (trypsin, and/or collagenase, and/or dispase), and the cells from these pigmented islands were selectively picked with a glass capillary. Although care was taken to pick only pigmented cells, the population of isolated cells invariably contained some non-pigmented cells. After plating cells on gelatin or laminin for 1-2 days, the cells were considered to be primary cultures (P0). Primary cultures contained islands of pigmented polygonal cells as well as some single pigmented cells. After 3-4 days in culture, non-pigmented cells that seemed to have lost epithelial morphology (flatter and cells with lamellipodia) appeared at the periphery of some islands (fig.2).
  • enzymes trypsin, and/or collagenase, and/or dispase
  • the epithelial structure and cobblestone morphology are not shared by other pigmented cells, e.g. melanocytes. It is also noteworthy that RPE cells have been shown to lose and regain their pigment and epithelial morphology when grown in culture (Zhao 1997, Opas and Dziak, 1994), and the pigmented cells behaved in a similar manner, so to test the hypothesis that the ES derived cells may be RPE, they were stained with antibodies to known markers for RPE: bestrophin and CRALBP. Figure 4 (left panel) shows membrane localization of bestrophin (A) and CRALBP (C), both are found in pigmented epithelial islands.
  • FIG. 4 Another known PRE marker, RPE65, was found in the RPE-like cells by real-time RT-PCR ( Figure 4, right panel, bottom), the PEDF ELISA assay showed the presence of PEDF in cell lysates of all presumed RPE cultures, and Western blot showed a band of approximately 48 kD (not shown).
  • Figure 4 shows localization of PAX-6, Pax2, mitf, and tubulin beta III in recently passaged and old cultures of hES cells-derived RPE.
  • tubulin beta III had a similar pattern of distribution (not shown).
  • mitf- positive and Pax6-negative cells located close to the periphery of pigmented islands (figure 4, A-C).
  • Pax2 was found only in a very small subset of mitf-negative cells (Figure 4, E-H). No presence of either of these proteins was ever detected in the cells of "mature" pigmented epithelial islands.
  • these markers in cells that only had some RPE features were often visible, i.e. either looked epithelial but had no pigment or in certain single pigmented cells away from pigmented epithelial islands.
  • Example 4 Characterization of RPE-like cells derived from hES cell lines H9 and ACT J-l from Cyno-1 ES cells and derivation of RPE-like cells from existing hES cell lines HI and H7.
  • An RPE-like cell line is expanded, tested for freezing and recovery, and characterized using the following methods and molecular markers of RPE cells: bestrophin and CRALBP by Western blot and immunofluorescence, PEDF by ELISA and Western blot, and REP65 by RT-PCR.
  • the cells are injected in SCID mice with undifferentiated hES or Cyno-1 cells as a control to evaluate tumorigenicity.
  • Karyotyping of RPE-like cells will be done by a clinical laboratory on a commercial basis.
  • RPE-like cells Characterization of the functional properties of RPE-like cells and studies of their transplantation potential are then carried out as otherwise described in this application and also using those techniques known to those skilled in the art. Gene expression profiling experiments are done using Affymetrix human genome arrays. Gene expression is compared in RPE-like cells derived from ES cells and in retinal samples from autopsies. Several animal models can be used to verify the effectiveness of the transplanted RPE-like cells, including but not limited to, rhesus monkey, rat, and rabbit.
  • ES cells are cultured on feeder cells or as embryoid bodies (EB) in the presence of bFGF, insulin, TGF-beta, IBMX, bmp-2, bmp-4 or their combinations, including stepwise addition.
  • EB embryoid bodies
  • ES cells are grown on various extracellular matrix-coated plates (laminin, fibronectin, collagen I, collagen IV, Matrigel, etc.) in evaluating the role of ECM in RPE formation.
  • RPE progenitors Pax6, Pax2, mitf
  • CRALBP bestrophin, PEDF, REP65
  • RPE progenitors Pax6, Pax2, mitf
  • CRALBP bestrophin, PEDF, REP65
  • This approach can also be used to produce common progenitors of RPE and other eye tissues, such as photoreceptor or neural retina which can be isolated and further characterized for their differentiation potential and used in transplantation studies.
  • Example 6 Derivation of RPE and other eye tissue progenitors from existing and new ES cell lines.
  • RPE progenitor markers will be correlated with the expression of the surface proteins in order to find a unique combination of surface markers for RPE progenitor cells. If such markers are found, antibodies to surface proteins can be used to isolate a pure population of RPE progenitors that can be then cultured and further differentiated in culture or used in transplantation studies to allow their differentiation after grafting. If the data from the gene expression profiling experiments is insufficient, to isolate the RPE progenitors the following approach will be used. ES cells and RPE- like cells will be transfected with GFP under the control of a Pax6 promoter, and stable transfectants will be selected.
  • GFP/Pax6-positive cells will be isolated by FACS and used as an antigen source for mouse injection to raise monoclonal antibodies to the surface molecules of Pax6 positive cells. Because Pax6 is present not only in RPE progenitors, screening will be done (by FACS) using several strategies: a) against proliferating RPE-like cells, b) against Pax2-positive RPE cells, c) against mitf-positive RPE cells. For b) and c) RPE cells will be transfected with GFP under the corresponding promoter; as a negative control, RPE or ES cells negative by these antigens will be used.
  • RPE-like cells Three ES cell lines that already produced RPE-like cells (H9, ACT J-l, Cyno-1), RPE-like cells will be used to continue to derive RPE-like cells and their progenitors as described in Aims 1 and 2, and HI and H7 hES cell lines will be used to produce new RPE-like cell lines. After expansion and characterization for molecular markers of RPE, these lines will be single-cloned, and the resulting lines will be characterized as described in Aim 1. The lines meeting criteria for RPE cells will be used for transplantation studies.
  • New human ES cell lines will be derived from unused INF embryos, from donated oocytes, stimulated to develop without fertilization (parthenote), and from generated developing blastocysts obtained from donated oocytes with the application of nuclear transfer technology. RPE-like cells and common eye progenitors will be derived from these lines using the approach in Aim 2, and the resulting lines will be characterized as in Aim 1. [Optional] new human ES cell lines will be derived in a virus-free system, characterized and submitted for clinical trials.
  • Example 7 Therapeutic potential of RPE-like cells and progenitors in various animal models of retinitis pigmentosa & macular degeneration.
  • Primate ES cells are tested in cynomologus monkeys (Macaques). Initially, vitrectomy surgery is performed and the cells are transplanted into the subretinal space of the animals. The first step is the transplantation of the cells in the suspension format after which a substrate or matrix is used to produce a monolayer transplantation.
  • Example 8 Direct differentiation of RPE cells from human embryo-derived cells. Human blastocyst-staged embryos are plated in the presence of murine or chick embryo fibroblasts with or without immunosurgery to remove the trophectoderm or directly plates on extracellular matrix protein-coated tissue cultureware. Instead of culturing and passaging the cells to produce a human ES cell line, the cells are directly differentiated.
  • hEDC cell cultures When hEDC cell cultures are allowed to overgrow on MEF in the absence of LIF, FGF and Plasmanate, they will form a thick multilayer of cells. (Alternate growth factors, media, and FBS can be used to alternate direct differentiation as is known to those skilled in the art.) About 6 weeks later, dark islands of cells will appear within the larger clusters. These dark cells are easily seen with the naked eye and looked like "freckles" in a plate of cells as shown in Fig 5B. At higher magnification these islands appear as tightly packed polygonal cells in a cobblestone monolayer, typical of epithelial cells, with brown pigment in the cytoplasm (Fig. 5A).
  • hEDC cells When hEDC cells are directly differentiated they may, though typically have not, fonned embryoid bodies (EB). Pigmented epithelial cells appear in about 1-2% of these differentiated cells and/or EBs in the first 6-8 weeks. Over time more and more EBs develop pigmented cells, and by 3 months nearly every EB had a pigmented epithelial region. Morphology of the cells in the pigmented regions of EBs was very similar to that of adherent cultures.
  • MEF medium high glucose DMEM, supplemented with 2mM GlutaMAX I, and 500 u/ml Penicillin, 500 ug/ml streptomycin (all from Invitrogen) and 16% FCS (HyCLone).
  • hES Cells Growth medium knockout high glucose DMEM supplemented with 500 u/ml Penicillin, 500 ug/mlstreptomycin, 1 % non-essential amino acids solution, 2mM GlutaMAX I securely 0.1 mM beta-mercaptoethanol, 4 ng/ml bFGF (Invitrogen), 1 -ng/ml human LIF (Chemicon, Temecula, CA), 8.4% of Serum Replacement (SR, Invitrogen) and 8.4% Plasmanate (Bayer).
  • Derivation medium contained the same components as growth medium except that it had lower concentration of SR and Plasmanate (4.2% each) and 8.4 % FCS and 2x concentration of human LIF and bFGF, as compared to growth medium.
  • EB medium same as growth medium except bFGF, LIF, and Plasmanate; the SR concentration was 13%.
  • RPE medium 50% EB medium and 50% MEF medium.
  • hES cell lines The cell lines, hES 35, 36, 45, used for these studies were derived with modifications of previously reported procedures (Thomson et. al., 1998, Reubinoff et. al, 2000, Lanzendorf et. al., 2001).
  • Immunostaining Cells were fixed with 2% paraformaldehyde, permeabilized with 0.1% NP- 40 for localization of intracellular antigens, and blocked with 10% goat serum, 10% donkey serum (Jackson Immunoresearch Laboratories, West Grove, PA) in PBS (Invitrogen) for at least one hour. Incubation with primary antibodies was carried out overnight at 4oC, the secondary antibodies (Jackson Immunoresearch Laboratories, West Grove, PA) were added for one hour. Between all incubations specimens were washed with 0.1% Tween-20 (Sigma) in PBS 3-5 times, 10-15 minutes each wash.
  • Specimens were mounted using Nectashield with DAPI (Vector Laboratories, Burlingame, CA) and observed under fluorescent microscope (Nikon). Localization of alkaline phosphatase was done either by Vector Red (Vector Laboratories, Burlingame, CA) to live cells or after the second wash during immunostaining according to manufacturer's instructions.
  • Trizol reagent Invitrogen
  • Qiagen Quantitect Probe RT-PCR reagents
  • hES cell lines Two female one male hES cell lines were used in these studies. Details on the derivation of these hES lines are reported elsewhere. All lines have been passaged more than 50 times during which time they maintain an undifferentiated colony morphology, high alkaline phosphatase activity, and express Oct-4, SSEA-3, SSEA- 4, TRA 1-60, and TRA 1-81 (data not shown). Two lines have normal karyotype (hES36, hES35), while there were both normal and aneuploid subpopulations in hES45. Upon spontaneous differentiation both in vitro and in teratomas all lines expressed the markers of three germ layers - muscle actin, alpha-fetoprotein, and tubulin beta III.
  • Example 9 Use of transcript genomics to identify normal differentiated cells differentiated ex vivo.
  • Transcriptomics - hES-cell derivatives are likely to play an important role in the future of regenerative medicine. Qualitative assessment of these and other stem cell derivatives remains a challenge that could be approached using functional genomics.
  • the gene expression profile of our data set was compared to two human RPE cell lines (non-transformed ARPE-19 and transformed D407, Rogojina et. al., 2003) to determine whether hES-RPE have similar global transcriptional profiles.
  • Example 10 Use of RPE cells for treatment of Parkinson 's Disease.
  • hRPE can be used as an alternative source of cells for cell therapy of Parkinson's Disease because they secrete L-DOPA.
  • One of the many advantages to the use of hES cell-derived RPE is that it circumvents the shortage of donor eye tissue. It also facilitates the use of gene therapy.

Abstract

This invention relates to methods for improved cell-based therapies for retinal degeneration and for differentiating human embryonic stem cells and human embryo-derived into retinal pigment epithelium (RPE) cells and other retinal progenitor cells.

Description

IMPROVED MODALITIES FOR THE TREATMENT OF DEGENERATIVE DISEASES OF THE RETINA
This application is being filed as a PCT International Patent application on 24 January 2005, in the name of Advanced Cell Technology, Inc, a U.S. national corporation, applicant for the designation of all countries except the U.S., and Irina V. Klir anskaya, a U.S. citizen, applicant for the designation of the US only, and claims priority to U.S. Provisional Application Serial No. 60/538,964, filed January 23, 2004.
FIELD OF THE INVENTION This invention relates generally to methods for improved cell-based therapies for retinal degeneration and other visual disorders as well as treatment of Parkinson's disease and for differentiating mammalian embryonic stem cells and mammalian embryo-derived cells into retinal pigment epithelium (RPE) cells and other eye tissue including, but not limited to) rods, cones, bipolar, comeal, neural, iris epithelium, and progenitor cells.
BACKGROUND OF THE INVENTION Many parts of the central nervous system (CNS) exhibit laminar organization, and neuropatho logical processes generally involve more than one of these multiple cellular layers. Diseases of the CNS frequently include neuronal cell loss, and, because of the absence of endogenous repopulation, effective recovery of function following CNS-related disease is either extremely limited or absent, hi particular, the common retinal condition known as age-related macular degeneration (AMD) results from the loss of photoreceptors together with the retinal pigment epithelium (RPE), with additional variable involvement of intemuncial ("relay") neurons of the inner nuclear layer (INL). Restoration of moderate-to-high acuity vision, therefore, requires the functional replacement of some or all of the damaged cellular layers. Anatomically, retinitis pigmentosa (RP), a family of inherited retinal degenerations, is a continuing decrease in the number of photocreceptor cell nuclei which leads to loss of vision. Although the phenotype is similar across most forms of RP, the underlying cellular mechanisms are diverse and can result from various mutations in many genes. Most involve mutations that alter the expression of photoreceptor-cell-specific genes, with mutations in the rhodopsin gene accounting for approximately 10% of these. In other forms of the disease, the regulatory genes of apoptosis are altered (for example, Box and Pax2). AMD is a clinical diagnosis encompassing a range of degenerative conditions that likely differ in etiology at the molecular level. All cases of AMD share the feature of photoreceptor cell loss within the central retina. However, this common endpoint appears to be a secondary consequence of earlier abnormalities at the level of the RPE, neovascularization, and underlying Bruch's membrane. The latter may relate to difficulties with photoreceptor membrane turnover, which are as yet poorly understood.
Additionally, the retinal pigment epithelium is one of the most important cell types in the eye, as it is crucial to the support of the photoreceptor function. It performs several complex tasks, including phagocytosis of shed outer segments of rods and cones, vitamin A metabolism, synthesis of mucoploysacharides involved in the metabolite exchange in the subretinal space, transport of metabolites, regulation of angiogenesis, absorption of light, enhancement of resolution of images, and the regulation of many other functions in the retina through secreted proteins such as proteases and protease inhibitors.. An additional feature present in some cases of AMD is the presence of aberrant blood vessels, which result in a condition known as choroidal neovascularization (CNN). This neovascular ("wet") form of AMD is particularly destructive and seems to result from a loss of proper regulation of angiogenesis. Breaks in Bruch's membrane as a result of RPE dysfunction allows new vessels from the choroidal circulation access to the subretinal space, where they can physically disrupt outer-segment organization and cause vascular leakage or hemorrhage leading to additional photoreceptor loss. CΝN can be targeted by laser treatment. Thus, laser treatment for the "wet" form of AMD is in general use in the United States. There are often undesirable side effects, however, and therefore patient dissatisfaction with treatment outcome. This is due to the fact that laser bums, if they occur, are associated with photoreceptor death and with absolute, irreparable blindness within the corresponding part of the visual field. In addition, laser treatment does not fix the underlying predisposition towards developing CΝV. Indeed, laser burns have been used as a convenient method for induction of CΝN in monkeys (Archer and Gardiner,, 1981). Macular laser treatments for CΝN are used much more sparingly in other countries such as the U.K. There is no generally recognized treatment for the more common "dry" form of AMD, in which there is photoreceptor loss overlying irregular patches of RPE atrophy in the macula and associated extracellular material called drusen. Since RPE plays an important role in photoreceptor maintenance, and regulation of angiogenesis, various RPE malfunctions in vivo are associated with vision-altering ailments, such as retinitis pigmentosa, RPE detachment, displasia, atlrrophy, retinopathy, macular dystrophy or degeneration, including age-related macular degeneration, which can result in photoreceptor damage and blindness. Specifically and in addition to AMD, the variety of other degenerative conditions affecting the macula include, but are not limited to, cone dystrophy, cone-rod dystrophy, malattia leventinese, Doyne honeycomb dystrophy, Sorsby's dystrophy, Stargardt disease, pattern/butterfly dystrophies, Best vitelliform dystrophy, North Carolina dystrophy, central areolar choroidal dystrophy, angioid streaks, and toxic maculopathies. General retinal diseases that can secondarily effect the macula include retinal detachment, pathologic myopia, retinitis pigmentosa, diabetic retinopathy, CMV retinitis, occlusive retinal vascular disease, retinopathy of prematurity (ROP), choroidal rupture, ocular histoplasmosis syndrome (POHS), toxoplasmosis, and Leber's congenital amaurosis. None of the above lists is exhaustive. All of the above conditions involve loss of photoreceptors and, therefore, treatment options are few and insufficient. Because of its wound healing abilities, RPE has been extensively studied in application to transplantation therapy. In 2002, one year into the trial, patients were showing a 30-50% improvement. It has been shown in several animal models and in humans (Gouras et. al., 2002, Stanga et. al., 2002, Binder et. al., 2002, Schraermeyer et. al., 2001, reviewed by Lund et. al., 2001) that RPE transplantation has a good potential of vision restoration. However, even in an immune-privileged site such as the eye, there is a problem with graft rejection, hindering the progress of this approach if allogenic transplantation is used. Although new photoreceptors (PRCs) have been introduced experimentally by transplantation, grafted PRCs show a marked reluctance to link up with surviving neurons of the host retina. Reliance on RPE cells derived from fetal tissue is another problem, as these cells have shown a very low proliferative potential. Emory University researchers performed a trial where they cultured RPE cells from a human eye donor in vitro and transplanted them into six patients with advanced Parkinson's Disease. Although a 30-50% decrease in symptoms was found one year after transplantation, there is a shortage of eye donors, this is not yet FDA approved, and there would still exist a need beyond what could be met by donated eye tissue. Thus far, therapies using ectopic RPE cells have been shown to behave like fibroblasts and have been associated with a number of destructive retinal complications including axonal loss (Villegas-Perez, et. al., 1998) and proliferative vitreoretinopathy (PVR) with retinal detachment (Cleary and Ryan, 1979). RPE delivered as a loose sheet tends to scroll up. This results in poor effective coverage of photoreceptors as well as a multilayered RPE with incorrect polarity, possibly resulting in cyst formation or macular edema. Delivery of neural retinal grafts to the subretinal (submacular) space of the diseased human eye has been described in Kaplan et. al. (1997), Humayun et. al. (2000), and del Cerro et. al. (2000). A serious problem exists in that the neural retinal grafts typically do not functionally integrate with the host retina. In addition, the absence of an intact RPE mono layer means that RPE dysfunction or disruption of Bruch's membrane has not been rectified. Both are fundamental antecedents of visual loss. Thus, there exists no effective means for reconstituting RPE in any of the current therapies and there remain deficiencies in each, particularly the essential problem of a functional discomiection between the graft and the host retina. Therefore there exists the need for an improved retinal therapy.
SUMMARY OF THE INVENTION The purpose of the present invention is to provide improved methods for the derivation of eye cells including, but not limited to, neural cells, including horizontal cells and amacrine cells, retinal cells such as rods and cones, corneal cells, vascular cells, and RPE and RPE-like cells from stem cells and to provide improved methods and therapies for the treatment of retinal degeneration, h particular, these methods involve the use of RPE and RPE-like cells derived from human embryonic stem cells. One embodiment of the present invention provides an improved method of generating cells for therapy for retinal degeneration using RPE cells, RPE-like cells, the progenitors of these cells or a combination of two or three of any of the preceding derived from mammalian embryonic stem cells in order to treat various conditions including but not limited to retinitis pigmentosa and macular degeneration and associated conditions. The cell types which can be produced using this invention include, but are not limited to, RPE, RPE-like cells, and RPE progenitors. Cells which may also be produced include iris pigmented epithelial (IPE) cells. Vision associated neural cells including intemuncial neurons (e.g. "relay" neurons of the inner nuclear layer (INL)) and amacrine cells (interneurons that interact at the second synaptic level of the vertically direct pathways consisting of the photoreceptor-bipolar- ganglion cell chain - they are synaptically active in the inner plexiform layer (IPL) and serve to integrate, modulate and interpose a temporal domain to the visual message presented to the ganglion cell) can also be produced using this invention. Additionally, retinal cells, rods, cones, and comeal cells can be produced. In a further embodiment of the present invention^ cells providing the vasculature of the eye can also be produced. The cells of the present invention may be transplanted into the subretinal space by using vitrectomy surgery. Non-limiting examples include the transplantation of these cells in a suspension, matrix, or substrate. Animal models of retinitis pigmentosa that may be treated 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 "rcdl, rcd2 & rcd3" dogs, photoreceptor dysplasia "pd" dog, and Briard "RPE-65" (dog). Evaluation is performed using behavioral tests, fluorescent angiography, histology, or functional testing such as measuring the ability of the cells to perform phagocytosis (photoreceptor fragments), vitamin A metabolism, tight junctions conductivity, or evaluation using electron microscopy. One of the many advantages to the methods presented here is the ability to produce and treat many more patients than it would be possible to treat if one were limited to using eye donor tissue. A further embodiment of the present invention provides methods for the spontaneous differentiation of hES cells into cells with numerous characteristics of RPE. These RPE preparations are capable of phenotypic changes in culture and maintaining RPE characteristics through multiple passages. The present invention also provides for methods of differentiation of established RPE cell lines into alternate neuronal lineages, comeal cells, retinal cells as a non-limiting example through the use of bFGF or FGF. Another embodiment of the present invention is a method for the derivation of new RPE lines and progenitor cells from existing and new ES cell lines. There can be variations in the properties, such as growth rate, expression of pigment, or de- differentiation and re-differentiation in culture, of RPE-like cells when they are derived from different ES cell lines. There can be certain variations in their functionality and karyotypic stability, so it is desirable to provide methods for the derivation of new RPE lines and new ES cell lines which would allow choosing the lines with desired properties that can be clonally selected to produce a pure population of high quality RPE-like cells. Cells which may also be derived from existing and new ES cell lines include iris pigmented epithelial (IPE) cells. In an additional embodiment, vision associated neural cells including intemuncial neurons (e.g. "relay" neurons of the inner nuclear layer (INL)) and amacrine cells can also be produced using this invention. Additionally, retinal cells, rods, cones, and comeal cells can be produced. In a further embodiment of the present invention, cells providing the vasculature of the eye can also be produced. 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). Alternatively such cells may be genetically modified with exogenous genes that inhibit neovascularization. Another embodiment of the present invention utilized a bank of ES or embryo-derived cells with homozygosity in the HLA region such that said cells have reduced complexity of their HLA antigens. Therefore, an additional embodiment of the present invention includes the characterization of ES-derived RPE-like cells. Although the ES-derived pigmented epithelial cells strongly resemble RPE by their morphology, behavior and molecular markers, their therapeutic value will depend on their ability to perform RPE functions and to remain non-carcinogenic. Therefore, the ES-derived RPE cells are characterized using one or more of the following techniques: (i) assessment of their functionality, i.e. phagocytosis of the photoreceptor fragments, vitamin A metabolism, wound healing potential; (ii) evaluation of the pluripotency of RPE-like ES cells derivatives through animal model transplantations, (as a non-limiting example this can include SCID mice); (iii) phenoytping and karyotyping of RPE-like cells; (iv) evaluation of ES cells-derived RPE-like cells and RPE tissue by gene expression profiling, (v) evaluation of the expression of molecular markers of RPE at the protein level, including bestrophin, CRALBP, RPE-65, PEDF. The cells can also be evaluated based on their expression of transcriptional activators normally required for the eye development, including rx/rax, chxlO/vsx-2/alx, ots-1, otx-2, six3/optx, six6/optx2, mitf, pax6/mitf, and pax6/pax2 (Fischer and Reh, 2001, Baumer et. al., 2003). An additional embodiment of the present invention is a method for the characterization of ES-derived RPE-like cells using at least one of the techniques selected from the group consisting of (i) assessment of the ES-derived RPE-like cells functionality; (ii) evaluation of the pluripotency of RPE-like ES cell derivatives through animal model transplantations; (iii) phenoytping and karyotyping of RPE- like cells; (iv) evaluation of gene expression profiling, (v) evaluation of the expression of molecular markers of RPE at the protein level; and (vi) the expression of transcriptional activators normally required for the eye development. In a further embodiment these techniques may be used for the assessment of multiple hES cell- derived cell types. Another embodiment of the present invention is a method for the derivation of RPE cells and RPE precursor cells directly from human and non-human animal morula or blastocyst-staged embryos (EDCs) without the generation of ES cell lines. Embryonic stem cells (ES) can be indefinitely maintained in vitro in an undifferentiated state and yet are capable of differentiating into virtually any cell type. Thus 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. To date, the differentiation of human and mouse ES cells into numerous cell types have been reported (reviewed by Smith, 2001) including cardiomyocytes [Kehat et. al. 2001, Mummery et. al., 2003 Carpenter et. al, 2002], neurons and neural precursors (Reubinoff et. al.2000, Carpenter et. al.2001, Schuldiner et. al, 2001), adipocytes (Bpst et. al, 2002, Aubert et. al., 1999), hepatocyte-like cells (Rambhatla et. al., 2003), hematopoetic cells (Chadwick et. al., 2003). oocytes (Hubner et. al., 2003), thymocyte-like cells (Lin RY et. al., 2003), pancreatic islet cells (Kalian, 2003), and osteob lasts (Zur Nieden et. al., 2003).
Another embodiment of the present invention is a method of identifying cells such as RPE cells, hematopoietic cells, muscle cells, liver cells, pancreatic beta cells, neurons, endothelium, progenitor cells or other cells useful in cell therapy or research, derived from embryos, embryonic stem cell lines, or other embryonic cells with the capacity to differentiate into useful cell types by comparing the messenger RNA transcripts of such cells with cells derived in- vivo. This method facilitates the identification of cells with a normal phenotype and for deriving cells optimized for cell therapy for research. 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, and it's 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 is easily recognized by its cobblestone cellular morphology of black pigmented cells. In addition, there are several known markers of the RPE, including cellular retinaldehyde-binding protein (CRALBP), a cytoplasmic protein that is also found in apical microvilli (Bunt-Milam and Saari, 1983); 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 48kD secreted protein with angiostatic properties (Karakousis et. al., 2001, Jablonski et. al., 2000). An unusual feature of the RPE is its apparent plasticity. RPE cells are normally mitotically quiescent, but can begin to divide in response to injury or photocoagulation. RPE cells adjacent to the injury flatten and proliferate forming a new monolayer (Zhao et. al, 1997). Several studies have indicated that the RPE monolayer can produce cells of fibroblast appearance that can later revert to their original RPE morphology (Grierson et. al., 1994, Kirchhof et. al., 1988, Lee et. al, 2001). It is unclear whether the dividing cells and pigmented epithelial layer are from the same lineage as two populations of RPE cells have been isolated: epithelial and fusiforms. (McKay and Burke, 1994). hi vitro, depending on the combination of growth factors and substratum, RPE can be maintained as an epithelium or rapidly dedifferentiate and become proliferative (Zhao 1997, Opas and Dziak, 1994). Interestingly, the epithelial phenotype can be reestablished in long-term quiescent cultures (Griersion et. al., 1994). In mammalian development, RPE shares the same progenitor with neural retina, the neuroepithelium of the optic vesicle. Under certain conditions, it has been suggested that RPE can transdifferentiate into neuronal progenitors (Opas and Dziak, 1994), neurons (Chen et. al., 2003, Vinores e.t al., 1995), and lens epithelium (Eguchi, 1986). One of the factors which can stimulate the change of RPE into neurons is bFGF (Opaz and Dziak, 1994, a process associated with the expression of transcriptional activators normally required for the eye development, including rx/rax, chxlO/vsx-2/alx, ots-1, otx-2, six3/optx, six6/optx2, mitf, and pax6/pax2 (Fischer and Reh, 2001, Baumer et. al, 2003). Recently, it has been shown that the margins of the chick retina contain neural stem cells (Fischer and Reh, 2000) and that the pigmented cells in that area, which express pax6/mitf, can form neuronal cells in response to FGF (Fisher and Reh, 2001). The present invention provides for the derivation of trabecular meshwork cells from hES and also for genetically modified trabecular meshwork cells for the treatment of glaucoma. The present invention also provides for the derivation of trabecular meshwork cells from RPE progenitors and RPE-like cells and also for genetically modified trabecular meshwork cells for the treatment of glaucoma. The present invention includes methods for the derivation of RPE cells and RPE precursor cells directly from human and non-human animal morula or blastocyst-staged embryos (EDCs) without the generation of ES cell lines, comprising a) maintaining ES cells in vitro in an undifferentiated state; b) differentiating the ES cells into RPE and RPE precursor cells; and, c) identifying cells the RPE cells by comparing the messenger RNA transcripts of such cells with cells derived in- vivo. Further provided by the present invention are methods for the derivation of RPE lines or precursors to RPE cells that have an increased ability to prevent neovascularization, said methods comprising: a) aging a somatic cell from an animal such that telomerase is shortened wherein at least 10% of the nonnal replicative lifespan of the cell has been passed; and, b) using the somatic cell as a nuclear transfer donor cell to create cells that overexpress angiogenesis inhibitors, wherein the angiogenesis inhibitors can be Pigment Epithelium Derived Factor (PEDF/EPC-1). The present invention provides methods for the treatment of Parkinson's disease with hES cell-derived RPE, RPE-like and/or RPE progenitor cells. These may be delivered by stereotaxic intrastriatal implantation with or microcarriers. Alternately, they may be delivered without the use of microcarriers. The cells may also be expanded in culture and used in the treatment of Parkinson's disease by any method known to those skilled in the art. Other features and advantages of the invention will be apparent from the following detailed description. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1A-F. is a series of photographs showing the appearance of pigmented areas (characteristic of RPE cells) in spontaneously differentiating hES cells. Figure 1A is a photograph of pigmented regions in a 2.5 month old adherent culture, a well of a 6-well plate, scanned; Figure IB is a photograph of pigmented regions in a 2.5 month old cultured grown in EB, at 45x magnification; Figure IC is a photograph of a pigmented area of an adherent culture; Figure ID is a photograph of a pigmented region of an EB grown culture; Figure IE is a photograph of the boundary between pigmented region and the rest of the culture, x200; Figure F same as Figure E but at x400 magnification. Arrows in A and B point to pigmented regions Figure 2A-F. is a series of photographs which show the loss and regain of pigmentation and epithelial morphology in culture. Figure 2A is a photograph showing primary EB outgrowth, 1 week; Figure 2B is a photograph showing the primary culture of cells, isolated by trypsin, 1 week; Figure 2C is a photograph showing epithelial islet surrounded by proliferating cells; Figure 2D is a photograph showing the regain of pigmentation and epithelial morphology in 1 month old culture; Figure 2E is a photograph showing the culture after 3 passages, x200 magnification; Figure 2F shows the same culture as in E, x400 magnification, Hoffman microscopy. Black arrows point to pigmented cells, white arrows show outgrowing cells with no pigment. Figure 3 Left Panel (A-D) and Right Panel is a series of photographs and one graph - these show markers of RPE in hES cells-derived pigmented epithelial cells. Figures 3 A and 3B are photographs showing immunolocalization of RPE marker, bestrophin and corresponding phase microscopy field, x200 magnification; Figures 3C and 3D are photographs showing CRALBP and corresponding phase contrast microscopy field, x400 magnification. Arrows show the colocalization of bestrophin (A) and CRALBP (C) to pigmented cells (C,D); arrowheads point to the absence of staining for these proteins (A,B) in non-pigmented regions (C,D) Figure 3, Right Panel shows a photograph and graph of western blot of cell lysates (line hES #36) with antibodies to bestrophin (a) and CRALBP (b); c,d - undifferentiated hES cells , c~control to anti-CRALBP antibody, d — control to anti- bestrophin antibody Figure 4 shows photographs which demonstrate the expression of markers of
Pax6 (Figure 4A), Pax2 (Figure 4E) and mitf (Figure 4B, Figure 4F) in RPE-like cells in long-term quiescent cultures. Figure 4C, Figure 4G - phase contrast, Figure 4D, Figure 4H - merged images of Pax6/mitf phase contrast (Figure 4 A, Figure 4B, Figure 4C) and Pax2/mitf/phase contrast (Figure 4E, Figure 4F, Figure 4G). Figure 5A-B show photographs of RPE differentiation in the culture of human embryo-derived cells: bypassing the stage of derivation of ES cell lines. Figure 6 shows the transcriptional comparison of RPE preparations. Figure 6A-F - Based on the Ontological annotation, this table represents the expression patterns of RPE related genes for hES cell-derived retinal pigment epithelium (hES- RPE), hES cell derived transdifferentiated (hES-RPE-TD), ARPE- 19 and D407, and freshly isolated human RPE (fe-RPE). Figure 6G - Further data mining revealed known RPE specific ontologies, such as melanin biosynthesis, vision, retinol- binding, only in fetal RPE and ES-RPE but not ARPE- 19.
DETAILED DESCRIPTION OF THE INVENTION Various embodiments of the invention are described in detail and may be further illustrated by the provided examples. As used in the description herein and throughout the claims that follow, the meaning of "a," "an," and "the" includes plural reference unless the context clearly dictates otherwise. Also, as used in the description herein, the meaning of "in" includes "in" and "on" unless the context clearly dictates otherwise. The terms used in this specification generally have their ordinary meanings in the art, within the context of the invention, and in the specific context where each term is used. Certain terms that are used to describe the invention are discussed below, or elsewhere in the specification, to provide additional guidance to the practitioner in describing the compositions and methods of the invention and how to make and use them. For convenience, certain terms may be highlighted, for example using italics and/or quotation marks. The use of highlighting has no influence on the scope and meaning of a term; the scope and meaning of a term is the same, in the same context, whether or not it is highlighted. It will be appreciated that the same thing can be said in more than one way. Consequently, alternative language and synonyms may be used for any one or more of the terms discussed herein, nor is any special significance to be placed upon whether or not a term is elaborated or discussed herein. Synonyms for certain terms are provided. A recital of one or more synonyms does not exclude the use of other synonyms. The use of examples anywhere in this specification, including examples of any terms discussed herein, is illustrative only, and in no way limits the scope and scope of the invention so long as data are processed, sampled, converted, or the like according to the invention without regard for any particular theory or scheme of action. Definitions By "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. The term "embryonic 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 "human embryonic stem cells" (hES cells) refers human embryo- derived cells. More specifically hES refers to cells isolated from the inner cell mass of human blastocysts or morulae and that have been serially passaged as cell lines and can also include blastomeres and aggregated blastomeres. The term "human embryo-derived cells" (hEDC) 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. Embryonic stem (ES) cells which have the ability to differentiate into virtually any tissue of a human body can provide a limitless supply of rejuvenated and histocompatible cells for transplantation therapy, as the problem of immune rejection can be overcome with nuclear transfer and partheno genetic technology. The recent findings of Hirano et. al. (2003) have shown that mouse ES cells can produce eye-like structures in differentiation experiments in vitro. Among those, pigmented epithelial cells were described, resembling retinal pigment epithelium. Preliminary experiments carried out at Advanced Cell Technology with primate and human ES cell lines show that a in a specialized culture system these cells differentiate into RPE-like cells that can be isolated and passaged. Human and mouse NT, Cyno parthenote ES cell derivatives have multiple features of RPE: these pigmented epithelial cells express four molecular markers of RPE - bestrophin, CRALBP, PEDF, and RPE65; like RPE, their proliferation in culture is accompanied by dedifferentiation - loss of pigment and epithelial morphology, both of which are restored after the cells form a monolayer and become quiescent. Such RPE-like cells can be easily passaged, frozen and thawed, thus allowing their expansion. The inventors have further shown that human ES cells also produce multiple eye (vitreous body)-like structures in differentiation experiments in vitro. Histological analysis of these structures show a pattern of cells consistent with early retinal development, including aggregates of cells similar to rods and cones. RPE Transplantation At present, chronic, slow rejection of the RPE allo grafts prevents scientists from determining the therapeutic efficacy of this RPE transplantation. Several methods are being considered to overcome this obstacle. The easiest way is to use systemic immunosuppression, which is associated with serious side-effects such as cancer and infection. A second approach is to transplant the patient's own RPE, i.e. homografts, but this has the drawback of using old, diseased RPE to replace even more diseased RPE. Yet, a third approach is to use iris epithelium (IPE) from the same patient but this has the drawback that IPE may not perform all the vision related functions of RPE. Ultimately a method will need to be found to eliminate rejection and then scientists can determine the true efficacy of RPE transplantation in AMD and APJVID. Nuclear transfer and parthenogenesis facilitate histocompatibility of grated RPE cells and progenitors. RPE defects in Retinitis Pigmentosa 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 RP 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), a second involves another protein unique to RPE, RPE65. Once rejection is conquered, both of these forms of RP should be treatable immediately by RPE transplantation. This treatment was inconceivable a few years ago when RP was a hopelessly untreatable and a poorly understood form of blindness. New research in RPE transplantation suggests there is promise for the treatment of retinal degeneration, including macular degeneration. In addition, a number of patients with advanced RP have regained some useful vision following fetal retinal cell transplant. One of the patients, for instance, improved from barely seeing light to being able to count fingers held at a distance of about six feet from the patient's face. In a second case, vision improved to ability to see letters through tunnel vision. The transplants in these studies were performed by injection, introducing the new retinal cells underneath the existing neural retina. Not all of the cells survived since the transplanted fetal cells were allogeneic (i.e. not genetically- matched), although those that did survive formed comiections with other neurons and begin to function like the photoreceptors around them. Approximately a year after the first eight people received the transplants, four have recovered some visual function and a fifth shows signs of doing so. Three newly derived human embryonic stem cell lines are similar in properties to those described earlier (Thomson et. al. 1998, Reibunoff et. al., 2000, Richards et. al., 2000, Lanzendorf et. al., 2001): they maintain undifferentiated phenotype and express known markers of undifferentiated hES cells, Oct-4, alkaline phosphatase, SSEA-3, SSEA-4, TRA-I-60, TRA-I-81 through 45 passages in culture or over 130 population doublings. All hES cell lines differentiate into derivatives of three germ layers in EB or long term adherent cultures and in teratomas. One of the differentiation derivatives of hES cells is similar to retinal pigment epithelium by the following criteria: morphologically, they have a typical epithelial cobblestone monolayer appearance and contain dark brown pigment in their cytoplasm, which is known to be present in the human body only in melanocytes, keratinocytes, retinal and iris pigment epithelium (IPE). Melanocytes, however, are non-epithelial cells, and keratynocytes don't secrete but only accumulate melanin. The set of RPE- specific proteins — bestrophin, CRALBP, PEDF - present in these cells indicates that they are likely to be similar to RPE and not IPE. Another similarity is the behavior of isolated pigmented cells in culture, when little or no pigment was seen in proliferating cells but was retained in tightly packed epithelial islands or re- expressed in newly established cobblestone monolayer after the cells became quiescent. Such behavior was described for RPE cells in culture (reviewed by Zhao et. al., 1997), and it was previously reported (Vinores et. al, 1995) that a neuronal marker tubulin beta III was specifically localized in dedifferentiating RPE cells in vitro and not in the cells with the typical RPE morphology suggesting that it reflects the plasticity of RPE and its ability to dedifferentiate to a neural lineage. The inventors have observed the same pattern of tubulin beta III localization in primary and passaged cultures of RPE and RPE-like cells which can reflect a dedifferentiation of such cells in culture or indicate a separate population of cells committed to a neuronal fate, that were originally located next to pigmented cells through differentiation of hES cells in long-term cultures and could have been co- isolated with RPE-like cells. In the growing optic vesicle RPE and the neural retina share the same bipotential neuroepithelial progenitor, and their fate was shown to be determined by Pax2, Pax6, and Mitf (Baumer et. al, 2003), the latter being a target of the first two. Pax6 at earlier stages acts as an activator of proneural genes and is downregulated in the RPE in further development, remaining in amacrine and ganglion cells in mature retina (reviewed by Ashery -Padan and Grass, 2001). In goldfish, it is also found in mitotically active progenitors of regenerating neurons (Hitchcock et. al., 1996). The inventors have found that many of the RPE-like cells expressed mitf and Pax6 in a pattern similar to tubulin beta III and were found only in non-pigmented cells of non-epithelial morphology that surround pigmented epithelial islands in long term cultures or in cells with a "partial" RPE phenotype (lightly pigmented and loosely packed). In proliferating cells in recently passaged cultures all these markers were found nearly in every cell suggesting either a reversal of RPE-like cells to progenitor stage at the onset of proliferation or massive proliferation of retinal progenitors. Interestingly, in teratomas where islands of pigmented cells of epithelial morphology were also found, Pax6 was expressed in non-pigmented cells adjacent to pigmented regions (data not shown). Multiple studies have previously shown dedifferentiation of RPE in culture and their transdifferentiation into cells of neuronal phenotype (Reh and Gretton, 1987, Skaguchi et. al, 1997, Vinores et. al, 1995, Chen et. al, 2003), neuronal, amacrine and photoreceptor cells (Zhao et. al., 1995), glia (Skaguchi et. al, 1997), neural retina (Galy et. al, 2002), and to neuronal progenitors (Opaz and Dziak, 1993). Such progenitors can in turn coexist with mature RPE-like cells in culture or appear as a result of dedifferentiation of RPE-like cells. At the same time, cells of neural retina can transdifferentiate into RPE in vitro (Opas et. al., 2001), so alternatively, tubulin beta III and Pax6 positive cells could represent a transient stage of such transdifferentiation of co-isolated neural cells or neural progenitors into RPE-like cells. Differentiation of hES cells into RPE-like cells happened spontaneously when using methods described in the Examples below, and the inventors noticed that pigmented epithelial cells reliably appeared in cultures older than 6-8 weeks and their number progressed overtime — in 3-5 months cultures nearly every EB had a large pigmented region. In addition to the described hES lines, six more newly derived hES lines turned into RPE-like cells, which suggests that since neural fate is usually chosen by ES cells spontaneously, RPE-like cells can arise by default as an advanced stage of such pathway. It is also possible that in such long term cultures, where differentiating hES cells form a multi-layered environment, permissive and/or instructive differentiation signals come from extracellular matrix and growth factors produced by differentiating derivatives of hES cells. The model of differentiation of hES cells into RPE-like cells could be a useful tool to study how such microenvironment orchestrates RPE differentiation and transdifferentiation. 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, athrophy, 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. It has been shown in several animal models and in humans (Gouras et. al., 2002, Stanga et. al., 2002, Binder et. al., 2002, Schraermeyer et. al., 2001, reviewed by Lund et. al., 2001) that RPE transplantation has a good potential of vision restoration. Recently another prospective niche for RPE transplantation was proposed and even reached the phase of clinical trials: since these cells secrete dopamine, they could be used for treatment of Parkinson disease (Subramanian, 2001). However, even in an immune-privileged eye, there is a problem of graft rejection, hindering the progress of this approach if allogenic transplant is used. The other problem is the reliance on fetal tissue, as adult RPE has a very low proliferative potential. 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 new differentiation derivative of human ES cells, retinal pigment epithelium-like cells and the reliability and simplicity of such differentiation system may offer an attractive potential supply of RPE cells for transplantation. EXAMPLES
Example 1 Spontaneous differentiation into pigmented epithelial cells in long term cultures When hES cell cultures are allowed to overgrow on MEF in the absence of
LIF, FGF and Plasmanate, they form a thick multilayer of cells. About 6 weeks later, dark islands of cells appear within the larger clusters (Figl). These dark cells are easily seen with the naked eye and looked like "freckles" in a plate of cells as shown in Fig 1 A. At higher magnification these islands appear as tightly packed polygonal cells in a cobblestone monolayer, typical of epithelial cells, with brown pigment in the cytoplasm (Fig. IC). There are differences in the amount of pigment in the cells with cells in the central part of the islands having the most pigment and those near the edges the least. (Fig 1, E,F). When hES cells form embryoid bodies (EB) - pigmented epithelial cells appear in about 1-2% of EBs in the first 6-8 weeks (fig IB) . Over time more and more EBs develop pigmented cells, and by 3 months nearly every EB had a pigmented epithelial region (fig ID). Morphology of the cells in the pigmented regions of EBs was very similar to that of adherent cultures (fig ID). Example 2 Isolation and culture of pigmented epithelial cells The inventors isolated pigmented epithelial cells from both adherent hES cell cultures and from EBs. Pigmented polygonal cells were digested with enzymes (trypsin, and/or collagenase, and/or dispase), and the cells from these pigmented islands were selectively picked with a glass capillary. Although care was taken to pick only pigmented cells, the population of isolated cells invariably contained some non-pigmented cells. After plating cells on gelatin or laminin for 1-2 days, the cells were considered to be primary cultures (P0). Primary cultures contained islands of pigmented polygonal cells as well as some single pigmented cells. After 3-4 days in culture, non-pigmented cells that seemed to have lost epithelial morphology (flatter and cells with lamellipodia) appeared at the periphery of some islands (fig.2). The number of such peripheral cells increased over time, suggesting that these cells were proliferating, and after 2 weeks most cells in the newly formed monolayer contained very little or no pigment. After continued culture, for another 2-3 weeks, pigmented epithelial cells began to reappear, visibly indistinguishable from those in the original cultures (fig 2). Example 3 Detection of RPE markers The preliminary characterization of these differentiated human cells as RPE is based on their similarity to RPE cultures previously described; principally, their epithelial morphology and possession of pigment. There are three types of pigmented epithelial cells in human body: retinal and iris pigmented epithelium and keratinocytes, but the latter don't secrete pigment. The epithelial structure and cobblestone morphology are not shared by other pigmented cells, e.g. melanocytes. It is also noteworthy that RPE cells have been shown to lose and regain their pigment and epithelial morphology when grown in culture (Zhao 1997, Opas and Dziak, 1994), and the pigmented cells behaved in a similar manner, so to test the hypothesis that the ES derived cells may be RPE, they were stained with antibodies to known markers for RPE: bestrophin and CRALBP. Figure 4 (left panel) shows membrane localization of bestrophin (A) and CRALBP (C), both are found in pigmented epithelial islands. Not all of the cells stain with these antibodies and intensity of staining correlated with pigment expression and "tightness" of colonies - the borders of each pigmented island where cells were larger and more loosely packed showed lower expression of both proteins. To further characterize presumably RPE cells, analysis was performed on the expression of bestrophin, CRALBP by Western blotting. Figure 4 (right panel) shows the bands, corresponding to bestrophin, 68 kD (a), CRALBP, 36 kD (b) in cell lysates. All these proteins were found in both primary cultures and subsequent passages. Another known PRE marker, RPE65, was found in the RPE-like cells by real-time RT-PCR (Figure 4, right panel, bottom), the PEDF ELISA assay showed the presence of PEDF in cell lysates of all presumed RPE cultures, and Western blot showed a band of approximately 48 kD (not shown). Detection of markers of neuronal and retinal progenitors in RPE-like cultures Figure 4 shows localization of PAX-6, Pax2, mitf, and tubulin beta III in recently passaged and old cultures of hES cells-derived RPE. In proliferating cultures (day 3 after trypsinization, not shown ) where RPE-like morphology of the proliferating cells is lost, nearly every cell showed the presence of mitf, Pax6, tubulin beta III and nestin (not shown). Pax2 was found only a small subset of cells which appeared mitf-negative, while there was a strong degree of co-localization of Pax6/mitf, mitf/tubulin beta III, and Pax6/tubulin beta III. In 21 days old quiescent cultures after pigmented epithelial islands were reestablished, groups of PAX-6 and mitf were found mostly in non-pigmented cells of non-epithelial morphology between pigmented epithelial islands (Figure 4, A-C). and tubulin beta III had a similar pattern of distribution (not shown). However, there were populations of mitf- positive and Pax6-negative cells, located close to the periphery of pigmented islands (figure 4, A-C). Pax2 was found only in a very small subset of mitf-negative cells (Figure 4, E-H). No presence of either of these proteins was ever detected in the cells of "mature" pigmented epithelial islands. However, these markers in cells that only had some RPE features were often visible, i.e. either looked epithelial but had no pigment or in certain single pigmented cells away from pigmented epithelial islands.
Example 4 Characterization of RPE-like cells derived from hES cell lines H9 and ACT J-l from Cyno-1 ES cells and derivation of RPE-like cells from existing hES cell lines HI and H7. An RPE-like cell line is expanded, tested for freezing and recovery, and characterized using the following methods and molecular markers of RPE cells: bestrophin and CRALBP by Western blot and immunofluorescence, PEDF by ELISA and Western blot, and REP65 by RT-PCR. The cells are injected in SCID mice with undifferentiated hES or Cyno-1 cells as a control to evaluate tumorigenicity. Karyotyping of RPE-like cells will be done by a clinical laboratory on a commercial basis. Characterization of the functional properties of RPE-like cells and studies of their transplantation potential are then carried out as otherwise described in this application and also using those techniques known to those skilled in the art. Gene expression profiling experiments are done using Affymetrix human genome arrays. Gene expression is compared in RPE-like cells derived from ES cells and in retinal samples from autopsies. Several animal models can be used to verify the effectiveness of the transplanted RPE-like cells, including but not limited to, rhesus monkey, rat, and rabbit.
Example 5 Optimization of the differentiation culture system ensuring high yields of RPE-like cells. ES cells are cultured on feeder cells or as embryoid bodies (EB) in the presence of bFGF, insulin, TGF-beta, IBMX, bmp-2, bmp-4 or their combinations, including stepwise addition. Alternatively, ES cells are grown on various extracellular matrix-coated plates (laminin, fibronectin, collagen I, collagen IV, Matrigel, etc.) in evaluating the role of ECM in RPE formation. Expression of molecular markers of early RPE progenitors (Pax6, Pax2, mitf) and of RPE cells (CRALBP, bestrophin, PEDF, REP65) are evaluated at various time intervals by real-time RT-PCR to verify and determine successful combinations of the above mentioned agents and stepwise procedure that produces enrichment in RPE-like cells or their progenitors. This approach can also be used to produce common progenitors of RPE and other eye tissues, such as photoreceptor or neural retina which can be isolated and further characterized for their differentiation potential and used in transplantation studies. Example 6 Derivation of RPE and other eye tissue progenitors from existing and new ES cell lines. Using the data from the gene expression profiling, expression of the RPE progenitor markers will be correlated with the expression of the surface proteins in order to find a unique combination of surface markers for RPE progenitor cells. If such markers are found, antibodies to surface proteins can be used to isolate a pure population of RPE progenitors that can be then cultured and further differentiated in culture or used in transplantation studies to allow their differentiation after grafting. If the data from the gene expression profiling experiments is insufficient, to isolate the RPE progenitors the following approach will be used. ES cells and RPE- like cells will be transfected with GFP under the control of a Pax6 promoter, and stable transfectants will be selected. From a culture of transfected differentiating ES cells or proliferating (dedifferentiated) RPE cells, GFP/Pax6-positive cells will be isolated by FACS and used as an antigen source for mouse injection to raise monoclonal antibodies to the surface molecules of Pax6 positive cells. Because Pax6 is present not only in RPE progenitors, screening will be done (by FACS) using several strategies: a) against proliferating RPE-like cells, b) against Pax2-positive RPE cells, c) against mitf-positive RPE cells. For b) and c) RPE cells will be transfected with GFP under the corresponding promoter; as a negative control, RPE or ES cells negative by these antigens will be used. After expansion of positive clones selected by all three strategies, antibodies will be tested against all types of cells used in screening and further analyzed: since this strategy can produce antibodies that recognize cell surface antigens specific and non-specific for RPE progenitors, the cells from differentiating total population of ES cells or of RPE cells selected with these antibodies will be assessed for molecular markers of RPE progenitors and for their ability to produce RPE. Using the optimized defined stepwise procedures to produce RPE or other early progenitors of eye tissues and the antibodies to their unique surface markers, such progenitors will be isolated from differentiated ES cells and cultured in vitro. Their ability to differentiate into various tissues of the eye will be investigated using the strategy described in Aim 2. Three ES cell lines that already produced RPE-like cells (H9, ACT J-l, Cyno-1), RPE-like cells will be used to continue to derive RPE-like cells and their progenitors as described in Aims 1 and 2, and HI and H7 hES cell lines will be used to produce new RPE-like cell lines. After expansion and characterization for molecular markers of RPE, these lines will be single-cloned, and the resulting lines will be characterized as described in Aim 1. The lines meeting criteria for RPE cells will be used for transplantation studies. New human ES cell lines will be derived from unused INF embryos, from donated oocytes, stimulated to develop without fertilization (parthenote), and from generated developing blastocysts obtained from donated oocytes with the application of nuclear transfer technology. RPE-like cells and common eye progenitors will be derived from these lines using the approach in Aim 2, and the resulting lines will be characterized as in Aim 1. [Optional] new human ES cell lines will be derived in a virus-free system, characterized and submitted for clinical trials.
Example 7 Therapeutic potential of RPE-like cells and progenitors in various animal models of retinitis pigmentosa & macular degeneration. Primate ES cells are tested in cynomologus monkeys (Macaques). Initially, vitrectomy surgery is performed and the cells are transplanted into the subretinal space of the animals. The first step is the transplantation of the cells in the suspension format after which a substrate or matrix is used to produce a monolayer transplantation. This can also be performed in immunosuppressed rabbits using cells derived from human ES-cells and also in various other animal models of retinitis pigmentosa, including 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 "rcdl, rcd2 & rcd3" dogs, photoreceptor dysplasia "pd" dog, and Briard "RPE-65) dog). Evaluation is performed using fluorescent angiography, histology (whether or not there is photoreceptor restoration and possibly ERG. Functional testing will also be carried out, including phagocytosis (photoreceptor fragments), vitamin A metabolism, tight junctions conductivity, and electron microscopy. Example 8 Direct differentiation of RPE cells from human embryo-derived cells. Human blastocyst-staged embryos are plated in the presence of murine or chick embryo fibroblasts with or without immunosurgery to remove the trophectoderm or directly plates on extracellular matrix protein-coated tissue cultureware. Instead of culturing and passaging the cells to produce a human ES cell line, the cells are directly differentiated. When hEDC cell cultures are allowed to overgrow on MEF in the absence of LIF, FGF and Plasmanate, they will form a thick multilayer of cells. (Alternate growth factors, media, and FBS can be used to alternate direct differentiation as is known to those skilled in the art.) About 6 weeks later, dark islands of cells will appear within the larger clusters. These dark cells are easily seen with the naked eye and looked like "freckles" in a plate of cells as shown in Fig 5B. At higher magnification these islands appear as tightly packed polygonal cells in a cobblestone monolayer, typical of epithelial cells, with brown pigment in the cytoplasm (Fig. 5A). There are differences in the amount of pigment in the cells with cells in the central part of the islands having the most pigment and those near the edges the least. (Fig. 5B). When hEDC cells are directly differentiated they may, though typically have not, fonned embryoid bodies (EB). Pigmented epithelial cells appear in about 1-2% of these differentiated cells and/or EBs in the first 6-8 weeks. Over time more and more EBs develop pigmented cells, and by 3 months nearly every EB had a pigmented epithelial region. Morphology of the cells in the pigmented regions of EBs was very similar to that of adherent cultures.
Materials and methods: MEF medium: high glucose DMEM, supplemented with 2mM GlutaMAX I, and 500 u/ml Penicillin, 500 ug/ml streptomycin (all from Invitrogen) and 16% FCS (HyCLone). hES Cells Growth medium: knockout high glucose DMEM supplemented with 500 u/ml Penicillin, 500 ug/mlstreptomycin, 1 % non-essential amino acids solution, 2mM GlutaMAX I„ 0.1 mM beta-mercaptoethanol, 4 ng/ml bFGF (Invitrogen), 1 -ng/ml human LIF (Chemicon, Temecula, CA), 8.4% of Serum Replacement (SR, Invitrogen) and 8.4% Plasmanate (Bayer). Derivation medium contained the same components as growth medium except that it had lower concentration of SR and Plasmanate (4.2% each) and 8.4 % FCS and 2x concentration of human LIF and bFGF, as compared to growth medium. EB medium: same as growth medium except bFGF, LIF, and Plasmanate; the SR concentration was 13%. RPE medium: 50% EB medium and 50% MEF medium. hES cell lines The cell lines, hES 35, 36, 45, used for these studies were derived with modifications of previously reported procedures (Thomson et. al., 1998, Reubinoff et. al, 2000, Lanzendorf et. al., 2001). Human frozen blastocysts (line hES35) or cleaved embryos (lines hES 36 and hES45) were donated to the study, approved by two institutional review board, by couples who had completed their fertility treatment. Differentiation experiments were performed with adherent hES cells or with embryoid bodies (EBs). For adherent differentiation, hES cells were allowed to overgrow on MEFs until the liES colonies lost their tight borders at which time the culture media was replaced with EB medium (usually, 8-10 days after passaging). The medium was changed every 1-2 days. For EB formation, hES cells were trypsinized and cultured in EB medium on low adherent plates (Costar).
Immunostaining Cells were fixed with 2% paraformaldehyde, permeabilized with 0.1% NP- 40 for localization of intracellular antigens, and blocked with 10% goat serum, 10% donkey serum (Jackson Immunoresearch Laboratories, West Grove, PA) in PBS (Invitrogen) for at least one hour. Incubation with primary antibodies was carried out overnight at 4oC, the secondary antibodies (Jackson Immunoresearch Laboratories, West Grove, PA) were added for one hour. Between all incubations specimens were washed with 0.1% Tween-20 (Sigma) in PBS 3-5 times, 10-15 minutes each wash. Specimens were mounted using Nectashield with DAPI (Vector Laboratories, Burlingame, CA) and observed under fluorescent microscope (Nikon). Localization of alkaline phosphatase was done either by Vector Red (Vector Laboratories, Burlingame, CA) to live cells or after the second wash during immunostaining according to manufacturer's instructions. Antibodies used: bestrophin (Novus Biologicals, Littleton, CO), anti-CRALBP antibody was a generous gift from Dr. Saari, University of Washington. Secondary antibodies were from Jackson Immunoresearch Laboratories, and Streptavidin-FITC was purchased from Amersham.
Isolation and passaging of RPE-like cells Adherent cultures of hES cells or EBs were rinsed with PBS twice and incubated in 0.25% Trypsin/1 mM EDTA (Invitrogen) at 37oC until the monolayer loosened. Cells from the pigmented regions were scraped off with a glass capillary, transferred to MEF medium, centrifuged at 200X g, and plated onto gelatin-coated plates in RPE medium. The medium was changed after the cells attached (usually in 1-2 days) and every 5-7 days after that; the cells were passaged every 2-4 weeks with 0.05% Trypsin/0.53mM EDTA (invitrogen).
Western blot and ELISA
Samples were prepared in Laemmli buffer (Laemmli, 1970), supplemented with 5% Mercaptoethanol and Protease hihibitor Cocktail (Roche), boiled for 5 minutes and loaded onto a 8-16% gradient gel (Bio-Rad, Hercules, CA) using a Mini-Protean apparatus; the gels were run at 25-30 mA per gel; proteins were transferred to a 0.2 Nitrocellulose membrane (Schleicher and Shull, Keene, NH) at 20 volt overnight. Blots were briefly stained with Ponceau Red (Sigma) to visualize the bands, washed with Milli-Q water, and blocked for 1 hour with 5% non-fat dry milk in 0.1% TBST (Bio-Rad). Primary antibodies to bestrophin, CRALBP or PEDF (Chemicon) were added for 2 hours followed by three 15-minute washes with TBST; peroxidase- conjugated secondary antibodies were added for 1 hour, and the washes were repeated. Blots were detected using ECL system with Super-Signal reagent (Pierce). PEDF ELISA was performed on cell lysates using PEDF ELISA kit (Chemicon) according to manufacturer's protocol.
Real-time RT-PCR Total RNA was purified from differentiating ES cultures by a two-step procedure Crude RNA was isolated using Trizol reagent (Invitrogen) and further purified on RNeazy minicolumns (Qiagen). The levels of RPE65 transcripts were monitored by real-time PCR using a commercial primer set for RPE65 detection (Assay on Demand # Hs00165642_ml, Applied Biosystems) and Quantitect Probe RT-PCR reagents (Qiagen), according to the manufacturer's (Qiagen) protocol.
Derivation and characterization of undifferentiated hES cell lines
Two female one male hES cell lines were used in these studies. Details on the derivation of these hES lines are reported elsewhere. All lines have been passaged more than 50 times during which time they maintain an undifferentiated colony morphology, high alkaline phosphatase activity, and express Oct-4, SSEA-3, SSEA- 4, TRA 1-60, and TRA 1-81 (data not shown). Two lines have normal karyotype (hES36, hES35), while there were both normal and aneuploid subpopulations in hES45. Upon spontaneous differentiation both in vitro and in teratomas all lines expressed the markers of three germ layers - muscle actin, alpha-fetoprotein, and tubulin beta III. Example 9 Use of transcript genomics to identify normal differentiated cells differentiated ex vivo. Transcriptomics - hES-cell derivatives are likely to play an important role in the future of regenerative medicine. Qualitative assessment of these and other stem cell derivatives remains a challenge that could be approached using functional genomics. We compared the transcriptional profile of hES-RPE vs. its in vivo counterpart, fetal RPE cells, which have been extensively researched for its transplantation value. Both profiles were then compared with previously published (Rogojina et. al., 2003) transcriptomics data on human RPE cell lines. The gene expression profile of our data set was compared to two human RPE cell lines (non-transformed ARPE-19 and transformed D407, Rogojina et. al., 2003) to determine whether hES-RPE have similar global transcriptional profiles. To account for common housekeeping genes expressed in all cells, we used publicly available Affymetrix data sets from undifferentiated hES cells (HI line, hi -hES, ~ sato et. al, 2003) and bronchial epithelial cells (BE, Wright et. al., 2004) as a control based on its common epithelial origin that would allow to exclude common housekeeping and epithelial genes and identify RPE-specific genes. There were similarities and differences between hES-RPE, hES-RPE-TD, ARPE-19, D407. The similarities were further demonstrated by analyzing the exclusive intersection between those genes present in hES-RPE/ARPE-19 but not in BE (1026 genes). To account for background, we compared this to the exclusive intersection of genes present in BE/hES-RPE, but not ARPE-19 (186 genes), which results in a five- to six-fold greater similarity in hES-RPE and ARPE-19 when compared to BE. D407/ARPE19 appear to lose RPE specific genes such as RPE65, Bestrophin, CRALBP, PEDF, which is typical of long-term passaged cells (figure 6). Further data mining revealed known RPE specific ontologies such as melanin biosynthesis, vision, retinol-binding, only in fetal RPE and ES-RPE but not ARPE19. Comparison of hES-RPE, ARPE-19 and D407 to their in vivo counterpart, freshly isolated human fetal RPE (feRPE), was in concordance with our previous data, demonstrating that the transcriptional identity of hES-RPE to human feRPE is significantly greater than D407 to fe RPE (2.3 fold difference- 849 genes/373 genes) and ARPE-19 to feRPE (1.6 fold difference - 588 genes/364 genes (Figure 5c/5d). The RPE specific markers identified above, which were only present in hES-RPE and not in ARPE-19 or D407 were also present in feRPE, demonstrating a higher similarity of hES-RPE to its in vivo counterpart than of the cultured RPE lines. Seven-hundred-and-eighty-four genes present in hES-RPE were absent in feRPE and ARPE-19 data sets. Since the retention of "stemness" genes could potentially cause transformation of hES derivatives into malignant teratomas if transplanted into patients, we created a conservative potential "stemness" genes data using currently available Affymetrix microarray data sets Abeyta et. al. 2004 Sato 2003). This resulted in a list of 3806 genes present in all 12 data sets (including common housekeeping genes), j Only 36 of the 784 genes present in the hES-RPE dats et but not feRPE-ARPE-19 were common to the 3806 potential stemness genes. None of these were known stemness genes such as Oct4, Sox2, TDGF1.
Example 10 , Use of RPE cells for treatment of Parkinson 's Disease. hRPE can be used as an alternative source of cells for cell therapy of Parkinson's Disease because they secrete L-DOPA. Studies have showed that such cells attached to gelatin-coated microcarriers can be successfully transplanted in hemiparkinsonian monkeys and produced notable improvements (10-50) thousand cells per target), and in FDA-approved trial started in 2000 the patients received hRPE intrastriatial transplants without adverse effects. One of the many advantages to the use of hES cell-derived RPE is that it circumvents the shortage of donor eye tissue. It also facilitates the use of gene therapy. Other Embodiments From the foregoing description, it will be apparent that variations and modifications may be made to the invention described herein to adopt it to various usages and conditions.

Claims

WE CLAIM:
1. A method of treating or preventing retinal degeneration, comprising use of a cell selected from the group consisting of at least one of: RPE cells, RPE- like cells, RPE or RPE-like progenitors derived from mammalian embryonic stem cells.
2. The method of claim 1, wherein the condition of retinal degeneration is selected from the group consisting of at least one of: retinitis pigmentosa and macular degeneration.
3. The method of claim 1, further comprising transplantation of the cell by vitrectomy surgery into the subretinal space of the eye.
4. The method of claim 3, wherein the cells are transplanted in a suspension, matrix, or substrate.
5. The method of claim 2, wherein the retinitis pigmentosa is associated with an animal model.
6. The method of claim 5, where in the animal model is selected from the group consisting of: rd mouse, RPE-65 knockout mouse, tubby-like mouse, RCS rat, Abyssinian cat, cone degeneration "cd" dog, progressive rod-cone degeneration "prcd" dog, early retinal degeneration "erd" dog, rod-cone dysplasia 1, 2 & 3 "rcdl, rcd2 and rcd3" dogs, photoreceptor dysplasia "pd" dog, and Briard "RPE-65" dog.
7. The method of claim 6, wherein the outcome of the therapy in the animal model is evaluated using one or more of behavioral tests, fluorescent angiography, histology, and functional testing such as measuring the ability of the cells to perform phagocytosis (photoreceptor fragments), vitamin A metabolism, tight junctions conductivity, or evaluation using electron microscopy.
8. A method for the spontaneous differentiation of hES cells into cells RPE, RPE-like, or RPE progenitor cells, said method comprising: a) allowing hES cell cultures to overgrow on MEF; b) allowing the hES cell cultures to form a thick multilayer of cells; c) culturing the hES cells; d) isolation and culture of the pigmented RPE, RPE-like, and/or RPE progenitor cells from the resultant cell cultures.
9. The method of claim 8, wherein the isolation and culture of the RPE- like cells comprises: a) digesting the cultured hES cells or EB's with an enzyme; b) selectively isolating the pigmented cells; c) plating the isolated cells on gelatin or laminin for 1-2 days to form primary cultures (P0); d) continued culture of the primary culture for a period of up to 3 weeks; and, e) isolation of the RPE-like cells.
10. The method of claim 9, where in the enzyme is selected from the group consisting of one or more of trypsin, collagenase, and dispase.
11. The method of claim 8, wherein the RPE cells are grown to establish a new RPE cell line.
12. The method of claim 11 , wherein the RPE cell line is differentiated into alternate lineages comprising treatment of the RPE cell line in culture with bFGF or FGF.
13. The method of claim 11, wherein the new RPE cell lines varies from the already established RPE cell lines in at least one of the characteristics selected from the group consisting of: growth rate, expression of pigment, de-differentiation in culture, and re-differentiation in culture, of RPE-like cells when they are derived from different ES cell lines.
14. A method for the derivation of RPE lines or precursors to RPE cells that have an increased ability to prevent neovascularization, said method comprising: a) aging a somatic cell from an animal such that telomerase is shortened wherein at least 10% of the normal replicative lifespan of the cell has been passed; and, b) using the somatic cell as a nuclear transfer donor cell to create cells that overexpress angiogenesis inhibitors, wherein the angiogenesis inhibitors can be Pigment Epithelium Derived Factor (PEDF/EPC-1).
15. The method of claim 14, wherein the somatic cells are genetically modified with exogenous genes that inhibit neovascularization.
16. The method of claim 8, wherein the RPE-like cells are derived form a bank of ES or embryo-derived cells with homozygosity in the HLA region such that
ES-derived cells have reduced complexity of their HLA antigens.
17. The method of claim 8, wherein the ES cells are derived from a human.
18. A method for the treatment of Parkinson' s disease comprising the transplantation of RPE-like cells or progenitor cells.
PCT/US2005/002273 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina WO2005070011A2 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
CA2555370A CA2555370C (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
KR1020067016837A KR101258292B1 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
CN2005800073590A CN1968608B (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
EP05711960A EP1708575A4 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
KR1020127008183A KR101398356B1 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
JP2006551392A JP2007522131A (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of retinal degenerative diseases
BRPI0507074-0A BRPI0507074A (en) 2004-01-23 2005-01-24 improved modalities for the treatment of degenerative retinal diseases
NZ548929A NZ548929A (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
KR1020137002345A KR20130025953A (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
AU2005207042A AU2005207042B2 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
EP11183613.6A EP2438816B1 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
IL177015A IL177015B (en) 2004-01-23 2006-07-20 Isolated human rpe cells and use thereof for the treatment of degenerative disease of the retina
US11/490,953 US7795025B2 (en) 2004-01-23 2006-07-21 Methods for producing enriched populations of human retinal pigment epithelium cells
US12/857,866 US9649340B2 (en) 2004-01-23 2010-08-17 Methods for producing enriched populations of human retinal pigment epithelium cells
AU2010249263A AU2010249263B2 (en) 2004-01-23 2010-12-09 Improved Modalities for the Treatment of Degenerative Diseases of the Retina
IL267126A IL267126B (en) 2004-01-23 2019-06-05 Isolated human rpe cells and use thereof for the treatment of degenerative diseases of the retina
IL291162A IL291162A (en) 2004-01-23 2022-03-07 Isolated human rpe cells and use thereof for the treatment of degenerative diseases of the retina

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US53896404P 2004-01-23 2004-01-23
US60/538,964 2004-01-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP22188897.7A Previously-Filed-Application EP4248751A3 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina
US11/490,953 Continuation US7795025B2 (en) 2004-01-23 2006-07-21 Methods for producing enriched populations of human retinal pigment epithelium cells

Publications (2)

Publication Number Publication Date
WO2005070011A2 true WO2005070011A2 (en) 2005-08-04
WO2005070011A3 WO2005070011A3 (en) 2005-10-13

Family

ID=34807248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/002273 WO2005070011A2 (en) 2004-01-23 2005-01-24 Improved modalities for the treatment of degenerative diseases of the retina

Country Status (15)

Country Link
US (6) US7736896B2 (en)
EP (8) EP2438814A3 (en)
JP (10) JP2007522131A (en)
KR (3) KR101398356B1 (en)
CN (3) CN102204930B (en)
AU (6) AU2005207042B2 (en)
BR (1) BRPI0507074A (en)
CA (2) CA2555370C (en)
ES (1) ES2721174T3 (en)
HK (3) HK1216068A1 (en)
IL (3) IL177015B (en)
MX (2) MX2020009456A (en)
NZ (1) NZ548929A (en)
SG (4) SG10201606441SA (en)
WO (1) WO2005070011A2 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101688178A (en) * 2007-04-18 2010-03-31 哈达锡特医学研究服务及发展有限公司 Stem cell-derived retinal pigment epithelium
US7838727B2 (en) 2004-11-04 2010-11-23 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
CN1966080B (en) * 2005-11-17 2011-06-08 李凌松 Neural stem cell injection for treating senile dementia and Parkinson's disease
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
EP2501802A2 (en) * 2009-11-17 2012-09-26 Advanced Cell Technology, Inc. Methods of producing human rpe cells and pharmaceutical preparations of human rpe cells
EP2612906A2 (en) 2006-05-03 2013-07-10 Advanced Cell Technology, Inc. Derivation of embryonic stem cells and embryo-derived cells
US8796021B2 (en) 2007-02-23 2014-08-05 Advanced Cell Technology, Inc. Blastomere culture to produce mammalian embryonic stem cells
US9040770B2 (en) 2004-01-23 2015-05-26 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
WO2015087231A1 (en) 2013-12-11 2015-06-18 Pfizer Limited Method for producing retinal pigment epithelial cells
US10077424B2 (en) 2007-10-12 2018-09-18 Astellas Institute For Regenerative Medicine Methods of producing RPE cells and compositions of RPE cells
WO2020218480A1 (en) 2019-04-26 2020-10-29 国立研究開発法人理化学研究所 Composite including neural retina, retinal pigment epithelial cells, and hydrogel, and method for producing same
WO2021086911A1 (en) 2019-10-30 2021-05-06 Astellas Institute For Regenerative Medicine Methods for producing retinal pigment epithelium cells
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
WO2022191216A1 (en) 2021-03-09 2022-09-15 国立研究開発法人理化学研究所 Method for producing hypoimmunogenic retinal pigment epithelial cells
WO2023201361A1 (en) 2022-04-15 2023-10-19 Aspen Neuroscience, Inc. Methods of classifying the differentiation state of cells and related compositions of differentiated cells

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8043614B2 (en) * 2004-03-09 2011-10-25 Ahlfors Jan-Eric W Autogenic living scaffolds and living tissue matrices: methods and uses thereof
US7541186B2 (en) * 2006-02-22 2009-06-02 University Of Washington Method of generating human retinal progenitors from embryonic stem cells
US9133435B2 (en) * 2007-01-18 2015-09-15 Riken Method for induction/differentiation into photoreceptor cell
EP2607477B1 (en) 2007-05-03 2020-09-23 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US8574567B2 (en) 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
US20090028831A1 (en) * 2007-07-23 2009-01-29 University Of Kentucky Research Foundation Stem cell regulator, compositions and methods of use
AU2015201435B2 (en) * 2007-10-12 2017-02-02 Astellas Institute For Regenerative Medicine Improved methods of producing RPE cells and compositions of RPE cells
EP2279247B1 (en) * 2008-04-22 2019-01-02 Regenerative Research Foundation Retinal pigment epithelial stem cells
WO2011028524A1 (en) 2009-08-24 2011-03-10 Wisconsin Alumni Research Foundation Substantially pure human retinal progenitor, forebrain progenitor, and retinal pigment epithelium cell cultures and methods of making the same
EP2593117B1 (en) 2010-07-12 2019-03-20 University of Southern California Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
DK2596119T3 (en) 2010-07-23 2021-07-26 Astellas Inst For Regenerative Medicine METHODS FOR DETECTING RARE SUBPOPULATIONS OF CELLS AND HIGHLY PURIFIED COMPOSITIONS OF CELLS
WO2013015835A1 (en) 2011-07-22 2013-01-31 Seven Networks, Inc. Mobile application traffic optimization
JP2012231786A (en) * 2011-04-18 2012-11-29 Kyushu Univ Method for evaluating susceptibility to food functional component and pharmaceutical by expression change of target gene
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
WO2012149468A2 (en) 2011-04-29 2012-11-01 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
EP2707479B1 (en) 2011-05-13 2018-01-10 The United States of America, as represented by The Secretary, Department of Health and Human Services Use of zscan4 and zscan4-dependent genes for direct reprogramming of somatic cells
EP3636286A1 (en) * 2011-05-18 2020-04-15 The Regents of The University of California Compositions and methods for treating retinal diseases
CN103007355B (en) * 2011-09-20 2014-08-13 同济大学 Hydrogel-nanometer fiber membrane, preparation method and uses thereof
SI2780022T1 (en) * 2011-11-14 2019-09-30 Astellas Institute For Regenerative Medicine Pharmaceutical preparations of human rpe cells and uses thereof
US8961956B2 (en) 2011-11-30 2015-02-24 Ocata Therapeutics, Inc. Mesenchymal stromal cells and uses related thereto
ES2690212T3 (en) 2011-11-30 2018-11-19 Astellas Institute For Regenerative Medicine Mesenchymal stromal cells and related uses
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
CA2864818C (en) * 2012-02-17 2022-05-03 The Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
US10519422B2 (en) 2012-02-29 2019-12-31 Riken Method of producing human retinal pigment epithelial cells
JP6292557B2 (en) * 2012-08-24 2018-03-14 国立研究開発法人理化学研究所 Method for producing retinal pigment epithelial cell sheet
US11241460B2 (en) 2013-03-15 2022-02-08 Astellas Institute For Regenerative Medicine Photoreceptors and photoreceptor progenitors produced from pluripotent stem cells
IL307568A (en) * 2013-03-15 2023-12-01 Astellas Inst For Regenerative Medicine Photoreceptors and photoreceptor progenitors produced from pluripotent stem cells
JP6473686B2 (en) * 2013-03-25 2019-02-20 公益財団法人神戸医療産業都市推進機構 Cell sorting method
EP2796545A1 (en) * 2013-04-26 2014-10-29 Université Pierre et Marie Curie (Paris 6) Methods for obtaining retinal progenitors, retinal pigmented epithelial cells and neural retinal cells
CN105814195A (en) 2013-11-27 2016-07-27 京都府公立大学法人 Application of laminin to corneal endothelial cell culture
EP3213761B1 (en) * 2014-10-31 2021-05-19 Kyoto Prefectural Public University Corporation Novel treatment of retina using laminin
CA2965770C (en) 2014-10-31 2024-02-13 Kyoto Prefectural Public University Corporation Treatment of cornea using laminin
KR102651291B1 (en) 2015-08-18 2024-03-26 아스텔라스 인스티튜트 포 리제너러티브 메디슨 Clinical formulations
EP3347457B1 (en) 2015-09-08 2021-10-27 FUJIFILM Cellular Dynamics, Inc. Macs-based purification of stem cell-derived retinal pigment epithelium
MY189778A (en) 2015-09-08 2022-03-04 Healios Kk Method for producing retinal pigment epithelial cells
CA3002157A1 (en) 2015-10-20 2017-04-27 FUJIFILM Cellular Dynamics, Inc. Methods for directed differentiation of pluripotent stem cells to immune cells
CN106609263B (en) * 2015-10-22 2020-04-07 同济大学 Method for efficiently inducing differentiation of pluripotent stem cells to retinal pigment epithelial cells
CN106609257B (en) * 2015-10-22 2020-04-10 同济大学 Method for efficiently separating in-vitro induced RPE (RPC-like protein) cells and RPC (RPC-like protein)
CN106609255B (en) * 2015-10-22 2019-11-05 同济大学 Cell suspension and its application containing PJ34 and retinal pigment epithelium
CN106609256B (en) * 2015-10-22 2020-04-07 同济大学 Method for inducing human embryonic stem cells to differentiate into retinal pigment epithelial cells in vitro
CN106282096A (en) * 2016-10-19 2017-01-04 江苏艾尔康生物医药科技有限公司 A kind of isolated culture method of human retinal pigment epithelial cells layer
CN110573610A (en) 2017-03-08 2019-12-13 大日本住友制药株式会社 Preparation method of retinal pigment epithelial cells
IL269363B2 (en) * 2017-03-16 2024-02-01 Lineage Cell Therapeutics Inc Methods for measuring therapeutic effects of retinal disease therapies
CA3075883A1 (en) * 2017-09-14 2019-03-21 Riken Method for amplifying cone photoreceptors or rod photoreceptors using dorsalization signal transmitter or ventralization signal transmitter
CN111726985B (en) * 2017-11-15 2022-06-10 弗里德里克·米谢尔生物医学研究所 Primate retinal pigment epithelial cell specific promoter
CN108029638B (en) * 2017-12-04 2020-04-21 四川省人民医院 Construction method and application of animal model of retinitis pigmentosa disease
US11946069B2 (en) 2018-03-12 2024-04-02 RxCell, Inc. Method for generating multiple cellular products from single pluripotent cell source
RU2730937C1 (en) * 2019-11-01 2020-08-26 Федеральное государственное бюджетное учреждение "Национальный медицинский исследовательский центр глазных болезней имени Гельмгольца" Министерства здравоохранения Российской Федерации (ФГБУ "НМИЦ ГБ им. Гельмгольца" Миндрава России) Method of transplantation of retinal pigment epithelium (rpe) cells differentiated from induced human pluripotent stem cells, with retinal pigment epithelium atrophy

Family Cites Families (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE196923T1 (en) 1993-04-30 2000-10-15 Photogenesis Inc TRANSPLANTATION OF THE RETINAL PIGMENTAL EPITHELIUM
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
WO1998030679A1 (en) 1997-01-10 1998-07-16 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
KR20010033834A (en) 1998-01-02 2001-04-25 타이탄 파마슈티칼스 인코퍼레이티드 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
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
CN100497598C (en) 1999-10-28 2009-06-10 马萨诸塞大学 Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
JP3454206B2 (en) * 1999-11-10 2003-10-06 三菱電機株式会社 Noise suppression device and noise suppression method
EP1248517A2 (en) * 2000-01-07 2002-10-16 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
WO2002016620A2 (en) 2000-08-19 2002-02-28 Axordia Limited Modulation of stem cell differentiation
US6576464B2 (en) 2000-11-27 2003-06-10 Geron Corporation Methods for providing differentiated stem cells
DE60114229T2 (en) 2000-11-29 2006-07-06 Allergan, Inc., Irvine PREVENTING TRANSPLANT DISCHARGE IN THE EYE
DE10108412B4 (en) * 2001-02-21 2006-03-09 Cevec Pharmaceuticals Gmbh Pigment epithelial cell of the eye, its preparation and use in the treatment of an ocular or nervous disease
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 (en) 2001-07-12 2011-10-17 제론 코포레이션 Cells of the cardiomyocyte lineage produced from human pluripotent stem cells
JP2005510232A (en) * 2001-11-26 2005-04-21 アドバンスド セル テクノロジー、インク. Production and use of reprogrammed human somatic cell nuclei and autologous and syngeneic human stem cells
JP2005519881A (en) 2001-12-11 2005-07-07 ファイブローゲン、インコーポレーテッド How to suppress eye processes
US20050222061A1 (en) 2002-04-18 2005-10-06 Schulte Ralf W Means and methods for the specific inhibition of genes in cells and tissue of the cns and/or eye
US7422736B2 (en) 2002-07-26 2008-09-09 Food Industry Research And Development Institute Somatic pluripotent cells
US7824671B2 (en) * 2002-10-04 2010-11-02 Tissuetech, Inc. Retinal pigment epithelial cell cultures on amniotic membrane and transplantation
WO2004034734A1 (en) * 2002-10-08 2004-04-22 Nec Corporation Array device and portable terminal
WO2004045374A2 (en) * 2002-11-14 2004-06-03 Ethicon Endo-Surgery, Inc. Methods and devices for detecting tissue cells
EP1599730A2 (en) * 2003-03-03 2005-11-30 Kouyama, Yoshihisa Methods and apparatus for use in detection and quantitation of various cell types and use of optical bio-disc for performing same
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
EP2438814A3 (en) * 2004-01-23 2012-10-03 Advanced Cell Technology, Inc. Improved modalities for the treatment of degenerative diseases of the retina
AU2005304978A1 (en) 2004-11-04 2006-05-18 Advanced Cell Technology, Inc. Derivation of embryonic stem cells
SG148171A1 (en) 2005-02-11 2008-12-31 Agency Science Tech & Res Methods of proliferating stem cells
KR100832592B1 (en) 2006-08-17 2008-05-27 박현숙 Method for co-culture of stem cells and feeder cells using a polymer membrane
EP2147094B2 (en) 2007-04-18 2018-02-21 Hadasit Medical Research Services & Development Limited Stem cell-derived retinal pigment epithelial cells
JP2008307007A (en) * 2007-06-15 2008-12-25 Bayer Schering Pharma Ag Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
CA2702386C (en) 2007-10-12 2018-07-24 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
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
US8652123B2 (en) * 2008-09-02 2014-02-18 Geoffrey C. GURTNER Methods and devices for improving the appearance of tissue
WO2011063005A2 (en) 2009-11-17 2011-05-26 Advanced Cell Technology, Inc. Methods of producing human rpe cells and pharmaceutical preparations of human rpe cells
DK2596119T3 (en) * 2010-07-23 2021-07-26 Astellas Inst For Regenerative Medicine METHODS FOR DETECTING RARE SUBPOPULATIONS OF CELLS AND HIGHLY PURIFIED COMPOSITIONS OF CELLS
EP2702135B1 (en) 2011-04-29 2019-04-17 University of Southern California Method of cryopreservation of stem cell-derived retinal pigment epithelial cells on polymeric substrate
SI2780022T1 (en) 2011-11-14 2019-09-30 Astellas Institute For Regenerative Medicine 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

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
HIRANO: "Generation of Structures Formed by Lens and Retinal Cells Differentiating from Embryonic Stem Cells", DEVELOPMENTAL DYNAMICS, vol. 228, no. 4, 1 December 2013 (2013-12-01), pages 664 - 671, XP008040482, DOI: doi:10.1002/dvdy.10425
KAWASAKI: "Generation of Dopaminergic Neurons and Pigmented Epithelia from Primate ES Cells by Stromal Cell-Derived Inducing Activity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, vol. 99, no. 3, 5 February 2002 (2002-02-05), pages 1580 - 1585, XP002971360, DOI: doi:10.1073/pnas.032662199
OOTA: "Induction of the Differentiation of Lentoids from Primate Embryonic Stem Cells", INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, vol. 44, no. 6, 1 June 2003 (2003-06-01), pages 2689 - 2693, XP002904369, DOI: doi:10.1167/iovs.02-1168
REUBINOFF: "Embryonic Stem Cell Lines from Human Blastocysts: Somatic Differentiation in Vitro", NATURE BIOTECHNOLOGY, vol. 18, no. 4, 1 April 2000 (2000-04-01), pages 399 - 404, XP002195338, DOI: doi:10.1038/74447
YING: "Conversion of Embryonic Stem Cells into Neuroectodermal Precursors in Adherent Monoculture", NATURE BIOTECHNOLOGY, vol. 21, no. 2, 1 February 2003 (2003-02-01), pages 183 - 186, XP002350599, DOI: doi:10.1038/nbt780
ZHAO: "Differentiation of Embryonic Stem Cells into Retinal Neurons", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 297, no. 2, 20 September 2002 (2002-09-20), pages 177 - 184, XP002544407, DOI: doi:10.1016/S0006-291X(02)02126-5

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US9040770B2 (en) 2004-01-23 2015-05-26 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
US9040039B2 (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
US9193950B2 (en) 2004-01-23 2015-11-24 Ocata Therapeutics, Inc. Modalities for the treatment of degenerative diseases of the retina
US9730962B2 (en) 2004-01-23 2017-08-15 Astellas Institute For Regenerative Medicine 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
US9650607B2 (en) 2004-01-23 2017-05-16 Astellas Institute For Regenerative Medicine 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
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
US7893315B2 (en) 2004-11-04 2011-02-22 Advanced Cell Technology, Inc. Derivation of embryonic stem cells and embryo-derived cells
US8642328B2 (en) 2004-11-04 2014-02-04 Advanced Cell Technology, Inc. Derivation of embryonic stem cells
US7838727B2 (en) 2004-11-04 2010-11-23 Advanced Cell Technology, Inc. Derivation of embryonic stem cells
US9617512B2 (en) 2004-11-04 2017-04-11 Astellas Institute For Regenerative Medicine Derivation of embryonic stem cells and embryo-derived cells
US9550974B2 (en) 2004-11-04 2017-01-24 Astellas Institute For Regenerative Medicine Derivation of embryonic stem cells
US10072243B2 (en) 2004-11-04 2018-09-11 Astellas Institute For Regenerative Medicine Derivation of embryonic stem cells and embryo-derived cells
CN1966080B (en) * 2005-11-17 2011-06-08 李凌松 Neural stem cell injection for treating senile dementia and Parkinson's disease
EP3133150A2 (en) 2006-05-03 2017-02-22 Astellas Institute for Regenerative Medicine Derivation of embryonic stem cells and embryo-derived cells
EP2612906A2 (en) 2006-05-03 2013-07-10 Advanced Cell Technology, Inc. Derivation of embryonic stem cells and embryo-derived cells
US8796021B2 (en) 2007-02-23 2014-08-05 Advanced Cell Technology, Inc. Blastomere culture to produce mammalian embryonic stem cells
EP3190178A1 (en) 2007-02-23 2017-07-12 Astellas Institute for Regenerative Medicine Highly efficient methods for reprogramming differentiated cells and for generating animals and embryonic stem cells from reprogrammed cells
EP2982744A1 (en) 2007-02-23 2016-02-10 Advanced Cell Technology, Inc. Highly efficient methods for reprogramming differentiated cells and for generating animals and embryonic stem cells from reprogrammed cells
US10584313B2 (en) 2007-02-23 2020-03-10 Astellas Institute For Regenerative Medicine Method of producing a differentiated mammalian cell comprising culturing a single mammalian blastomere
CN101688178A (en) * 2007-04-18 2010-03-31 哈达锡特医学研究服务及发展有限公司 Stem cell-derived retinal pigment epithelium
US10077424B2 (en) 2007-10-12 2018-09-18 Astellas Institute For Regenerative Medicine Methods of producing RPE cells and compositions of RPE cells
US20220049217A1 (en) * 2007-10-12 2022-02-17 Astellas Institute For Regenerative Medicine Methods of producing rpe cells and compositions of rpe cells
EP2501802A4 (en) * 2009-11-17 2013-08-21 Advanced Cell Tech Inc Methods of producing human rpe cells and pharmaceutical preparations of human rpe cells
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
EP2501802A2 (en) * 2009-11-17 2012-09-26 Advanced Cell Technology, Inc. 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
WO2015087231A1 (en) 2013-12-11 2015-06-18 Pfizer Limited Method for producing retinal pigment epithelial cells
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
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
WO2020218480A1 (en) 2019-04-26 2020-10-29 国立研究開発法人理化学研究所 Composite including neural retina, retinal pigment epithelial cells, and hydrogel, and method for producing same
WO2021086911A1 (en) 2019-10-30 2021-05-06 Astellas Institute For Regenerative Medicine Methods for producing retinal pigment epithelium cells
WO2022191216A1 (en) 2021-03-09 2022-09-15 国立研究開発法人理化学研究所 Method for producing hypoimmunogenic retinal pigment epithelial cells
WO2023201361A1 (en) 2022-04-15 2023-10-19 Aspen Neuroscience, Inc. Methods of classifying the differentiation state of cells and related compositions of differentiated cells

Also Published As

Publication number Publication date
AU2013201791B2 (en) 2016-08-18
JP2022153630A (en) 2022-10-12
CA2937099A1 (en) 2005-08-04
JP2007522131A (en) 2007-08-09
MX2020009456A (en) 2020-10-12
EP4269561A3 (en) 2024-01-03
KR20130025953A (en) 2013-03-12
AU2016259323B2 (en) 2019-01-31
JP2015096556A (en) 2015-05-21
SG10201912182UA (en) 2020-02-27
US20150328261A1 (en) 2015-11-19
EP2438816A3 (en) 2012-10-03
IL267126B (en) 2022-04-01
CA2555370C (en) 2024-02-06
EP2438815A2 (en) 2012-04-11
CN102204930B (en) 2014-12-03
US20110117062A1 (en) 2011-05-19
US20060018886A1 (en) 2006-01-26
AU2019200513A1 (en) 2019-02-14
EP2929782A1 (en) 2015-10-14
EP2438815A3 (en) 2012-10-03
US9649340B2 (en) 2017-05-16
KR101258292B1 (en) 2013-04-25
HK1216068A1 (en) 2016-10-14
AU2005207042B2 (en) 2010-09-09
CA2555370A1 (en) 2005-08-04
BRPI0507074A (en) 2007-06-19
AU2021200601A1 (en) 2021-03-04
SG155236A1 (en) 2009-09-30
IL177015A0 (en) 2006-12-10
EP4248751A2 (en) 2023-09-27
EP2438814A2 (en) 2012-04-11
US20070031386A1 (en) 2007-02-08
US9730962B2 (en) 2017-08-15
EP4248752A2 (en) 2023-09-27
CN102204930A (en) 2011-10-05
CN1968608B (en) 2011-06-29
CN104434979A (en) 2015-03-25
JP2020146575A (en) 2020-09-17
CN1968608A (en) 2007-05-23
HK1162931A1 (en) 2012-09-07
EP1708575A2 (en) 2006-10-11
HK1207821A1 (en) 2016-02-12
EP2438814A3 (en) 2012-10-03
US9040770B2 (en) 2015-05-26
NZ548929A (en) 2010-10-29
US7795025B2 (en) 2010-09-14
JP2024038460A (en) 2024-03-19
WO2005070011A3 (en) 2005-10-13
JP2019217356A (en) 2019-12-26
ES2721174T3 (en) 2019-07-29
EP4248752A3 (en) 2024-01-03
KR101398356B1 (en) 2014-05-23
KR20070069087A (en) 2007-07-02
US20100299765A1 (en) 2010-11-25
EP2438816B1 (en) 2019-03-27
EP4269561A2 (en) 2023-11-01
JP2018086274A (en) 2018-06-07
MX343606B (en) 2016-11-11
KR20120051754A (en) 2012-05-22
EP1708575A4 (en) 2009-07-01
IL177015B (en) 2019-06-30
AU2005207042A1 (en) 2005-08-04
SG188122A1 (en) 2013-03-28
IL267126A (en) 2019-08-29
IL291162A (en) 2022-05-01
JP2016195862A (en) 2016-11-24
AU2016259323A1 (en) 2016-12-01
AU2010249263A1 (en) 2011-01-06
AU2013201791A1 (en) 2013-04-11
US20180064761A1 (en) 2018-03-08
AU2010249263B2 (en) 2013-04-18
JP2012148085A (en) 2012-08-09
EP2438816A2 (en) 2012-04-11
EP4248751A3 (en) 2023-11-01
SG10201606441SA (en) 2016-09-29
JP2014079650A (en) 2014-05-08
US7736896B2 (en) 2010-06-15

Similar Documents

Publication Publication Date Title
US20210102164A1 (en) Modalities for the treatment of degenerative diseases of the retina
AU2016259323B2 (en) Improved Modalities for the Treatment of Degenerative Diseases of the Retina

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 177015

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/008299

Country of ref document: MX

Ref document number: 2006551392

Country of ref document: JP

Ref document number: 11490953

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2555370

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2005207042

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005711960

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 548929

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2005207042

Country of ref document: AU

Date of ref document: 20050124

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005207042

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1020067016837

Country of ref document: KR

Ref document number: 2373/KOLNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 200580007359.0

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005711960

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11490953

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0507074

Country of ref document: BR