WO2007079183A2 - Placental stem cell populations - Google Patents

Placental stem cell populations Download PDF

Info

Publication number
WO2007079183A2
WO2007079183A2 PCT/US2006/049491 US2006049491W WO2007079183A2 WO 2007079183 A2 WO2007079183 A2 WO 2007079183A2 US 2006049491 W US2006049491 W US 2006049491W WO 2007079183 A2 WO2007079183 A2 WO 2007079183A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
placental
stem cells
population
cell
Prior art date
Application number
PCT/US2006/049491
Other languages
French (fr)
Other versions
WO2007079183A3 (en
Inventor
Sascha Dawn Abramson
James W. Edinger
Herbert Faleck
Robert J. Hariri
Kristen S. Labazzo
Marian Periera
Jia-Lun Wang
Qian Ye
Original Assignee
Anthrogenesis Corporation
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 KR1020177008335A priority Critical patent/KR101900809B1/en
Priority to NZ595786A priority patent/NZ595786A/en
Priority to KR1020187026653A priority patent/KR20180105266A/en
Priority to KR1020197025129A priority patent/KR20190104428A/en
Priority to EP12150934.3A priority patent/EP2471907B1/en
Priority to EP12150924.4A priority patent/EP2471905B1/en
Priority to EP12150919.4A priority patent/EP2471904B1/en
Priority to PL12150934T priority patent/PL2471907T3/en
Priority to PL12150928T priority patent/PL2471906T3/en
Priority to EP18196002.2A priority patent/EP3486314B1/en
Priority to KR1020227015349A priority patent/KR20220063304A/en
Priority to EP06848281.9A priority patent/EP1976977B1/en
Priority to KR1020237019606A priority patent/KR20230091193A/en
Priority to AU2006332678A priority patent/AU2006332678B2/en
Priority to PL12150919T priority patent/PL2471904T3/en
Priority to EP12150928.5A priority patent/EP2471906B1/en
Priority to PL06848281T priority patent/PL1976977T3/en
Priority to KR1020207030297A priority patent/KR20200123283A/en
Application filed by Anthrogenesis Corporation filed Critical Anthrogenesis Corporation
Priority to ES06848281.9T priority patent/ES2549111T3/en
Priority to KR1020217031368A priority patent/KR20210122908A/en
Priority to KR1020157020367A priority patent/KR101722577B1/en
Priority to PL12150924T priority patent/PL2471905T3/en
Priority to CA2635253A priority patent/CA2635253C/en
Priority to JP2008548745A priority patent/JP5550235B2/en
Priority to NZ569562A priority patent/NZ569562A/en
Priority to CN200680053575.3A priority patent/CN101395266B/en
Publication of WO2007079183A2 publication Critical patent/WO2007079183A2/en
Publication of WO2007079183A3 publication Critical patent/WO2007079183A3/en
Priority to IL192457A priority patent/IL192457A0/en
Priority to IL243876A priority patent/IL243876B/en
Priority to IL223156A priority patent/IL223156A0/en
Priority to IL223194A priority patent/IL223194A/en
Priority to AU2014201181A priority patent/AU2014201181B2/en
Priority to IL243876A priority patent/IL243876A0/en
Priority to AU2017248523A priority patent/AU2017248523A1/en
Priority to IL266958A priority patent/IL266958A/en
Priority to AU2019268061A priority patent/AU2019268061A1/en
Priority to AU2022201955A priority patent/AU2022201955A1/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
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem 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
    • A61K35/56Materials from animals other than mammals
    • A61K35/63Arthropods
    • A61K35/64Insects, e.g. bees, wasps or fleas
    • A61K35/644Beeswax; Propolis; Royal jelly; Honey
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • 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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0605Cells from extra-embryonic tissues, e.g. placenta, amnion, yolk sac, Wharton's jelly
    • 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
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • 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
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/36Lipids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/42Organic phosphate, e.g. beta glycerophosphate
    • 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
    • C12N2501/11Epidermal growth factor [EGF]
    • 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
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • 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
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/52Fibronectin; Laminin

Definitions

  • the present invention provides isolated placental stem cells, populations of placental stem cells, compositions comprising the stem cells, and methods of obtaining the stem cells.
  • Human stem cells are totipotential or pluripotential precursor cells capable of generating a variety of mature human cell lineages. Evidence exists that demonstrates that stem cells can be employed to repopulate many, if not all, tissues and restore physiologic and anatomic functionality.
  • the present invention provides isolated placental stem cells, populations of placental stem cells, compositions comprising the stem cells, and methods of obtaining the stem cells.
  • the invention first provides isolated stem cells, and cell populations comprising such stem cells, wherein the stem cells are present in, and isolatable from placental tissue ⁇ e.g., amnion, chorion, placental cotyledons, etc.)
  • placental stem cells exhibit one or more characteristics of a stem cell ⁇ e.g., exhibit markers associated with stem cells, replicate at least 10-20 times in culture in an undifferentiated state, differentiate into adult cells representative of the three germ layers, etc.), and can adhere to a tissue culture substrate ⁇ e.g., tissue culture plastic such as the surface of a tissue culture dish or multiwell plate).
  • the invention provides an isolated placental stem cell that is CQ200 + or HLA-G + .
  • said cell is CD200 + and HLA-G + .
  • said stem cell is CD73 + and CD105 + .
  • said stem cell is CD34 " , CD38 “ or CD45 " .
  • said stem cell is CD34 " , CD38 " and CD45 ⁇
  • said stem cell is CD34 " , CD38 " , CD45 ⁇ , CD73 + and CD105 + .
  • said stem cell facilitates the formation of one or more embryoid-like bodies from a population of isolated placental cells comprising placental stem cells when said population is cultured under conditions that allow formation of embryoid-like bodies.
  • the invention provides a population of isolated placental cells comprising, e.g., that is enriched for, CD200 + , HLA-G + stem cells.
  • at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% or more of said isolated placental cells are CD200 + , HLA-G + stem cells.
  • said stem cells are CD73 + and CD105 + .
  • said stem cells are CD34 ⁇ , CD38 ⁇ or CD45 " .
  • said stem cells are CD34 " , CD3S " , CD45 ⁇ , CD73 + and CD105 + .
  • said population has been expanded, e.g., passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • said population forms one or more embryoid-like bodies when cultured under conditions that allow formation of embryoid-like bodies.
  • the invention provides an isolated stem cell that is CD73 + , CD105 + , and CD200 + .
  • said stem cell is HLA-G + .
  • said stem cell is CD34 " , CD38 ⁇ or CD45 ⁇ .
  • said stem cell is CD34 ⁇ , CD38 " and CD45 " .
  • said stem cell is CD34 ⁇ , CD38 ⁇ , CD45 ⁇ , and HLA-G + .
  • said stem cell facilitates development of one or more embryoid-like bodies from a population of isolated placental cells comprising the stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies.
  • the invention provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73 + , CD105 + , CD200 + stem cells.
  • at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD73 + , CDl 05 + , CD200 + stem cells.
  • said stem cells are HLA-G + .
  • said stem cells are CD34 , CD38 " or CD45 " .
  • said stem cells are CD34 ⁇ , CD38 " and CD45 " .
  • said stem cells are CD34 " , CD38 " , CD45 ⁇ , and HLA-G + .
  • said population has been expanded, for example, passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • said population forms one or more embryoid-like bodies in culture under conditions that allow formation of embryoid-like bodies.
  • the invention also provides an isolated stem cell that is CD200 + and OCT-4 + .
  • the stem cell is CD73 + and CD105 + .
  • said stem cell is HLA-G + .
  • said stem cell is CD34 ⁇ , CD38 ⁇ or CD45 " .
  • said stem cell is CD34 ⁇ , CD38 " and CD45 ⁇ .
  • said stem cell is CD34 " , CD38 " , CD45 " , CD73 + , CDl 05 + and HLA-G + .
  • said stem cell facilitates the formation of one or more embryoid-like bodies from a population of isolated placental cells comprising placental stem cells when said population is cultured under conditions that allow formation of embryoid-like bodies.
  • the invention provides a population of isolated cells comprising, e.g., that is enriched for, CD200 + , OCT-4 + stem cells.
  • at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD200 + , OCT- 4 + stem cells.
  • said stem cells are CD73 + and CD105 + .
  • said stem cells are HLA-G + .
  • said stem cells are CD34 " , CD38 ⁇ and CD45 ⁇ ⁇
  • said stem cells are CD34 " , CD38 " CD45 " , CD73 + , CD105 + and HLA-G + .
  • said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • said population forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • the invention provides an isolated stem cell that is CD73 + and CDl 05 + and which facilitates the formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies.
  • said stem cell is CD34 , CD38 or CD45 .
  • said stem cell is CD34 " , CD38 " and CD45 ⁇ .
  • said stem cell is OCT4 + .
  • said stem cell is OCT4+, CD34 " , CD38 ⁇ and CD45 " .
  • the invention further provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73 + , CD105 + stem cells, wherein said population forms one or more embryoid-like bodies under conditions that allow formation of embryoid-like bodies.
  • at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD73 + , CD105 + stem cells.
  • said stem cells are CD34 ⁇ , CD38 ⁇ or CD45 ⁇ .
  • said stem cells are CD34 " , CD38 ⁇ and CD45 ⁇ .
  • said stem cells are OCT-4 + .
  • said stem cells are OCT-4 + , CD34 ⁇ , CD38 ⁇ and CD45 " .
  • said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • the invention further provides an isolated stem cell that is CD73 + , CDlOS + and HLA- G + .
  • said stem cell is CD34 ⁇ , CD38 ⁇ or CD45 ⁇ .
  • said stem cell is CD34 ⁇ , CD38 ⁇ and CD45 ⁇ .
  • said stem cell is OCT-4 + .
  • said stem cell is CD200 + .
  • said stem cell is CD34 " , CD38 " , CD45 " , OCT-4 + and CD200 + .
  • said stem cell facilitates the formation of one or more embryoid-like bodies from a population of isolated placental cells comprising placental stem cells in culture under conditions that allow formation of embryoid-like bodies.
  • the invention further provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73 + , CD105 + and HLA-G + stem cells.
  • CD73 + , CD105 + and HLA-G + stem cells are CD73 + , CD105 + and HLA-G + stem cells.
  • said stem cells are CD34 ⁇ , CD38 ⁇ or CD45 " .
  • said stem cells are CD34 ⁇ , CD38 ⁇ and CD45 " .
  • said stem cells are OCT-4 + .
  • said stem cells are CD200 + .
  • said stem cells are CD34 " , CD38 " , CD45 " , OCT-4 + and CD200 + .
  • said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • said population forms embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • the invention further provides an isolated stem cell that is OCT-4 + and which facilitates formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when cultured under conditions that allow formation of embryoid-like bodies.
  • said stem cell is CD73 + and CDl 05 + .
  • said stem cell is CD34 ⁇ , CD38 ⁇ , or CD45 " .
  • said stem cell is CD200 + .
  • said stem cell is CD73 + , CDl 05 + , CD200 + , CD34 " , CD38 " , and CD45 ⁇ .
  • the invention also provides a population of isolated cells comprising, e.g., that is enriched for, OCT-4 + placental stem cells, wherein said population forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid- like bodies.
  • at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are OCT4 + placental stem cells.
  • said stem cells are CD73 + and CD105 + .
  • said stem cells are CD34 " , CD38 ⁇ , or CD45 ⁇ .
  • said stem cells are CD200 + .
  • said stem cells are CD73 + , CD105 "1" , CD200 + , CD34 " ⁇ , CD38 ⁇ , and CD45 " .
  • said population has been expanded, for example, passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • the invention further provides an isolated population of the placental stem cells described herein that is produced according to a method comprising perfusing a mammalian placenta that has been drained of cord blood and perfused to remove residual blood; perfusing said placenta with a perfusion solution; and collecting said perfusion solution, wherein said perfusion solution after perfusion comprises a population of placental cells that comprises placental stem cells; and isolating a plurality of said placental stem cells from said population of cells.
  • the perfusion solution is passed through both the umbilical vein and umbilical arteries and collected after it exudes from the placenta.
  • the perfusion solution is passed through the umbilical vein and collected from the umbilical arteries, or passed through the umbilical arteries and collected from the umbilical vein.
  • the invention further provides an isolated population of the placental stem cells described herein that is produced according to a method comprising digesting placental tissue with a tissue-disrupting enzyme to obtain a population of placental cells comprising placental stem cells, and isolating a plurality of placental stem cells from the remainder of said placental cells.
  • said placental tissue is a whole placenta, an amniotic membrane, chorion, a combination of amnion and chorion, or a combination of any of the foregoing.
  • the tissue-disrupting enzyme is trypsin or collagenase.
  • the invention provides any of the isolated stem cells above, wherein said stem cell expresses one or more genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, ILl 8, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PJP2, RTNl 5 SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, T
  • said stem cell expresses ACTG2, ADARBl, AMIG02, ARTS-I, B4GALT6, BCHE, Cl lor ⁇ , CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJ10781, GAT A6, GPRl 26, GPRC5B, HLA-G, ICAMl , IER3, IGFBP7, ILIA, IL6, IL 18, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl 5 PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12AS, TCF21, TGFB2, VTN, and ZC3H12A at a detectably higher level than a bone marrow-derived mesenchymal stem cell.
  • the invention also provides any of the populations of isolated stem cells above, wherein said stem cells express one or more genes at a detectably higher level than a population of bone marrow-derived mesenchymal stem cells, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl , AMIG02, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CPA4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, ILl 8, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12
  • the population of isolated stem cells expresses ACTG2, ADARBl 5 AMIGO2, ARTS-I, B4GALT6, BCHE, Cl lorfP, CD200, COL4A1 , COL4A2, CPA4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G 5 ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A at a detectably higher level than said population of isolated bone marrow-derived mesenchymal stem cells.
  • the invention also provides methods of selecting one of the above-mentioned cell populations, comprising selecting cells that express one or more genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJ 10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILlA 5 IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL
  • said selecting comprises selecting cells that express ACTG2, ADARBl 5 AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl 5 IER3, IGFBP7, ILIA, IL6, ILl 8, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl 5 PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLCl 2 A8, TCF21, TGFB2, VTN and ZC3H12A at a detectably higher level than a bone marrow-derived mesenchymal stem cell.
  • the invention also provides compositions that comprise one or more of the stem cells of the invention, wherein the stem cell has been isolated from the placenta.
  • the invention further provides a composition comprising a stem cell, wherein said stem cell is CD200 + and HLA-G + .
  • said stem cell is CD73 + and CD105 + .
  • said stem cell is CD34 " , CD38 ⁇ or CD45 ⁇ .
  • said stem cell is CD34 ⁇ , CD38 ⁇ and CD45 " .
  • said stem cell is CD34 " CD38 ⁇ , CD45 " , CD73 + , CD105 + , CD200 + and HLA-G + .
  • the invention provides a composition comprising a stem cell, wherein said stem cell is CD73 + , CDlOS + and CD200 + .
  • said stem cell is HLA-G + .
  • said stem cell is CD34 ⁇ , CD38 ⁇ or CD45 " .
  • said stem cell is CD34 ⁇ , CD38 ⁇ and CD45 ⁇ .
  • said stem cell is CD34 " , CD38 ⁇ , CD45 " and HLA-G + .
  • the invention provides a composition comprising a stem cell, wherein said stem cell is CD200 + and OCT-4 + .
  • said stem cell is CD73 + and CD105 + . In another specific embodiment, said stem cell is HLA-G + . In another specific embodiment, said stem cell is CD34 ⁇ , CD38 ⁇ or CD45 ⁇ . In another specific embodiment, said stem cell is CD34 " , CD38 ⁇ and CD45 ⁇ . In another specific embodiment, said stem cell is CD34 ' , CD38 " , CD45 " , CD73 + , CD105 + , and HLA-G + .
  • the invention provides a composition comprising a stem cell that is CD73 + and CD105 + , wherein said stem cell facilitates formation of an embryoid-like body in a population of isolated placental cells comprising said stem cell under conditions that allow the formation of an embryoid-like body.
  • said stem cell is CD34 " , CD38 ⁇ or CD45 ⁇ .
  • said stem cell is OCT-4 + .
  • said stem cell is CD200 + .
  • said stem cell is OCT-4+, CD200 + , CD34 " , CD38 " and CD45 " .
  • the invention provides a composition comprising a stem cell that is CD73 + , CDl 05 + and HLA-G + .
  • said stem cell is CD34 ⁇ , CD38 " or CD45 .
  • said stem cell is OCT-4 + .
  • said stem cell is CD200 + .
  • said stem cell is OCT-4+, CD200 + , CD34 " , CD38 ⁇ and CD45 " .
  • the invention provides a composition comprising a stem cell that is OCT-4 "1" , wherein said stem cell facilitates formation of an embryoid-Iike body in a population of isolated placental cells comprising said stem cell under conditions that allow the formation of an embryoid-like body.
  • said stem cell is CD73 + and CD105 + .
  • said stem cell is CD34 " , CD38 " and CD45 ⁇ .
  • said stem cell is CD200 .
  • said stem cell is CD73 + , CDl 05 + , CD200 + , CD34 " , CD38 “ and CD45 " .
  • said stem cell expresses one or more genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1 , COL4A2, CPA4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJl 0781, GATA6, GPRl 26, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, ILl 8, KRTI 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB
  • said stem cells express ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD 5 DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRTI8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A at a detectably higher level than a population of isolated bone marrow-derived mesenchymal stem cell, wherein said population of stem cells and said population of bone
  • any of the foregoing compositions comprises a matrix.
  • said matrix is a three-dimensional scaffold.
  • said matrix comprises collagen, gelatin, laminin, fibronectin, pectin, ornithine, or vitronectin.
  • the matrix is an amniotic membrane or an amniotic membrane-derived biomaterial.
  • said matrix comprises an extracellular membrane protein.
  • said matrix comprises a synthetic compound.
  • said matrix comprises a bioactive compound.
  • said bioactive compound is a growth factor, cytokine, antibody, or organic molecule of less than 5,000 daltons.
  • the invention further provides a composition comprising medium conditioned by any of the foregoing stem cells, or any of the foregoing stem cell populations.
  • any such composition comprises a stem cell that is not derived from a placenta.
  • said stem cell is an embryonic stem cell.
  • said stem cell is a mesenchymal stem cell.
  • said stem cell is a bone marrow-derived stem cell.
  • said stem cell is a hematopoietic progenitor cell.
  • said stem cell is a somatic stem cell.
  • said somatic stem cell is a neural stem cell, a hepatic stem cell, a pancreatic stem cell, an endothelial stem cell, a cardiac stem cell, or a muscle stem cell.
  • the invention also provides methods for producing populations of stem cells derived from mammalian placenta.
  • the invention provides a method of producing a cell population comprising selecting cells that (a) adhere to a substrate, and (b) express CD200 and HLA-G; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, and (b) express CD73, CDl 05, and CD200; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate and (b) express CD200 and OCT-4; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, (b) express CD73 and CDl 05, and (c) facilitate the formation of one or more embryoid-like bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, and (b) express CD73, CDl 05 and HLA-G; and isolating said cells from other cells to form a cell population.
  • the invention also provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, (b) express OCT-4, and (c) facilitate the formation of one or more embryoid-like bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies; and isolating said cells from other cells to form a cell population.
  • said substrate comprises fibronectin.
  • the methods comprise selecting cells that express ABC-p.
  • the methods comprise selecting cells exhibiting at least one characteristic specific to a mesenchymal stem cell.
  • said characteristic specific to a mesenchymal stem cell is expression of CD29, expression of CD44, expression of CD90, or expression of a combination of the foregoing.
  • said selecting is accomplished using an antibody.
  • said selecting is accomplished using flow cytometry.
  • said selecting is accomplished using magnetic beads.
  • said selecting is accomplished by fluorescence-activated cell sorting.
  • said cell population is expanded.
  • the invention also provides a method of producing a stem cell line, comprising transforming a stem cell with a DNA sequence that encodes a growth-promoting protein; and exposing said stem cell to conditions that promote production of said growth-promoting protein.
  • said growth-promoting protein is v-myc, N-myc, c-myc, p53, SV40 large T antigen, polyoma large T antigen, EIa adenovirus or human papillomavirus E7 protein.
  • said DNA sequence is regulatable.
  • said DNA sequence is regulatable by tetracycline.
  • said growth-promoting protein has a regulatable activity.
  • said growth-promoting protein is a temperature-sensitive mutant.
  • the invention further provides cryopreserved stem cell populations.
  • the invention provides a population of CD200 + , HLA-G + stem cells, wherein said cells have been cryopreserved, and wherein said population is contained within a container.
  • the invention also provides a population of CD73 + , CD105 "1" , CD200 + stem cells, wherein said stem cells have been cryopreserved, and wherein said population is contained within a container.
  • the invention also provides a population of CD200 + , OCT-4 + stem cells, wherein said stem cells have been cryopreserved, and wherein said population is contained within a container.
  • the invention also provides a population of CD73 + , CD105 + stem cells, wherein said cells have been cryopreserved, and wherein said population is contained within a container, and wherein said stem cells facilitate the formation of one or more embryo id-like bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies.
  • the invention further provides a population of CD73 + , CD105 + , HLA-G + stem cells, wherein said cells have been cryopreserved, and wherein said population is contained within a container.
  • the invention also provides a population of OCT-4 + stem cells, wherein said cells have been cryopreserved, wherein said population is contained within a container, and wherein said stem cells facilitate the formation of one or more embryoid-iike bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies.
  • said container is a bag.
  • said population comprises about, at least, or at most 1 x 10 6 said stem cells, 5 x 10 6 said stem cells, 1 x 10 7 said stem cells, 5 x 10 7 said stem cells, 1 x 10 8 said stem cells, 5 x 10 8 said stem cells, 1 x 10 9 said stem cells, 5 x 10 9 said stem cells, or 1 x 10 10 said stem cells.
  • said stem cells have been passaged about, at least, or no more than 5 times, no more than 10 times, no more than 15 times, or no more than 20 times.
  • said stem cells have been expanded within said container.
  • the term “SH2" refers to an antibody that binds an epitope on the marker CD 105. Thus, cells that are referred to as SH2 + are CD105 + .
  • the terms “SH3” and SH4" refer to antibodies that bind epitopes present on the marker CD73. Thus, cells that are referred to as SH3 + and/or SH4 + are CD73 + .
  • the term “isolated stem cell” means a stem cell that is substantially separated from other, non-stem cells of the tissue, e.g., placenta, from which the stem cell is derived.
  • a stem cell is "isolated” if at least 50%, 60%, 70%, 80%, 90%, 95%, or at least 99% of the non-stem cells with which the stem cell is naturally associated, or stem cells displaying a different marker profile, are removed from the stem cell, e.g., during collection and/or culture of the stem cell.
  • the term "population of isolated cells” means a population of cells that is substantially separated from other cells of the tissue, e.g., placenta, from which the population of cells is derived.
  • a stem cell is "isolated” if at least 50%, 60%, 70%, 80%, 90%, 95%, or at least 99% of the cells with which the population of cells, or cells from which the population of cells is derived, is naturally associated, i.e., stem cells displaying a different marker profile, are removed from the stem cell, e.g., during collection and/or culture of the stem cell.
  • placental stem cell refers to a stem cell or progenitor cell that is derived from a mammalian placenta, regardless of morphology, cell surface markers, or the number of passages after a primary culture.
  • placental stem cell does not, however, refer to a trophoblast.
  • a cell is considered a “stem cell” if the cell retains at least one attribute of a stem cell, e.g., a marker or gene expression profile associated with one or more types of stem cells; the ability to replicate at least 10-40 times in culture, the ability to differentiate into cells of all three germ layers; the lack of adult (i.e., differentiated) cell characteristics, or the like.
  • the terms “placental stem cell” and “placenta- derived stem cell” may be used interchangeably.
  • a stem cell is "positive" for a particular marker when that marker is detectable above background.
  • a placental stem cell is positive for, e.g., CD73 because CD73 is detectable on placental stem cells in an amount detectably greater than background (in comparison to, e.g., an isotype control).
  • a cell is also positive for a marker when that marker can be used to distinguish the cell from at least one other cell type, or can be used to select or isolate the cell when present or expressed by the cell.
  • a particular cell surface marker in the context of, e.g., antibody-mediated detection, "positive,” as an indication a particular cell surface marker is present, means that the marker is detectable using an antibody, e.g., a fluorescently-labeled antibody, specific for that marker; "positive” also means that a cell bears that marker in a amount that produces a signal, e.g., in a cytometer, that is detectably above background.
  • a cell is "CD200 + " where the cell is detectably labeled with an antibody specific to CD200, and the signal from the antibody is detectably higher than a control (e.g., background).
  • a control e.g., background
  • a cell is "CD34 ⁇ " where the cell is not detectably labeled with an antibody specific to CD34.
  • cluster of differentiation (“CD") markers are detected using antibodies.
  • OCT-4 is determined to be present, and a cell is "OCT-4 + " if OCT-4 is detectable using RT-PCR. 4.
  • FIG. 1 Viability of placental stem cells from perfusion (A), amnion (B), chorion (C), amnion-chorion plate (D) or umbilical cord (E). Numbers on X-axis designate placenta from which stem cells were obtained.
  • FIG. 2 Percent HLA ABC7CD457CD347CD133 + cells from perfusion (A), amnion
  • FACSCalibur Numbers on X-axis designate placenta from which stem cells were obtained.
  • FIG. 3 Percent HLA ABC7CD457CD347CD133 + cells from perfusion (A), amnion
  • FIG. 4 HLA-G 5 CDlO 5 CDl 3, CD33, CD38, CD44, CD90, CD 105, CDl 17, CD200 expression in stem cells derived from placental perfusate.
  • FIG. 5 HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from amnion.
  • FIG. 6 HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from chorion.
  • FIG. 7 HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from amnion-chorion plate.
  • FIG. 8 HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from umbilical cord.
  • FIG. 9 Average expression of HLA-G, CDlO, CD 13, CD33, CD38, CD44, CD90,
  • FIG. 10 Culture time courses for amnion/chorion (AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (NHDF) cell lines used in this study. All cultures were grown and propagated using the same seeding and passage densities. Circles indicate which cultures were used for RNA isolation. Late cultures were harvested just prior to senescence. Two UC cultures were harvested at 38 doublings (UC-38) to compare the effect of trypsinization on gene expression. AU other cultures were lysed directly in their culture flasks prior to RNA isolation.
  • AC amnion/chorion
  • UC umbilical cord
  • BM-MSC bone marrow-derived stem cell
  • NHDF human dermal fibroblast
  • FIG. 11 Line plot of relative expression levels of 8215 genes in amnion/chorion
  • AC umbilical cord
  • BM-MSC bone marrow-derived stem cell
  • DF human dermal fibroblast
  • FIG. 12 Subset of the all genes list showing genes over-expressed > 6-fold in AC-03 for amnion/chorion (AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (DF) cells.
  • the number associated with each cell line designation on the X-axis indicates the number of days the cell line was cultured prior to evaluation of gene expression levels.
  • the chart was generated from RNA expression data analyzed by GeneSpring software. AC-03 was used as the selected condition.
  • Placental stem cell-specific or umbilical cord stem cell-specific genes found by fold change filtering for amnion/chorion (AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (DF) cells.
  • the number associated with each cell line designation on the X-axis indicates the number of days the cell line was cultured prior to evaluation of gene expression levels.
  • the chart was generated from RNA expression data analyzed by GeneSpring software. AC-03 was used as the selected condition.
  • Placental stem cells are stem cells, obtainable from a placenta or part thereof, that adhere to a tissue culture substrate and have the capacity to differentiate into non-placental cell types.
  • Placental stem cells can be either fetal or maternal in origin (that is, can have the genotype of either the fetus or mother, respectively).
  • the placental stem cells and placental stem cell populations of the invention are fetal in origin.
  • Populations of placental stem cells, or populations of cells comprising placental stem cells can comprise placental stem cells that are solely fetal or maternal in origin, or can comprise a mixed population of placental stem cells of both fetal and maternal origin.
  • the placental stem cells, and populations of cells comprising the placental stem cells can be identified and selected by the morphological, marker, and culture characteristic discussed below.
  • the placental stem cells of the present invention when cultured in primary cultures or in cell culture, adhere to the tissue culture substrate, e.g., tissue culture container surface (e.g., tissue culture plastic). Placental stem cells in culture assume a generally fibroblastoid, stellate appearance, with a number of cyotplasmic processes extending from the central cell body.
  • the placental stem cells are, however, morphologically differentiable from fibroblasts cultured under the same conditions, as the placental stem cells exhibit a greater number of such processes than do fibroblasts. Morphologically, placental stem cells are also differentiable from hematopoietic stem cells, which generally assume a more rounded, or cobblestone, morphology in culture.
  • Placental stem cells of the present invention express a plurality of markers that can be used to identify and/or isolate the stem cells, or populations of cells that comprise the stem cells.
  • the placental stem cells, and stem cell populations of the invention include stem cells and stem cell-containing cell populations obtained directly from the placenta, or any part thereof (e.g., amnion, chorion, placental cotyledons, and the like).
  • Placental stem cell populations also includes populations of (that is, two or more) placental stem cells in culture, and a population in a container, e.g., a bag.
  • Placental stem cells are not, however, trophoblasts.
  • the placental stem cells of the invention generally express the markers CD73, CDl 05, CD200, HLA-G, and/or OCT-4, and do not express CD34, CD38, or CD45.
  • Placental stem cells can also express HLA-ABC (MHC-I) and HLA-DR. These markers can be used to identify placental stem cells, and to distinguish placental stem cells from other stem cell types. Because the placental stem cells can express CD73 and CD 105, they can have mesenchymal stem cell-like characteristics.
  • the placental stem cells can express CD200 and HLA-G, a fetal-specific marker, they can be distinguished from mesenchymal stem cells, e.g., bone marrow-derived mesenchymal stem cells, which express neither CD200 nor HLA-G.
  • the lack of expression of CD34, CD38 and/or CD45 identifies the placental stem cells as non-hematopoietic stem cells.
  • the invention provides an isolated stem cell that is CD200 + or HLA-G + .
  • said stem cell is a placental stem cell.
  • the stem cell is CD200 + and HLA-G + .
  • said stem cell is CD73 + and CD105 + .
  • said stem cell is CD34 ⁇ , CD38 ⁇ or CD45 ⁇ .
  • said stem cell is CD34 " , CD38 ⁇ and CD45 " .
  • said stem cell is CD34 " , CD38 ⁇ , CD45 " , CD73 + and CDlOS + .
  • said CD200 + or HLA-G + stem cell facilitates the formation of ernbryoid-like bodies in a population of placental cells comprising the stem cells, under conditions that allow the formation of embryoid-like bodies.
  • said placental stem cell is isolated away from placental cells that are not stem cells.
  • said placental stem cell is isolated away from placental stem cells that do not display these markers.
  • the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD200 + or HLA-G + placental cell, whereby said cell is a placental stem cell.
  • said selecting comprises selecting a placental cell that is both CD200 + and HLA-G + .
  • said selecting comprises selecting a placental cell that is also CD73 + and CD105 + .
  • said selecting comprises selecting a placental cell that is also CD34 " , CD38 ⁇ or CD45 " .
  • said selecting comprises selecting a placental cell that is also CD34 ⁇ , CD38 ⁇ and CD45 ⁇ . In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34 ⁇ , CD38 ⁇ , CD45 ⁇ , CD73 + and CD105 + . In another specific embodiment, said selecting comprises selecting a placental cell that also facilitates the formation of embryoid-like bodies in a population of placental cells comprising the stem cells, under conditions that allow the formation of embryoid-like bodies.
  • the invention provides an isolated population of cells comprising, e.g., that is enriched for, CD200 + , HLA-G + stem cells.
  • said population is a population of placental cells.
  • at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at least about 60% of said cells are CD200 + , HLA-G + stem cells.
  • at least about 70% of said cells are CD200 + , HLA-G + stem cells. More preferably, at least about 90%, 95%, or 99% of said cells are CD200 + , HLA-G + stem cells.
  • said stem cells are also CD73 + and CDl 05 + .
  • said stem cells are also CD34 ⁇ , CD38 ⁇ or CD45 ⁇ .
  • said stem cells are also CD34 " , CD38 ⁇ , CD45 " CD73 + and CDl 05 + .
  • said isolated population produces one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • said population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these markers. W
  • the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said cells are CD200 + , HLA-G + stem cells.
  • said selecting comprises selecting stem cells that are also CD73 + and CD105 + .
  • said selecting comprises selecting stem cells that are also CD34 " , CD38 ⁇ or CD45 " .
  • said selecting comprises selecting stem cells that are also CD34 " , CD38 “ , CD45 “ , CD73 + and CD105 + .
  • said selecting also comprises selecting a population of placental stem cells that forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • the invention provides an isolated stem cell that is CD73 + , CD105 + , and CD200 + .
  • said isolated stem cell is an isolated placental stem cell.
  • said stem cell is HLA-G + .
  • said stem cell is CD34 " , CD38 ⁇ or CD45 " .
  • said stem cell is CD34 " , CD38 " and CD45 ⁇ .
  • said stem cell is CD34 " , CD38 " , CD45 " , and HLA-G + .
  • the isolated CD73 + , CDl 05 + , and CD200 + stem cell facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies.
  • said placental stern cell is isolated away from placental cells that are not stem cells.
  • said placental stem cell is isolated away from placental stem cells that do not display these markers.
  • the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD73 + , CD105 + , and CD200 + placental cell, whereby said cell is a placental stem cell.
  • said selecting comprises selecting a placental cell that is also HLA-G + .
  • said selecting comprises selecting a placental cell that is also CD34 " , CD38 " or CD45 ⁇ .
  • said selecting comprises selecting a placental cell that is also CD34 " , CD38 ⁇ and CD45 " In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34 " , CD38 " , CD45 " , and HLA-G + . In another specific embodiment, said selecting additionally comprises selecting a CD73 + , CD105 + , and CD200 + stem cell that facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell, when the population is cultured under conditions that facilitate formation of embryoid-like bodies.
  • the invention provides an isolated population of cells comprising, e.g., that is enriched for, CD73 + , CDl 05 + , CD200 + stem cells.
  • said stem cells are placental stem cells.
  • at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at " least about 60% of said cells are CD73 + , CDlOS + , CD200* stem cells.
  • at least about 70% of said cells in said population of cells are CD73 + , CD105 + , CD200 + stem cells.
  • At least about 90%, 95% or 99% of said cells in said population of cells are CD73 + , CD105 "1" , CD200 + stem cells.
  • said stem cells are HLA-G + .
  • said stem cells are CD34 " , CD38 " or CD45 " .
  • said stem cells are CD34 " , CD38 " and CD45 ⁇ .
  • said stem cells are CD34 ⁇ , CD38 ⁇ , CD45 ⁇ , and HLA-G + .
  • said population of cells produces one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • said population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said cells are CD73 + , CD105 + , CD200 + stem cells.
  • said selecting comprises selecting stem cells that are also HLA-G + .
  • said selecting comprises selecting stem cells that are also CD34 " , CD38 ⁇ or CD45 " .
  • said selecting comprises selecting stem cells that are also CD34 ⁇ , CD38 " and CD45 " . In another specific embodiment, said selecting comprises selecting stem cells that are also CD34 " , CD38 " , CD45 " , and HLA-G + . In another specific embodiment, said selecting additionally comprises selecting a population of placental cells that produces one or more embryoid-like bodies when the population is cultured under conditions that allow the formation of embryoid-like bodies.
  • the invention also provides an isolated stem cell that is CD200 + and OCT-4 + . In a specific embodiment, the stem cell is CD73 + and CD105 + . In a specific embodiment, the stem cell is a placental stem cell. In another specific embodiment, said stem cell is HLA-G + .
  • said stem cell is CD34 " , CD38 ⁇ or CD45 " .
  • said stem cell is CD34 " , CD38 ⁇ and CD45 ⁇ .
  • said stem cell is CD34 ⁇ , CD38 ⁇ , CD45 ⁇ CD73 + , CDl 05 + and HLA-G + .
  • the stem cell facilitates the production of one or more embryoid-like bodies by a population of placental cells that comprises the stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies.
  • said placental stem cell is isolated away from placental cells that are not stem cells.
  • said placental stem cell is isolated away from placental stem cells that do not display these markers.
  • the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD200 + and OCT-4 + placental cell, whereby said cell is a placental stem cell.
  • said selecting comprises selecting a placental cell that is also HLA-G + .
  • said selecting comprises selecting a placental cell that is also CD34 ⁇ , CD38 " or CD45 " .
  • said selecting comprises selecting a placental cell that is also CD34 " , CD38 " and CD45 " .
  • said selecting comprises selecting a placental cell that is also CD34 " CD38 " , CD45 “ , CD73 + , CD105 + and HLA-G + .
  • said selecting comprises selecting a placental stem cell that also facilitates the production of one or more embryoid-like bodies by a population of placental cells that comprises the stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies.
  • the invention also provides an isolated population of cells comprising, e.g., that is enriched for, CD200 "1* , OCT-4 + stem cells.
  • at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at least about 60% of said cells are CD200 + , OCT-4 + stem cells.
  • at least about 70% of said cells are said CD200 + , OCT-4 + stem cells.
  • at least about 90%, 95%, or 99% of said cells are said CD200 + , OCT-4 + stem cells.
  • said stem cells are CD73 + and CD105 + .
  • said stem cells are HLA-G + .
  • said stem cells are CD34 ⁇ , CD38 ⁇ and CD45 " .
  • said stem cells are CD34 ⁇ CD38 " , CD45 " CD73 + , CD105 + and HLA-G + .
  • the population produces one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • said population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said cells are CD200 + , OCT-4 + stem cells.
  • said selecting comprises selecting stem cells that are also CD73 + and CDlOS + .
  • said selecting comprises selecting stem cells that are also HLA-G + .
  • said selecting comprises selecting stem cells that are also CD34 " , CD38 " and CD45 " .
  • said stem cells are also CD34 ⁇ , CD38 " , CD45 " , CD73 + , CDlOS + and HLA-G + .
  • the invention further provides an isolated stem cell that is CD73 + , CD105 + and HLA- G + .
  • the stem cell is a placental stem cell.
  • said stem cell is CD34 ⁇ , CD38 ⁇ or CD45 ⁇ .
  • said stem cell is CD34 " , CD38 ⁇ and CD45 ⁇ " .
  • said stem cell is OCT-4 + .
  • said stem cell is CD200 + .
  • said stem cell is CD34 " , CD38 ⁇ , CD45 " , OCT-4 + and CD200 + .
  • said stem cell facilitates the formation of embryoid-like bodies in a population of placental ceils comprising said stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies.
  • said placental stem cell is isolated away from placental cells that are not stem cells.
  • said placental stem cell is isolated away from placental stem cells that do not display these characteristics.
  • the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD73 + , CDl 05 + and HLA-G + placental cell, whereby said cell is a placental stem cell.
  • said selecting comprises selecting a placental cell that is also CD34 ⁇ , CD38 ⁇ or CD45 " .
  • said selecting comprises selecting a placental cell that is also CD34 " , CD38 " and CD45 " .
  • said selecting comprises selecting a placental cell that is also OCT-4 + .
  • said selecting comprises selecting a placental cell that is also CD200 + .
  • said selecting comprises selecting a placental cell that is also CD34 ⁇ , CD38 " , CD45 " , OCT-4 + and CD200 + .
  • said selecting comprises selecting a placental cell that also facilitates the formation of one or more embryoid-like bodies in a population of placental cells that comprises said stem cell, when said population is culture under conditions that allow the formation of embryoid-like bodies.
  • the invention also provides an isolated population of cells comprising, e.g., that is enriched for, CD73 + , CD105 + and HLA-G + stem cells.
  • said stem cells are placental stem cells.
  • at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at least about 60% of said cells are CD73 + , CD105 + and HLA-G + stem cells.
  • at least about 70% of said cells are CD73 + , CD105 + and HLA-G + .
  • at least about 90%, 95% or 99% of said cells are CD73 + , CDl 05 + and HLA-G + stem cells.
  • said stem cells are CD34 " , CD38 ⁇ or CD45 " .
  • said stem cells are CD34 " , CD38 ⁇ and CD45 ⁇ .
  • said stem cells are OCT-4 + .
  • said stem cells are CD200 + .
  • said stem cells are CD34 ⁇ , CD38 " , CD45 " , OCT-4 + and CD200 + .
  • said population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein a majority of said cells are CD73 + , CD105 + and HLA-G + .
  • said majority of cells are also CD34 ⁇ , CD38 ⁇ and/or CD45 " .
  • said majority of cells are also CD200 + .
  • said majority of cells are also CD34 " , CD38 ⁇ , CD45 " , OCT-4 + and CD200 + .
  • the invention provides an isolated stem cell that is CD73 + and CD105 + and which facilitates the formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies.
  • said stem cell is CD34 ⁇ , CD38 " or CD45 ⁇ .
  • said stem cell is CD34 ⁇ , CD38 ⁇ and CD45 ⁇ .
  • said stem cell is OCT4 + .
  • said stem cell is OCT4 + , CD34 " , CD38 " and CD45 " .
  • placental stem cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these characteristics. [00771 The invention further provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73 + , CDl 05 + stem cells, wherein said population forms one or more embryoid-like bodies under conditions that allow formation of embryoid-like bodies.
  • At least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said isolated placental cells are CD73 + , CD105 + stem cells.
  • said stem cells are CD34 ⁇ , CD38 ⁇ or CD45 " .
  • said stem cells are CD34 ⁇ , CD38 ⁇ and CD45 " .
  • said stem cells are OCT-4 + .
  • said stem cells are OCT-4 + , CD34 " , CD38 " and CD45 " .
  • said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • said population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention further provides an isolated stem cell that is OCT-4 + and which facilitates formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when cultured under conditions that allow formation of embryoid-like bodies.
  • said stem cell is CD73 + and CD105 + .
  • said stem ceil is CD34 ⁇ , CD38 " , or CD45 " .
  • said stem cell is CD200 + .
  • said stem cell is CD73 + , CD105 + , CD200 + , CD34 " CD38 " , and CD45 "
  • said placental stem cell is isolated away from placental cells that are not stem cells.
  • said placental stem cell is isolated away from placental stem cells that do not display these characteristics.
  • the invention also provides a population of isolated cells comprising, e.g., that is enriched for, OCT-4 + stem cells, wherein said population forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
  • at least 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said isolated placental cells are OCT4 + stem cells.
  • said stem cells are CD73 + and CDl 05 + .
  • said stem cells are CD34 " , CD38 ⁇ , or CD45 " .
  • said stem cells are CD200 + .
  • said stem cells are CD73 + , CD105 + , CD200 "1" , CD34 " , CD38 ⁇ and CD45 "
  • said population has been expanded, for example, passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
  • said population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention also provides an isolated placental stem cell that is CDlO + , CD34 " , CDl 05 + , and CD200 + .
  • the invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO + , CD34 " , CD105 + , CD200 + .
  • said stem cells are additionally CD90 + and CD45 ⁇ .
  • said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • said isolated placental stem cell is non-maternal in origin.
  • at least about 90%, at least about 95%, or at least about 99% of said cells in said isolated population of placental stem cells are non-maternal in origin.
  • the invention provides an isolated placental stem cell that is HLA-A 5 B 5 C 5 CD45 " , CDl 33 " and CD34 " .
  • the invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are HLA- A,B,C ⁇ , CD45 " , CD 133 " and CD34 ⁇ .
  • said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • said isolated placental stem cell is non-maternal in origin.
  • the invention provides a method of obtaining a placental stem cell that is HLA-A 5 B J C-, CD45 , CD 133 " and CD34 " comprising isolating said cell from placental perfusate.
  • the invention provides an isolated placental stem cell that is CDlO + , CD13 + , CD33 + , CD45 " , CDl 17 " and CD133 "
  • the invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO + , CD13 + , CD33 + , CD45 " CDl IT and CD133 " .
  • said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said isolated placental stem cell is non-maternal in origin.
  • At least about 90%, at least about 95%, or at least about 99% of said cells in said isolated population of placental stem cells are non-maternal in origin.
  • said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention provides a method of obtaining a placental stem cell that is CDlO + , CD13 + , CD33 + , CD45 " , CDI lT and CD133 " comprising isolating said cell from placental perfusate.
  • the invention provides an isolated placental stem cell that is CDlO " , CD33 ⁇ , CD44 + , CD45 " , and CDl IT.
  • the invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO " , CD33 " , CD44 + , CD45 ⁇ , and CDl 17 " .
  • said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said isolated placental stem cell is non-maternal in origin.
  • the invention provides a method of obtaining a placental stem cell that is CDlO " , CD33 “ , CD44 + , CD45 " , CDl 17 " comprising isolating said cell from placental perfusate. [0084] In another embodiment, the invention provides an isolated placental stem cell that is CDlO " , CD 13 " , CD33 “ , CD45 “ ⁇ , and CDl 17 " .
  • the invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO " , CD13 " , CD33 " , CD45 " , and CDl 17 " .
  • said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said isolated placental stem cell is non-maternal in origin.
  • at least about 90%, at least about 95%, or at least 99% of said cells in said isolated population of placental stem cells are non-maternal in origin.
  • said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention provides a method of obtaining a placental stem cell that is CDlO " , CD 13 " , CD33 " , CD45 " , and CDl 17 + comprising isolating said cell from placental perfusate. [0085]
  • the invention provides an isolated placental stem cell that is HLA A,B,C ⁇ , CD45 " , CD34 “ , CD133 " , positive for CDlO, CD13, CD38, CD44, CD90, CD 105, CD200 and/or HLA-G, and/or negative for CDl 17.
  • the invention further provides an isolated population of placental stem cells, wherein said stem cells are HLA A,B,C, CD45 " CD34 " , CDl 33 " , and at least about 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or about 99% of the stem cells in the population are positive for CDlO, CD 13, CD38, CD44, CD90, CD 105, CD200 and/or HLA- G, and/or negative for CD 117.
  • said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells.
  • said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least about 99%, of said cells in said isolated population of placental stem cells, are non-maternal in origin. In another specific embodiment, said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
  • the invention provides a method of obtaining a placental stem cell that is HLA A 5 B, C ⁇ , CD45 " , CD34 " CD133 " and positive for CDlO, CD13, CD38, CD44, CD90, CD105, CD200 and/or HLA-G, and/or negative for CDl 17, comprising isolating said cell from placental perfusate.
  • the invention provides a placental stem cell that is CD200 + and CDlO + , as determined by antibody binding, and CDl 17 " , as determined by both antibody binding and RT-PCR.
  • the invention provides a placental stem cell that is CDlO + , CD29 " CD54 + , CD200 + , HLA-G + , HLA class T and ⁇ -2-microglobulin " .
  • the invention provides placental stem cells, wherein the expression of at least one marker is at least two-fold higher than for a mesenchymal stem cell (e.g., a bone marrow-derived mesenchymal stem cell).
  • said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about W
  • the invention provides an isolated population of placental stem cells, wherein a plurality of said placental stem cells are positive for aldehyde dehydrogenase (ALDH), as assessed by an aldehyde dehydrogenase activity assay.
  • ALDH aldehyde dehydrogenase
  • said ALDH assay uses ALDEFLUOR® (Aldagen, Inc., Ashland, Oregon) as a marker of aldehyde dehydrogenase activity.
  • said plurality is between about 3% and about 25% of cells in said population of cells.
  • the invention provides a population of umbilical cord stem cells, wherein a plurality of said umbilical cord stem cells are positive for aldehyde dehydrogenase, as assessed by an aldehyde dehydrogenase activity assay that uses ALDEFLUOR® as an indicator of aldehyde dehydrogenase activity.
  • said plurality is between about 3% and about 25% of cells in said population of cells.
  • said population of placental stem cells or umbilical cord stem cells shows at least three-fold, or at least five-fold, higher ALDH activity than a population of bone marrow-derived mesenchymal stem cells having the same number of cells and cultured under the same conditions.
  • the invention provides any of the above placental stem cells, or populations of placental stem cells, wherein the stem cell or population of placental stem cells has been passaged at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 times, or more, or expanded for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 population doublings, or more.
  • the karyotype of the cells, or at least about 95% or about 99% of the cells in said population is normal.
  • the cells, or cells in the population of cells are non-maternal in origin.
  • Isolated placental stem cells, or isolated populations of placental stem cells, bearing any of the above combinations of markers, can be combined in any ratio.
  • the invention also provides for the isolation of, or enrichment for, any two or more of the above placental stem ceil populations to form a placental stem cell population.
  • the invention provides an isolated population of placental stem cells comprising a first population of placental stem cells defined by one of the marker combinations described above and a second population of placental stem cells defined by another of the marker combinations described above, wherein said first and second populations are combined in a ratio of about 1 :99, 2:98, 3:97, 4:96, 5:95, 10:90, 20:80, 30:70, 40:60, 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98:2, or about 99:1.
  • any three, four, five or more of the above-described placental stem cells or placental stem cell populations can be combined.
  • the invention further provides placental stem cells that are obtained by disruption of placental tissue, with or without enzymatic digestion, followed by culture ⁇ see Section 5.2.3) or perfusion ⁇ see Section 5.2.4).
  • the invention provides an isolated population of placental stem cells that is produced according to a method comprising perfusing a mammalian placenta that has been drained of cord blood and perfused to remove residual blood; perfusing said placenta with a perfusion solution; and collecting said perfusion solution, wherein said perfusion solution after perfusion comprises a population of placental cells that comprises placental stem cells; and isolating a plurality of said placental stem cells from said population of cells.
  • the perfusion solution is passed through both the umbilical vein and umbilical arteries and collected after it exudes from the placenta.
  • Populations of placental stem cells produced by this method typically comprise a mixture of fetal and maternal cells.
  • the perfusion solution is passed through the umbilical vein and collected from the umbilical arteries, or passed through the umbilical arteries and collected from the umbilical vein.
  • Populations of placental stem cells produced by this method typically are substantially exclusively fetal in origin; that is, e.g., greater than 90%, 95%, 99%, or 99.5% of the placental stem cells in the population are fetal in origin.
  • the placental stem cells contained within a population of cells obtained from perfusion of a placenta, are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% of said population of placental cells.
  • the placental stem cells collected by perfusion comprise fetal and maternal cells.
  • the placental stem cells collected by perfusion are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% fetal cells.
  • the invention provides a composition comprising a population of isolated placental stem cells collected by perfusion, wherein said composition comprises at least a portion of the perfusion solution used to collect the placental stem cells.
  • the invention further provides an isolated population of the placental stem cells described herein that is produced according to a method comprising digesting placental tissue with a tissue-disrupting enzyme to obtain a population of placental cells comprising placental stem cells, and isolating a plurality of placental stem cells from the remainder of said placental cells. The whole, or any part of, the placenta can be digested to obtain placental stem cells.
  • said placental tissue is a whole placenta, an amniotic membrane, chorion, a combination of amnion and chorion, or a combination of any of the foregoing.
  • the tissue-disrupting enzyme is trypsin or collagenase.
  • the placental stem cells, contained within a population of cells obtained from digesting a placenta are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% of said population of placental cells.
  • Gene profiling confirms that isolated placental stem cells, and populations of isolated placental stem cells, are distinguishable from other cells, e.g., mesenchymal stem cells, e.g., bone marrow-derived stem cells.
  • mesenchymal stem cells e.g., bone marrow-derived stem cells.
  • the placental stem cells described herein can be distinguished from mesenchymal stem cells on the basis of the expression of one or more genes, the expression of which is specific to placental stem cells or umbilical cord stem cells in comparison to bone marrow-derived mesenchymal stem cells.
  • placental stem cells can be distinguished from mesenchymal stem cells on the basis of the expression of one or more gene, the expression of which is significantly higher (that is, at least twofold higher) in placental stem cells than in mesenchymal stem cells, wherein the one or more gene is(are) ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl lorf9, CD200, COL4A1, COL4A2, CPA4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9 ⁇ ST3GAL6, ST6GALNAC5, SLC12A
  • the placental stem cell-specific or umbilical cord stem cell-specific gene is CD200.
  • the level of expression of these genes can be used to confirm the identity of a population of placental cells, to identify a population of cells as comprising at least a plurality of placental stem cells, or the like.
  • the population of placental stem cells can be clonal, e.g., a population of placental stem cells expanded form a single placental stem cell, or a mixed population of stem cells, e.g., a population of cells comprising solely placental stem cells that are expanded from multiple placental stem cells, or a population of cells comprising placental stem cells and at least one other type of cell.
  • the level of expression of these genes can be used to select populations of placental stem cells.
  • a population of cells e.g., clonally-expanded cells
  • a population of cells is selected if the expression of one or more of these genes is significantly higher in a sample from the population of cells than in an equivalent population of mesenchymal stem cells.
  • Such selecting can be of a population from a plurality of placental stem cells populations, from a plurality of cell populations, the identity of which is not known, etc.
  • Placental stem cells can be selected on the basis of the level of expression of one or more such genes as compared to the level of expression in said one or more genes in a mesenchymal stem cell control.
  • the level of expression of said one or more genes in a sample comprising an equivalent number of mesenchymal stem cells is used as a control.
  • the control, for placental stem cells tested under certain conditions is a numeric value representing the level of expression of said one or more genes in mesenchymal stem cells under said conditions.
  • the placental stem cells of the invention display the above characteristics (e.g., combinations of cell surface markers and/or gene expression profiles) in primary culture, or during proliferation in medium comprising 60% DMEM-LG (Gibco), 40% MCDB- 201 (Sigma), 2% fetal calf serum (FCS) (Hyclone Laboratories), Ix insulin-transferrin- selenium (ITS), Ix lenolenic-acid-bovine-serum-albumin (LA-BSA), 10 "9 M dexamethasone (Sigma), 10 "4 M ascorbic acid 2-phosphate (Sigma), epidermal growth factor (EGF) 10ng/ml (R&D Systems), platelet derived-growth factor (PDGF-BB) lOng/ml (R&D Systems), and IOOU penicillin/100OU streptomycin.
  • FCS fetal calf serum
  • ITS insulin-transferrin- selenium
  • LA-BSA Ix len
  • the isolated populations of placental stem cells described above, and populations of placental stem cells generally, can comprise about, at least, or no more than, 1 x 10 5 , 5 x 10 s , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , 5 x 10 8 , 1 x 10 9 , 5 x 10 9 , 1 x 10 10 , 5 x 10 10 , 1 x 10 11 or more placental stem cells.
  • placental stem cells described herein depends in part upon the particular medium selected for growth. Under optimum conditions, placental stem cells typically double in number in 3-5 days.
  • the placental stem cells of the invention adhere to a substrate in culture, e.g. the surface of a tissue culture container ⁇ e.g., tissue culture dish plastic, fibronectin-coated plastic, and the like) and form a monolayer.
  • embryoid-like bodies that is, three-dimensional clusters of cells grow atop the adherent stem cell layer.
  • Cells within the embryoid-like bodies express markers associated with very early stem cells, e.g., OCT-4, Nanog, SSEA3 and SSEA4.
  • Cells within the embryoid-like bodies are typically not adherent to the culture substrate, as are the placental stem cells described herein, but remain attached to the adherent cells during culture.
  • Embryoid-like body cells are dependent upon the adherent placental stem cells for viability, as embryoid-like bodies do not form in the absence of the adherent stem cells.
  • the adherent placental stem cells thus facilitate the growth of one or more embryoid-like bodies in a population of placental cells that comprise the adherent placental stem cells.
  • the cells of the embryoid-like bodies are thought to grow on the adherent placental stem cells much as embryonic stem cells grow on a feeder layer of cells.
  • Mesenchymal stem cells e.g., bone marrow-derived mesenchymal stem cells, do not develop embryoid-like bodies in culture.
  • the present invention further provides methods of collecting and isolating placental stem cells.
  • stem cells are obtained from a mammalian placenta using a physiologically-acceptable solution, e.g., a stem cell collection composition.
  • a stem cell collection composition is described in detail in related U.S. Provisional Application No. 60/754,969, entitled "Improved Medium for Collecting Placental Stem Cells and Preserving Organs," filed on December 29, 2005.
  • the stem cell collection composition can comprise any physiologically-acceptable solution suitable for the collection and/or culture of stem cells, for example, a saline solution (e.g., phosphate-buffered saline, Kreb's solution, modified Kreb's solution, Eagle's solution, 0.9% NaCl. etc.), a culture medium (e.g., DMEM, H.DMEM, etc.), and the like.
  • a saline solution e.g., phosphate-buffered saline, Kreb's solution, modified Kreb's solution, Eagle's solution, 0.9% NaCl. etc.
  • a culture medium e.g., DMEM, H.DMEM, etc.
  • the stem cell collection composition can comprise one or more components that tend to preserve placental stem cells, that is, prevent the placental stem cells from dying, or delay the death of the placental stem cells, reduce the number of placental stem cells in a population of cells that die, or the like, from the time of collection to the time of culturing.
  • Such components can be, e.g., an apoptosis inhibitor (e.g., a caspase inhibitor or JNK inhibitor); a vasodilator (e.g., magnesium sulfate, an antihypertensive drug, atrial natriuretic peptide (ANP), adrenocorticotropin, corticotropin-releasing hormone, sodium nitroprusside, hydralazine, adenosine triphosphate, adenosine, indomethacin or magnesium sulfate, a phosphodiesterase inhibitor, etc.); a necrosis inhibitor (e.g., 2-(lH-Indol-3-yl)-3-pentylamino- maleimide, pyrrolidine dithiocarbamate, or clonazepam); a TNF- ⁇ inhibitor; and/or an oxygen-carrying perfluorocarbon (e.g., perfluorooctyl bromid
  • the stem cell collection composition can comprise one or more tissue-degrading enzymes, e.g., a metalloprotease, a serine protease, a neutral protease, an RNase, or a DNase, or the like.
  • tissue-degrading enzymes include, but are not limited to, collagenases (e.g., collagenase I, II, III or IV, a collagenase from Clostridium histolyticum, etc.); dispase, thermolysin, elastase, trypsin, LIBERASE, hyaluronidase, and the like.
  • the stem cell collection composition can comprise a bacteriocidally or bacteriostatically effective amount of an antibiotic.
  • the antibiotic is a macrolide (e.g., tobramycin), a cephalosporin (e.g., cephalexin, cephradine, cefuroxime, cefprozil, cefaclor, cefixime or cefadroxil), a clarithromycin, an erythromycin, a penicillin (e.g., penicillin V) or a quinolone (e.g., ofloxacin, ciprofloxacin or norfloxacin), a tetracycline, a streptomycin, etc.
  • the antibiotic is active against Gram(+) and/or Gram(— ) bacteria, e.g., Pseudomonas aeruginosa, Staphylococcus aureus, and the like.
  • the stem cell collection composition can also comprise one or more of the following compounds: adenosine (about 1 mM to about 50 mM); D-glucose (about 20 mM to about 100 mM); magnesium ions (about 1 mM to about 50 mM); a macromolecule of molecular weight greater than 20,000 daltons, in one embodiment, present in an amount sufficient to maintain endothelial integrity and cellular viability (e.g., a synthetic or naturally occurring colloid, a polysaccharide such as dextran or a polyethylene glycol present at about 25 g/1 to about 100 g/1, or about 40 g/1 to about 60 g/1); an antioxidant (e.g., butylated hydroxyanisole, butylated hydroxytoluene, glutathione, vitamin C or vitamin E present at about 25 ⁇ M to about 100 ⁇ M); a reducing agent (e.g., N-acetylcysteine present at about 0.1 m
  • a human placenta is recovered shortly after its expulsion after birth.
  • the placenta is recovered from a patient after informed consent and after a complete medical history of the patient is taken and is associated with the placenta.
  • the medical history continues after delivery.
  • Such a medical history can be used to coordinate subsequent use of the placenta or the stem cells harvested therefrom.
  • human placental stem cells can be used, in light of the medical history, for personalized medicine for the infant associated with the placenta, or for parents, siblings or other relatives of the infant.
  • the umbilical cord blood and placental blood are removed prior to recovery of placental stem cells.
  • the cord blood in the placenta is recovered.
  • the placenta can be subjected to a conventional cord blood recovery process.
  • a needle or cannula is used, with the aid of gravity, to exsanguinate the placenta (see, e.g., Anderson, U.S. Patent No. 5,372,581; Hessel etal, U.S. Patent No. 5,415,665).
  • the needle or cannula is usually placed in the umbilical vein and the placenta can be gently massaged to aid in draining cord blood from the placenta.
  • cord blood recovery may be performed commercially, e.g., LifeBank USA, Cedar Knolls, N. J., ViaCord, Cord Blood Registry and Cryocell.
  • the placenta is gravity drained without further manipulation so as to minimize tissue disruption during cord blood recovery.
  • a placenta is transported from the delivery or birthing room to another location, e.g., a laboratory, for recovery of cord blood and collection of stem cells by, e.g., perfusion or tissue dissociation.
  • the placenta is preferably transported in a sterile, thermally insulated transport device (maintaining the temperature of the placenta between 20-28 0 C), for example, by placing the placenta, with clamped proximal umbilical cord, in a sterile zip-lock plastic bag, which is then placed in an insulated container.
  • the placenta is transported in a cord blood collection kit substantially as described in pending United States Patent No. 7,147,626.
  • the placenta is delivered to the laboratory four to twenty-four hours following delivery.
  • the proximal umbilical cord is clamped, preferably within 4-5 cm (centimeter) of the insertion into the placental disc prior to cord blood recovery. In other embodiments, the proximal umbilical cord is clamped after cord blood recovery but prior to further processing of the placenta.
  • the placenta, prior to stem cell collection can be stored under sterile conditions and at either room temperature or at a temperature of 5 to 25°C (centigrade). The placenta may be stored for a period of for a period of four to twenty- four hours, up to forty-eight hours, or longer than forty eight hours, prior to perfusing the placenta to remove any residual cord blood.
  • the placenta is harvested from between about zero hours to about two hours post-expulsion.
  • the placenta is preferably stored in an anticoagulant solution at a temperature of 5 to 25°C (centigrade).
  • Suitable anticoagulant solutions are well known in the art.
  • a solution of heparin or warfarin sodium can be used.
  • the anticoagulant solution comprises a solution of heparin (e.g., 1% w/w in 1 : 1000 solution).
  • the exsanguinated placenta is preferably stored for no more than 36 hours before placental stem cells are collected.
  • the mammalian placenta or a part thereof, once collected and prepared generally as above, can be treated in any art-known manner, e.g., can be perfused or disrupted, e.g., digested with one or more tissue-disrupting enzymes, to obtain stem cells.
  • stem cells are collected from a mammalian placenta by physical disruption of part of all of the organ.
  • the placenta, or a portion thereof may be, e.g., crushed, sheared, minced, diced, chopped, macerated or the like.
  • the tissue can then be cultured to obtain a population of stem cells.
  • the placental tissue is disrupted using, e.g., in, a stem cell collection composition (see Section 5.2.1 and below).
  • the placenta can be dissected into components prior to physical disruption and/or enzymatic digestion and stem cell recovery.
  • Placental stem cells can be obtained from all or a portion of the amniotic membrane, chorion, umbilical cord, placental cotyledons, or any combination thereof, including from a whole placenta.
  • placental stem cells are obtained from placental tissue comprising amnion and chorion.
  • placental stem cells can be obtained by disruption of a small block of placental tissue, e.g., a. block of placental tissue that is about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900 or about 1000 cubic millimeters in volume.
  • any method of physical disruption can be used, provided that the method of disruption leaves a plurality, more preferably a majority, and more preferably at least 60%, 70%, 80%, 90%, 95%, 98%, or 99% of the cells in said organ viable, as determined by, e.g., trypan blue exclusion.
  • Stem cells can generally be collected from a placenta, or portion thereof, at any time within about the first three days post-expulsion, but preferably between about 8 hours and about 18 hours post-expulsion.
  • the disrupted tissue is cultured in tissue culture medium suitable for the proliferation of placental stem cells ⁇ see, e.g., Section 5.3, below, describing the culture of placental stem cells).
  • stem cells are collected by physical disruption of placental tissue, wherein the physical disruption includes enzymatic digestion, which can be accomplished by use of one or more tissue-digesting enzymes.
  • the placenta, or a portion thereof, may also be physically disrupted and digested with one or more enzymes, and the resulting material then immersed in, or mixed into, a stem cell collection composition.
  • a preferred stem cell collection composition comprises one or more tissue-disruptive enzyme(s).
  • Enzymatic digestion preferably uses a combination of enzymes, e.g., a combination of a matrix metalloprotease and a neutral protease, for example, a combination of collagenase and dispase.
  • enzymatic digestion of placental tissue uses a combination of a matrix metalloprotease, a neutral protease, and a mucolytic enzyme for digestion of hyaluronic acid, such as a combination of collagenase, dispase, and hyaluronidase or a combination of LIBERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) and hyaluronidase.
  • a matrix metalloprotease such as a combination of collagenase, dispase, and hyaluronidase or a combination of LIBERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) and hyaluronidase.
  • a mucolytic enzyme for digestion of hyaluronic acid, such as a combination of collagenase, dispase, and hyaluronidase or a combination of LIBERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) and hyaluronidase.
  • Serine proteases may be inhibited by alpha 2 microglobulin in serum and therefore the medium used for digestion is usually serum-free. EDTA and DNase are commonly used in enzyme digestion procedures to increase the efficiency of cell recovery.
  • the digestate is preferably diluted so as to avoid trapping stem cells within the viscous digest.
  • tissue digestion enzymes can be used. Typical concentrations for tissue digestion enzymes include, e.g., 50-200 U/mL for collagenase I and collagenase IV, 1- 10 U/mL for dispase, and 10-100 U/mL for elastase.
  • Proteases can be used in combination, that is, two or more proteases in the same digestion reaction, or can be used sequentially in order to liberate placental stem cells.
  • a placenta, or part thereof is digested first with an appropriate amount of collagenase I at about 1 to about 2 mg/ml for, e.g., 30 minutes, followed by digestion with trypsin, at a concentration of about 0.25%, for, e.g., 10 minutes, at 37°C.
  • Serine proteases are preferably used consecutively following use of other enzymes.
  • the tissue can further be disrupted by the addition of a chelator, e.g., ethylene glycol bis(2-aminoethyl ether)-N,N,N'N'-tetraacetic acid (EGTA) or ethylenediaminetetraacetic acid (EDTA) to the stem cell collection composition comprising the stem cells, or to a solution in which the tissue is disrupted and/or digested prior to isolation of the stem cells with the stem cell collection composition.
  • a digestion can proceed as follows. Approximately a gram of placental tissue is obtained and minced.
  • the tissue is digested in 10 mL of a solution comprising about 1 mg/mL collagenase IA and about 0.25% trypsin at 37°C in a shaker at about 100 RPM.
  • the digestate is washed three times with culture medium, and the washed cells are seeded into 2 T-75 flasks.
  • The. cells are then isolated by differential adherence, and characterized for, e.g., viability, cell surface markers, differentiation, and the like.
  • the placental stem cells collected will comprise a mix of placental stem cells derived from both fetal and maternal sources. Where a portion of the placenta that comprises no, or a negligible number of, maternal cells (for example, amnion), the placental stem cells collected will comprise almost exclusively fetal placental stem cells.
  • Stem cells can be isolated from disrupted tissue by differential trypsinization (see Section 5.2.5, below) followed by culture in one or more new culture containers in fresh proliferation medium, optionally followed by a second differential trypsinization step.
  • Placental stem cells can also be obtained by perfusion of the mammalian placenta. Methods of perfusing mammalian placenta to obtain stem cells are disclosed, e.g., in Hariri, U.S. Application Publication No. 2002/0123141, and in related U.S. Provisional Application No. 60/754,969, entitled "Improved Medium for Collecting Placental Stem Cells and Preserving Organs,” filed on December 29, 2005.
  • Placental stem cells can be collected by perfusion, e.g., through the placental vasculature, using, e.g., a stem cell collection composition as a perfusion solution.
  • a mammalian placenta is perfused by passage of perfusion solution through either or both of the umbilical artery and umbilical vein.
  • the flow of perfusion solution through the placenta may be accomplished using, e.g., gravity flow into the placenta.
  • the perfusion solution is forced through the placenta using a pump, e.g., a peristaltic pump.
  • the umbilical vein can be, e.g., cannulated with a cannula, e.g., a TEFLON ® or plastic cannula, that is connected to a sterile connection apparatus, such as sterile tubing.
  • the sterile connection apparatus is connected to a perfusion manifold.
  • the placenta is preferably oriented (e.g., suspended) in such a manner that the umbilical artery and umbilical vein are located at the highest point of the placenta.
  • the placenta can be perfused by passage of a perfusion fluid through the placental vasculature and surrounding tissue.
  • the placenta can also be perfused by passage of a perfusion fluid into the umbilical vein and collection from the umbilical arteries, or passage of a perfusion fluid into the umbilical arteries and collection from the umbilical vein.
  • the umbilical artery and the umbilical vein are connected simultaneously, e.g., to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution.
  • the perfusion solution is passed into the umbilical vein and artery.
  • the perfusion solution exudes from and/or passes through the walls of the blood vessels into the surrounding tissues of the placenta, and is collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation.
  • the perfusion solution may also be introduced through the umbilical cord opening and allowed to flow or percolate out of openings in the wall of the placenta which interfaced with the maternal uterine wall.
  • Placental cells that are collected by this method which can be referred to as a "pan” method, are typically a mixture of fetal and maternal cells.
  • the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or is passed through the umbilical artery and collected from the umbilical veins.
  • Placental cells collected by this method which can be referred to as a "closed circuit" method, are typically almost exclusively fetal.
  • perfusion using the pan method that is, whereby perfusate is collected after it has exuded from the maternal side of the placenta, results in a mix of fetal and maternal cells.
  • the cells collected by this method comprise a mixed population of placental stem cells of both fetal and maternal origin.
  • the closed circuit perfusion method can, in one embodiment, be performed as follows.
  • a post-partum placenta is obtained within about 48 hours after birth.
  • the umbilical cord is clamped and cut above the clamp.
  • the umbilical cord can be discarded, or can processed to recover, e.g., umbilical cord stem cells, and/or to process the umbilical cord membrane for the production of a biomaterial.
  • the amniotic membrane can be retained during perfusion, or can be separated from the chorion, e.g., using blunt dissection with the fingers.
  • amniotic membrane is separated from the chorion prior to perfusion, it can be, e.g., discarded, or processed, e.g., to obtain stem cells by enzymatic digestion, or to produce, e.g., an amniotic membrane biomaterial, e.g., the biomaterial described in U.S. Application Publication No. 2004/0048796.
  • an amniotic membrane biomaterial e.g., the biomaterial described in U.S. Application Publication No. 2004/0048796.
  • the umbilical cord vessels are exposed, e.g., by partially cutting the umbilical cord membrane to expose a cross-section of the cord.
  • the vessels are identified, and opened, e.g., by advancing a closed alligator clamp through the cut end of each vessel.
  • the apparatus e.g., plastic tubing connected to a perfusion device or peristaltic pump, is then inserted into each of the placental arteries.
  • the pump can be any pump suitable for the purpose, e.g., a peristaltic pump.
  • Plastic tubing, connected to a sterile collection reservoir, e.g., a blood bag such as a 250 mL collection bag, is then inserted into the placental vein.
  • the tubing connected to the pump is inserted into the placental vein, and tubes to a collection reservoir(s) are inserted into one or both of the placental arteries.
  • the placenta is then perfused with a volume of perfusion solution, e.g., about 750 ml of perfusion solution. Cells in the perfusate are then collected, e.g., by centrifugation.
  • the proximal umbilical cord is clamped during perfusion, and more preferably, is clamped within 4-5 cm (centimeter) of the cord's insertion into the placental disc.
  • the first collection of perfusion fluid from a mammalian placenta during the exsanguination process is generally colored with residual red blood cells of the cord blood and/or placental blood.
  • the perfusion fluid becomes more colorless as perfusion proceeds and the residual cord blood cells are washed out of the placenta.
  • 30 to 100 ml (milliliter) of perfusion fluid is adequate to initially exsanguinate the placenta, but more or less perfusion fluid may be used depending on the observed results.
  • the volume of perfusion liquid used to collect placental stem cells may vary depending upon the number of stem cells to be collected, the size of the placenta, the number of collections to be made from a single placenta, etc.
  • the volume of perfusion liquid may be from 50 mL to 5000 mL, 50 mL to 4000 mL, 50 mL to 3000 mL, 100 mL to 2000 mL, 250 mL to 2000 mL, 500 mL to 2000 mL, or 750 mL to 2000 mL.
  • the placenta is perfused with 700-800 mL of perfusion liquid following exsanguination.
  • the placenta can be perfused a plurality of times over the course of several hours or several days. Where the placenta is to be perfused a plurality of times, it may be maintained or cultured under aseptic conditions in a container or other suitable vessel, and perfused with the stem cell collection composition, or a standard perfusion solution (e.g., a normal saline solution such as phosphate buffered saline ("PBS”)) with or without an anticoagulant (e.g., heparin, warfarin sodium, coumarin, bishydroxycoumarin), and/or with or without an antimicrobial agent (e.g., ⁇ -mercaptoethanol (0.1 mM); antibiotics such as streptomycin (e.g., at 40-100 ⁇ g/ml), penicillin (e.g., at 40U/ml), amphotericin B (e.g., at 0.5 ⁇ g/ml).
  • a standard perfusion solution e.g., a
  • an isolated placenta is maintained or cultured for a period of time without collecting the perfusate, such that the placenta is maintained or cultured for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, or 2 or 3 or more days before perfusion and collection of perfusate.
  • the perfused placenta can be maintained for one or more additional time(s), e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12. 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and perfused a second time with, e.g., 700-800 mL perfusion fluid.
  • the placenta can be perfused 1, 2, 3, 4, 5 or more times, for example, once every 1, 2, 3, 4, 5 or 6 hours.
  • perfusion of the placenta and collection of perfusion solution e.g., stem cell collection composition
  • perfusion of the placenta and collection of perfusion solution is repeated until the number of recovered nucleated cells falls below 100 cells/ml.
  • the perfusates at different time points can be further processed individually to recover time-dependent populations of cells, e.g., stem cells. Perfusates from different time points can also be pooled.
  • stem cells are collected at a time or times between about 8 hours and about 18 hours post-expulsion.
  • placental stem cells are believed to migrate into the exsanguinated and perfused microcirculation of the placenta where, according to the methods of the invention, they are collected, preferably by washing into a collecting vessel by perfusion.
  • Perfusing the isolated placenta not only serves to remove residual cord blood but also provide the placenta with the appropriate nutrients, including oxygen.
  • the placenta may be cultivated and perfused with a similar solution which was used to remove the residual cord blood cells, preferably, without the addition of anticoagulant agents.
  • Perfusion according to the methods of the invention results in the collection of significantly more placental stem cells than the number obtainable from a mammalian placenta not perfused with said solution, and not otherwise treated to obtain stem cells (e.g., by tissue disruption, e.g., enzymatic digestion).
  • stem cells e.g., by tissue disruption, e.g., enzymatic digestion.
  • "significantly more” means at least 10% more.
  • Perfusion according to the methods of the invention yields significantly more placental stem cells than, e.g., the number of placental stem cells obtainable from culture medium in which a placenta, or portion thereof, has been cultured.
  • Stem cells can be isolated from placenta by perfusion with a solution comprising one or more proteases or other tissue-disruptive enzymes.
  • a placenta or portion thereof e. g. , amniotic membrane, amnion and chorion, placental lobule or cotyledon, umbilical cord, or combination of any of the foregoing
  • a placenta or portion thereof e. g. , amniotic membrane, amnion and chorion, placental lobule or cotyledon, umbilical cord, or combination of any of the foregoing
  • tissue-disruptive enzymes in 200 mL of a culture medium for 30 minutes.
  • Cells from the perfusate are collected, brought to 4°C, and washed with a cold inhibitor mix comprising 5 mM EDTA, 2 mM dithiothreitol and 2 mM beta-mercaptoethanol.
  • the stem cells are washed after several minutes with a cold ⁇ e.g., 4 0 C) stem cell collection composition.
  • J Stem cells from mammalian placenta can initially be purified from ⁇ i.e., be isolated from) other cells by Ficoll gradient centrifugation. Such centrifugation can follow any standard protocol for centrifugation speed, etc. In one embodiment, for example, cells collected from the placenta are recovered from perfusate by centrifugation at 5000 x g for 15 minutes at room temperature, which separates cells from, e.g., contaminating debris and platelets.
  • placental perfusate is concentrated to about 200 ml, gently layered over Ficoll, and centrifuged at about 1100 x g for 20 minutes at 22°C, and the low-density interface layer of cells is collected for further processing.
  • Cell pellets can be resuspended in fresh stem cell collection composition, or a medium suitable for stem cell maintenance, e.g., IMDM serum-free medium containing 2U/ml heparin and 2mM EDTA (GibcoBRL, NY).
  • IMDM serum-free medium containing 2U/ml heparin and 2mM EDTA
  • the total mononuclear cell fraction can be isolated, e.g., using Lymphoprep (Nycomed Pharma, Oslo, Norway) according to the manufacturer's recommended procedure.
  • placing placental stem cells means to remove at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99% of the cells with which the stem cells are normally associated in the intact mammalian placenta.
  • a stem cell from an organ is “isolated” when it is present in a population of cells that comprises fewer than 50% of the cells with which the stem cell is normally associated in the intact organ.
  • Placental cells obtained by perfusion or digestion can, for example, be further, or initially, isolated by differential trypsinization using, e.g., a solution of 0.05% trypsin with 0.2% EDTA (Sigma, St. Louis MO).
  • placental stem cells typically detach from plastic surfaces within about five minutes whereas other adherent populations typically require more than 20-30 minutes incubation.
  • the detached placental stem cells can be harvested following trypsinization and trypsin neutralization, using, e.g., Trypsin Neutralizing Solution (TNS, Cambrex).
  • Trypsin Neutralizing Solution e.g., Trypsin Neutralizing Solution (TNS, Cambrex).
  • TSS Trypsin Neutralizing Solution
  • aliquots of, for example, about 5-10 x 10 cells are placed in each of several T-75 flasks, preferably fibronectin-coated T75 flasks.
  • the cells can be cultured with commercially available Mesenchymal Stem Cell Growth Medium (MSCGM) (Cambrex), and placed in a tissue culture incubator (37°C, 5% CO 2 ). After 10 to 15 days, non-adherent cells are removed from the flasks by washing with PBS. The PBS is then replaced by MSCGM. Flasks are preferably examined daily for the presence of various adherent cell types and in particular, for identification and expansion of clusters of fibroblastoid cells.
  • MSCGM Mesenchymal Stem Cell Growth Medium
  • the number and type of cells collected from a mammalian placenta can be monitored, for example, by measuring changes in morphology and cell surface markers using standard cell detection techniques such as flow cytometry, cell sorting, immunocytochemistry (e.g., staining with tissue specific or cell-marker specific antibodies) fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), by examination of the morphology of cells using light or confocal microscopy, and/or by measuring changes in gene expression using techniques well known in the art, such as PCR and gene expression profiling. These techniques can be used, too, to identify cells that are positive for one or more particular markers.
  • standard cell detection techniques such as flow cytometry, cell sorting, immunocytochemistry (e.g., staining with tissue specific or cell-marker specific antibodies) fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), by examination of the morphology of cells using light or confocal microscopy, and/or by measuring changes in
  • a cell comprises a detectable amount of CD34; if so, the cell is CD34 + .
  • the cell is OCT-4 +
  • Antibodies to cell surface markers e.g., CD markers such as CD34
  • sequence of stem cell-specific genes such as OCT-4
  • Placental cells may be sorted using a fluorescence activated cell sorter (FACS).
  • Fluorescence activated cell sorting is a well-known method for separating particles, including cells, based on the fluorescent properties of the particles (Kamarch, 1987, Methods Enzymol, 151 :150- 165). Laser excitation of fluorescent moieties in the individual particles results in a small electrical charge allowing electromagnetic separation of positive and negative particles from a mixture.
  • cell surface marker-specific antibodies or ligands are labeled with distinct fluorescent labels. Cells are processed through the cell sorter, allowing separation of cells based on their ability to bind to the antibodies used.
  • FACS sorted particles may be directly deposited into individual wells of 96-well or 384-well plates to facilitate separation and cloning.
  • stem cells from placenta are sorted on the basis of expression of the markers CD34, CD38, CD44, CD45, CD73, CD105, OCT-4 and/or HLA-G. This can be accomplished in connection with procedures to select stem cells on the basis of their adherence properties in culture. For example, an adherence selection stem can be accomplished before or after sorting on the basis of marker expression.
  • cells are sorted first on the basis of their expression of CD34; CD34 " cells are retained, and cells that are CD200 + HLA-G + , are separated from all other CD34 ⁇ cells.
  • cells from placenta are based on their expression of markers CD200 and/or HLA-G; for example, cells displaying either of these markers are isolated for further use.
  • Cells that express, e.g., CD200 and/or HLA-G can, in a specific embodiment, be further sorted based on their expression of CD73 and/or CD 105, or epitopes recognized by antibodies SH2, SH3 or SH4, or lack of expression of CD34, CD38 or CD45.
  • placental cells are sorted by expression, or lack thereof, of CD200, HLA- G, CD73, CDl 05, CD34, CD38 and CD45, and placental cells that are CD200 + , HLA-G + , CD73 + , CD105 + , CD34 ⁇ , CD38 " and CD45 ' are isolated from other placental cells for further use.
  • any antibody specific for a particular marker, can be used, in combination with any fluorophore or other label suitable for the detection and sorting of cells (e.g., fluorescence-activated cell sorting).
  • Antibody/fluorophore combinations to specific markers include, but are not limited to, fluorescein isothiocyanate (FITC) conjugated monoclonal antibodies against HLA-G (available from Serotec, Raleigh, North Carolina), CDlO (available from BD Immunocytometry Systems, San Jose, California), CD44 (available from BD Biosciences Pharmingen, San Jose, California), and CD 105 (available from R&D Systems Inc., Minneapolis, Minnesota); phycoerythrin (PE) conjugated monoclonal antibodies against CD44, CD200, CDl 17, and CD 13 (BD Biosciences Pharmingen); phycoerythrin-Cy7 (PE Cy7) conjugated monoclonal antibodies against CD33 and CDlO (BD Biosciences Pharmingen); allophycocyanin (APC) conjugated streptavidin and monoclonal antibodies against CD38 (BD Biosciences Pharmingen); and Biotinylated CD90 (BD Biosciences Pharmingen).
  • FITC flu
  • antibodies that can be used include, but are not limited to, CDl 33-APC (Miltenyi), KDR-Biotin (CD309, Abeam), CytokeratinK-Fitc (Sigma or Dako), HLA ABC- Fitc (BD), HLA DRDQDP-PE (BD), ⁇ -2-microglobulin-PE (BD), CD80-PE (BD) and CD86- APC (BD).
  • CDl 33-APC Miltenyi
  • KDR-Biotin CD309, Abeam
  • CytokeratinK-Fitc Sigma or Dako
  • HLA ABC- Fitc BD
  • HLA DRDQDP-PE HLA DRDQDP-PE
  • BD ⁇ -2-microglobulin-PE
  • CD80-PE CD80-PE
  • CD86- APC CD86- APC
  • antibody/label combinations that can be used include, but are not limited to, CD45-PerCP (peridin chlorophyll protein); CD44-PE; CD 19-PE; CDlO-F (fluorescein); HLA-G-F and 7-am ⁇ no-actinomycin-D (7-AAD); HLA-ABC-F; and the like.
  • Placental stem cells can be assayed for CDl 17 or CD 133 using, for example, phycoerythrin-Cy5 (PE Cy5) conjugated streptavidin and biotin conjugated monoclonal antibodies against CDl 17 or CD 133; however, using this system, the cells can appear to be positive for CDl 17 or CD133, respectively, because of a relatively high background.
  • Placental stem cells can be labeled with an antibody to a single marker and detected and/sorted. Placental stem cells can also be simultaneously labeled with multiple antibodies to different markers.
  • magnetic beads can be used to separate cells.
  • the cells may be sorted using a magnetic activated cell sorting (MACS) technique, a method for separating particles based on their ability to bind magnetic beads (0.5-100 ⁇ m diameter).
  • MCS magnetic activated cell sorting
  • a variety of useful modifications can be performed on the magnetic microspheres, including covalent addition of antibody that specifically recognizes a particular cell surface molecule or hapten.
  • the beads are then mixed with the cells to allow binding. Cells are then passed through a magnetic field to separate out cells having the specific cell surface marker. In one embodiment, these cells can then isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers. The cells are again passed through a magnetic field, isolating cells that bound both the antibodies. Such cells can then be diluted into separate dishes, such as microtiter dishes for clonal isolation.
  • Placental stem cells can also be characterized and/or sorted based on cell morphology and growth characteristics.
  • placental stem cells can be characterized as having, and/or selected on the basis of, e.g., a fibroblastoid appearance in culture.
  • Placental stem cells can also be characterized as having, and/or be selected, on the basis of their ability to form embryoid-like bodies.
  • placental cells that are fibroblastoid in shape express CD73 and CD 105, and produce one or more embryoid-like bodies in culture are isolated from other placental cells.
  • OCT-4 + placental cells that produce one or more embryoid-like bodies in culture are isolated from other placental cells.
  • placental stem cells can be identified and characterized by a colony forming unit assay.
  • Colony forming unit assays are commonly known in the art, such as MESEN CULTTM medium (Stem Cell Technologies, Inc., Vancouver British Columbia)
  • Placental stem cells can be assessed for viability, proliferation potential, and longevity using standard techniques known in the art, such as trypan blue exclusion assay, fluorescein diacetate uptake assay, propidium iodide uptake assay (to assess viability); and thymidine uptake assay, MTT cell proliferation assay (to assess proliferation). Longevity may be determined by methods well known in the art, such as by determining the maximum number of population doubling in an extended culture.
  • Placental stem cells can also be separated from other placental cells using other techniques known in the art, e.g., selective growth of desired ceils (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation); unit gravity separation; countercurrent. distribution; electrophoresis; and the like.
  • other techniques known in the art e.g., selective growth of desired ceils (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation); unit gravity separation; countercurrent. distribution; electrophoresis; and
  • Isolated placental stem cells or placental stem cell population, or cells or placental tissue from which placental stem cells grow out, can be used to initiate, or seed, cell cultures.
  • Cells are generally transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen (e.g., native or denatured), gelatin, f ⁇ bronectin, ornithine, vitronectin, and extracellular membrane protein (e.g., MATRIGEL® (BD Discovery Labware, Bedford, Mass.)).
  • extracellular matrix or ligands such as laminin, collagen (e.g., native or denatured), gelatin, f ⁇ bronectin, ornithine, vitronectin, and extracellular membrane protein (e.g., MATRIGEL® (BD Discovery Labware, Bedford, Mass.)).
  • Placental stem cells can be cultured in any medium, and under any conditions, recognized in the art as acceptable for the culture of stem cells.
  • the culture medium comprises serum.
  • Placental stem cells can be cultured in, for example, DMEM-LG (Dulbecco's Modified Essential Medium, low glucose)/MCDB 201 (chick fibroblast basal medium) containing ITS (insulin-transferrin-selenium), LA+BSA (linoleic acid-bovine serum albumin), dextrose, L-ascorbic acid, PDGF, EGF, IGF-I, and penicillin/streptomycin; DMEM-HG (high glucose) comprising 10% fetal bovine serum (FBS); DMEM-HG comprising 15% FBS; IMDM (Iscove's modified Dulbecco's medium) comprising 10% FBS, 10% horse serum, and hydrocortisone; Ml 99 comprising 10% FBS, EGF, and heparin;
  • DMEM high or low glucose
  • Eagle's basal medium Ham's FlO medium (FlO)
  • Ham's F-12 medium F 12
  • Iscove's modified Dulbecco's medium Mesenchymal Stem Cell Growth Medium (MSCGM) 5 Liebovitz's L- 15 medium
  • MCDB DMEM/F12
  • RPMI 1640 advanced DMEM (Gibco), DMEM/MCDB201 (Sigma), and CELL-GRO FREE.
  • the culture medium can be supplemented with one or more components including, for example, serum ⁇ e.g., fetal bovine serum (FBS), preferably about 2-15% (v/v); equine (horse) serum (ES); human serum (HS)); beta-mercaptoethanol (BME), preferably about 0.001% (v/v); one or more growth factors, for example, platelet-derived growth factor (PDGF), epidermal growth factor (EGF) ?
  • FBS fetal bovine serum
  • ES equine (horse) serum
  • HS human serum
  • BME beta-mercaptoethanol
  • growth factors for example, platelet-derived growth factor (PDGF), epidermal growth factor (EGF) ?
  • bFGF basic fibroblast growth factor
  • IGF-I insulin-like growth factor- 1
  • LIF leukemia inhibitory factor
  • VEGF vascular endothelial growth factor
  • EPO erythropoietin
  • amino acids including L-valine
  • antibiotic and/or antimycotic agents to control microbial contamination such as, for example, penicillin G, streptomycin sulfate, amphotericin B, gentamicin, and nystatin, either alone or in combination.
  • Placental stem cells can be cultured in standard tissue culture conditions, e.g., in tissue culture dishes or multiwell plates. Placental stem cells can also be cultured using a hanging drop method. In this method, placental stem cells are suspended at about 1 x 10 4 cells per mL in about 5 mL of medium, and one or more drops of the medium are placed on the inside of the lid of a tissue culture container, e.g., a 100 mL Petri dish. The drops can be, e.g., single drops, or multiple drops from, e.g., a multichannel pipetter. The lid is carefully inverted and placed on top of the bottom of the dish, which contains a volume of liquid, e.g., sterile PBS sufficient to maintain the moisture content in the dish atmosphere, and the stem cells are cultured.
  • a volume of liquid e.g., sterile PBS
  • the placental stem cells are cultured in the presence of a compound that acts to maintain an undifferentiated phenotype in the placental stem cell.
  • the compound is a substituted 3,4-dihydropyridimol[4,5-d]pyrimid ⁇ ne.
  • the compound is a compound having the following chemical structure:
  • the compound can be contacted with a placental stem cell, or population of placental stem cells, at a concentration of, for example, between about 1 ⁇ M to about 10 ⁇ M.
  • an isolated placental stem cell, or isolated population of stem cells e.g., a stem cell or population of stem cells separated from at least 50% of the placental cells with which the stem cell or population of stem cells is normally associated in vivo
  • the stem cell or population of stem cells can be proliferated and expanded in vitro.
  • a population of placental stem cells can be cultured in tissue culture containers, e.g., dishes, flasks, multiwell plates, or the like, for a sufficient time for the stem cells to proliferate to 70-90% confluence, that is, until the stem cells and their progeny occupy 70-90% of the culturing surface area of the tissue culture container.
  • Placental stem cells can be seeded in culture vessels at a density that allows cell growth.
  • the cells may be seeded at low density (e.g., about 1,000 to about 5,000 cells/cm 2 ) to high density (e.g., about 50,000 or more cells/cm 2 ).
  • the cells are cultured at about 0 to about 5 percent by volume CO 2 in air.
  • the cells are cultured at about 2 to about 25 percent O 2 in air, preferably about 5 to about 20 percent O 2 in air.
  • the cells preferably are cultured at about 25°C to about 40 0 C, preferably 37°C.
  • the cells are preferably cultured in an incubator.
  • the culture medium can be static or agitated, for example, using a bioreactor.
  • Placental stem cells preferably are grown under low oxidative stress (e.g., with addition of glutathione, ascorbic acid, catalase, tocopherol, N-acetylcysteine, or the like).
  • the cells may be passaged.
  • the cells can be enzymatically treated, e.g., trypsinized, using techniques well-known in the art, to separate them from the tissue culture surface.
  • about 20,000-100,000 stem cells preferably about 50,000 stem cells, are passaged to a new culture container containing fresh culture medium.
  • the new medium is the same type of medium from which the stem cells were removed.
  • the invention encompasses populations of placental stem cells that have been passaged at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 times, or more.
  • Placental stem cell population can be isolated directly from one or more placentas; that is, the placental stem cell population can be a population of placental cells comprising placental stem cells obtained from, or contained within, perfusate, or obtained from, or contained within, disrupted placental tissue, e.g., placental tissue digestate (that is, the collection of cells obtained by enzymatic digestion of a placenta or part thereof).
  • Isolated placental stem cells of the invention can also be cultured and expanded to produce placental stem cell populations.
  • Populations of placental cells comprising placental stem cells can also be cultured and expanded to produce placental stem cell populations.
  • Placental stem cell populations of the invention comprise placental stem cells, for example, placental stem cells as described herein. In various embodiments, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the cells in an isolated placental stem cell population are placental stem cells. That is, a placental stem cell population can comprise, e.g., as much as 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% non-stem cells.
  • the invention provides methods of producing isolated placental stem cell population by, e.g., selecting placental stem cells, whether derived from enzymatic digestion or perfusion, that express particular markers and/or particular culture or morphological characteristics.
  • the invention provides a method of producing a cell population comprising selecting placental cells that (a) adhere to a substrate, and (b) express CD200 and HLA-G; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population comprising identifying placental cells that express CD200 and HLA-G, and isolating said cells from other cells to form a cell population.
  • the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, and (b) express CD73, CDl 05, and CD200; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population comprising identifying placental cells that express CD73, CD105, and CD200, and isolating said cells from other cells to form a cell population.
  • the method of producing a ceil population comprises selecting placental cells that (a) adhere to a substrate and (b) express CD200 and OCT-4; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population comprising identifying placental cells that express CD200 and OCT-4, and isolating said cells from other cells to form a cell population.
  • the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, (b) express CD73 and CDl 05, and (c) facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population comprising identifying placental cells that express CD73 and CD 105, and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body, and isolating said cells from other cells to form a cell population.
  • the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, and (b) express CD73, CD 105 and HLA-G; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population comprising identifying placental cells that express CD73, CD 105 and HLA-G, and isolating said cells from other cells to form a cell population.
  • the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, (b) express OCT-4, and (c) facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body; and isolating said cells from other cells to form a cell population.
  • the invention provides a method of producing a cell population comprising identifying placental cells that express OCT-4, and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body, and isolating said cells from other cells to form a cell population.
  • Such cell populations can be used to treat any of the diseases or conditions listed hereinbelow.
  • Such cell populations can also be used to assess populations of placental stem cells, e.g., as part of a quality control method.
  • the method can additionally comprise selecting placental cells that express ABC-p (a placenta-specific ABC transporter protein; see, e.g., Allikmets et ah, Cancer Res. 58(23):5337-9 (1998)).
  • the method can also comprise selecting cells exhibiting at least one characteristic specific to, e.g., a mesenchymal stem cell, for example, expression of CD29, expression of CD44, expression of CD90, or expression of a combination of the foregoing.
  • the substrate can be any surface on which culture and/or selection of Ce 1 IIs, e.g., placental stem cells, can be accomplished.
  • the substrate is plastic, e.g., tissue culture dish or multiwell plate plastic.
  • Tissue culture plastic can be coated with a biomolecule, e.g., laminin or fibronectin.
  • Cells e.g., placental stem cells
  • cells can be selected using an antibody or antibodies to one or more cell surface markers, for example, in flow cytometry or FACS. Selection can be accomplished using antibodies in conjunction with magnetic beads.
  • Antibodies that are specific for certain stem cell-related markers are known in the art. For example, antibodies to OCT-4 (Abeam, Cambridge, MA), CD200 (Abeam), HLA-G (Abeam), CD73 (BD Biosciences Pharmingen, San Diego, CA), CDl 05 (Abeam; BioDesign International, Saco, ME), etc.
  • Antibodies to other markers are also available commercially, e.g., CD34, CD38 and CD45 are available from, e.g., StemCell Technologies or BioDesign International.
  • the isolated placental stem cell population can comprise placental cells that are not stem cells, or cells that are not placental cells.
  • Isolated placental stem cell populations can be combined with one or more populations of non-stem cells or non-placental cells.
  • an isolated population of placental stem cells can be combined with blood (e.g., placental blood or umbilical cord blood), blood-derived stem cells (e.g., stem cells derived from placental blood or umbilical cord blood), umbilical cord stem cells, populations of blood-derived nucleated cells, bone marrow-derived mesenchymal cells, bone-derived stem cell populations, crude bone marrow, adult (somatic) stem cells, populations of stem cells contained within tissue, cultured stem cells, populations of fully-differentiated cells (e.g., chondrocytes, fibroblasts, amniotic cells, osteoblasts, muscle cells, cardiac cells, etc.) and the like.
  • blood e.g., placental blood or umbilical cord blood
  • blood-derived stem cells e.g., stem cells derived from placental blood or umbilical cord blood
  • the invention provides a population of stem cells comprising placental stem cells and umbilical cord stem cells.
  • Cells in an isolated placental stem cell population can be combined with a plurality of cells of another type in ratios of about 100,000,000:1, 50,000,000:1, 20,000,000:1, 10,000,000:1, 5,000,000:1, 2,000,000:1, 1,000,000:1, 500,000:1, 200,000:1, 100,000:1, 50,000: 1, 20,000:1, 10,000:1 , 5,000:1, 2,000:1, 1,000:1, 500: 1, 200: 1, 100:1, 50:1, 20:1, 10: 1, 5:1, 2:1, 1:1 ; 1 :2; 1:5; 1 :10; 1 : 100; 1:200; 1 :500; 1 :1,000; 1 :2,000; 1 :5,000; 1 :10,000; 1 :20,000; 1 :50,000; 1:100,000; 1 :500,000; 1: 1,000,000; 1 :2,000,000; 1 :5,000,000; 1 :10,000,000; 1 :20,000,000; 1 :50,000,000; or about 1,000,000:1, 50
  • Cells in an isolated placental stem cell population can be combined with a plurality of cells of a plurality of cell types, as well.
  • an isolated population of placental stem cells is combined with a plurality of hematopoietic stem cells.
  • Such hematopoietic stem cells can be, for example, contained within unprocessed placental, umbilical cord blood or peripheral blood; in total nucleated cells from placental blood, umbilical cord blood or peripheral blood; in an isolated population of CD34 + cells from placental blood, umbilical cord blood or peripheral blood; in unprocessed bone marrow; in total nucleated cells from bone marrow; in an isolated population of CD34 + cells from bone marrow, or the like.
  • Stem cells from postpartum placentas can be cultured in a number of different ways to produce a set of lots, e.g., a set of individually-administrable doses, of placental stem cells.
  • lots can, for example, be obtained from stem cells from placental perfusate or from enzyme-digested placental tissue.
  • Sets of lots of placental stem cells, obtained from a plurality of placentas, can be arranged in a bank of placental stem cells for, e.g., long-term storage.
  • adherent stem cells are obtained from an initial culture of placental material to form a seed culture, which is expanded under controlled conditions to form populations of cells from approximately equivalent numbers of doublings. Lots are preferably derived from the tissue of a single placenta, but can be derived from the tissue of a plurality of placentas.
  • stem cell lots are obtained as follows. Placental tissue is first disrupted, e.g., by mincing, digested with a suitable enzyme, e.g., collagenase ⁇ see Section 5.2.3, above).
  • the placental tissue preferably comprises, e.g., the entire amnion, entire chorion, or both, from a single placenta, but can comprise only a part of either the amnion or chorion.
  • the digested tissue is cultured, e.g., for about 1-3 weeks, preferably about 2 weeks. After removal of non-adherent cells, high-density colonies that form are collected, e.g., by trypsinization. These cells are collected and resuspended in a convenient volume of culture medium, and defined as Passage 0 cells.
  • Passage 0 cells are then used to seed expansion cultures.
  • Expansion cultures can be any arrangement of separate cell culture apparatuses, e.g., a Cell Factory by NUNCTM.
  • Cells in the Passage 0 culture can be subdivided to any degree so as to seed expansion cultures with, e.g., 1 x 10 3 , 2 x 10 3 , 3 x 10 3 , 4 x 10 3 , 5 x 10 3 , 6 x lO 3 , 7 x 10 3 , 8 x 10 3 , 9 x 10 3 , 1 x 10 4 , 1 x IO 4 , 2 x 10 4 , 3 x 10 4 , 4 x 10 4 , 5 x 10 4 , 6 x 10 4 , 7 x 10 4 , 8 x 10 4 , 9 x 10 4 , or 10 x 10 4 stem cells.
  • each expansion culture Preferably, from about 2 x 10 4 to about 3 x 10 4 Passage O cells are used to seed each expansion culture.
  • the number of expansion cultures can depend upon the number of Passage 0 cells, and may be greater or fewer in number depending upon the particular placenta(s) from which the stem cells are obtained.
  • Expansion cultures are grown until the density of cells in culture reaches a certain value, e.g., about 1 x 10 5 cells/cm 2 .
  • Cells can either be collected and cryopreserved at this point, or passaged into new expansion cultures as described above.
  • Cells can be passaged, e.g., 2, 3, 4 , 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 times prior to use.
  • a record of the cumulative number of population doublings is preferably maintained during expansion culture(s).
  • the cells from a Passage 0 culture can be expanded for 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 doublings, or up to 60 doublings.
  • the number of population doublings, prior to dividing the population of cells into individual doses is between about 15 and about 30, preferably about 20 doublings.
  • the cells can be culture continuously throughout the expansion process, or can be frozen at one or more points during expansion.
  • Cells to be used for individual doses can be frozen, e.g., cryopreserved for later use.
  • Individual doses can comprise, e.g., about 1 million to about 100 million cells per ml, and can comprise between about 10 6 and about 10 9 cells in total.
  • Passage 0 cells are cultured for a first number of doublings, e.g., approximately 4 doublings, then frozen in a first cell bank.
  • Cells from the first cell bank are frozen and used to seed a second cell bank, the cells of which are expanded for a second number of doublings, e.g., about another eight doublings.
  • Cells at this stage are collected and frozen and used to seed new expansion cultures that are allowed to proceed for a third number of doublings, e.g., about eight additional doublings, bringing the cumulative number of cell doublings to about 20.
  • Cells at the intermediate points in passaging can be frozen in units of about 100,000 to about 10 million cells per ml, preferably about 1 million cells per ml for use in subsequent expansion culture. Cells at about 20 doublings can be frozen in individual doses of between about 1 million to about 100 million cells per ml for administration or use in making a stem cell-containing composition.
  • the invention provides a method of making a placental stem cell bank, comprising: expanding primary culture placental stem cells from a human post-partum placenta for a first plurality of population doublings; cryopreserving said placental stem cells to form a Master Cell Bank; expanding a plurality of placental stem cells from the Master Cell Bank for a second plurality of population doublings; cryopreserving said placental stem cells to form a Working Cell Bank; expanding a plurality of placental stem cells from the Working Cell Bank for a third plurality of population doublings; and cryopreserving said placental stem cells in individual doses, wherein said individual doses collectively compose a placental stem cell bank.
  • the total number of population doublings is about 20.
  • said first plurality of population doublings is about four population doublings; said second plurality of population doublings is about eight population doublings; and said third plurality of population doublings is about eight population doublings.
  • said primary culture placental stem cells comprise placental stem cells from placental perfusate.
  • said primary culture placental stem cells comprise placental stem cells from digested placental tissue.
  • said primary culture placental stem cells comprise placental stem cells from placental perfusate and from digested placental tissue.
  • all of said placental stem cells in said placental stem cell primary culture are from the same placenta.
  • the method further comprises the step of selecting CD200 + or HLA-G + placental stem cells from said plurality of said placental stem cells from said Working Cell Bank to form individual doses.
  • said individual doses comprise from about 10 4 to about 10 5 placental stem cells.
  • said individual doses comprise from about 10 s to about 10 6 placental stem cells.
  • said individual doses comprise from about 10 6 to about 10 7 placental stem cells.
  • said individual doses comprise from about 10 7 to about 10 s placental stem cells.
  • the donor from which the placenta is obtained ⁇ e.g., the mother) is tested for at least one pathogen. If the mother tests positive for a tested pathogen, the entire lot from the placenta is discarded. Such testing can be performed at any time during production of placental stem cell lots, including before or after establishment of Passage 0 cells, or during expansion culture.
  • Pathogens for which the presence is tested can include, without limitation, hepatitis A, hepatitis B, hepatitis C, hepatitis D, hepatitis E, human immunodeficiency virus (types I and II), cytomegalovirus, herpesvirus, and the like.
  • Neuronal differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into neurons.
  • a neurogenic medium comprises DMEM/20% FBS and 1 mM beta- mercaptoethanol; such medium can be replaced after culture for about 24 hours with medium consisting of DMEM and 1-10 mM betamercaptoethanol.
  • the cells are contacted with DMEM/2% DMSO/200 ⁇ M butylated hydroxyanisole.
  • the differentiation medium comprises serum-free DMEMIF- 12, butylated hydroxyanisole, potassium chloride, insulin, forskolin, valproic acid, and hydrocortisone.
  • neuronal differentiation is accomplished by plating placental stem cells on laminin-coated plates in Neurobasal-A medium (Invitrogen, Carlsbad CA) containing B27 supplement and L-glutamine, optionally supplemented with bFGF and/or EGF. Placental stem cells can also be induced to neural differentiation by co-culture with neural cells, or culture in neuron-conditioned medium.
  • Neuronal differentiation can be assessed, e.g., by detection of neuron-like morphology (e.g., bipolar cells comprising extended processes) detection of the expression of e.g., nerve growth factor receptor and neurofilament heavy chain genes by RT-PCR; or detection of electrical activity, e.g., by patch-clamp.
  • a placental stem cell is considered to have differentiated into a neuronal cell when the cell displays one or more of these characteristics.
  • Adipogenic differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into adipocytes.
  • a preferred adipogenic medium comprises MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum.
  • placental stem cells are fed Adipogenesis Induction Medium (Cambrex) and cultured for 3 days (at 37°C, 5% CO 2 ), followed by 1-3 days of culture in Adipogenesis Maintenance Medium (Cambrex). After 3 complete cycles of induction/maintenance, the cells are cultured for an additional 7 days in adipogenesis maintenance medium, replacing the medium every 2-3 days.
  • placental stem cells are cultured in medium comprising 1 ⁇ M dexamethasone, 0.2 mM indomethacin, 0.01 mg/ml insulin, 0.5 mM IBMX, DMEM-high glucose, FBS, and antibiotics.
  • Placental stem cells can also be induced towards adipogenesis by culture in medium comprising one or more glucocorticoids ⁇ e.g., dexamethasone, indomethasone, hydrocortisone, cortisone), insulin, a compound which elevates intracellular levels of cAMP (e.g., dibutyryl-cAMP; 8-CPT-cAMP (8-(4)chlorophenylthio)-adenosine, 3',5' cyclic monophosphate); 8-bromo-cAMP; dioctanoyl-cAMP; forskolin) and/or a compound which inhibits degradation of cAMP (e.g., a phosphodiesterase inhibitor such as isobutylmethylxanthine (IBMX), methyl isobutylxanthine, theophylline, caffeine, indomethacin).
  • glucocorticoids ⁇ e.g., dexamethasone, indome
  • a hallmark of adipogenesis is the development of multiple intracytoplasmic lipid vesicles that can be easily observed using the lipophilic stain oil red O. Expression of lipase and/or fatty acid binding protein genes is confirmed by RT/PCR in placental stem cells that have begun to differentiate into adipocytes. A placental stem cell is considered to have differentiated into an adipocytic cell when the cell displays one or more of these characteristics.
  • Chondrogenic differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into chondrocytes.
  • a preferred chondrocytic medium comprises MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum.
  • placental stem cells are aliquoted into a sterile polypropylene tube, centrifuged (e.g., at 150 x g for 5 minutes), and washed twice in Incomplete Chondrogenesis Medium (Cambrex).
  • the cells are resuspended in Complete Chondrogenesis Medium (Cambrex) containing 0.01 ⁇ g/ml TGF- beta-3 at a concentration of about 1-20 x 10 s cells/ml.
  • placental stem cells are contacted with exogenous growth factors, e.g., GDF-5 or transforming growth factor beta3 (TGF-beta3), with or without ascorbate.
  • Chondrogenic medium can be supplemented with amino acids including proline and glutamine, sodium pyruvate, dexamethasone, ascorbic acid, and insulin/transferrin/selenium. Chondrogenic medium can be supplemented with sodium hydroxide and/or collagen.
  • the placental stem cells may be cultured at high or low density. Cells are preferably cultured in the absence of serum.
  • Chondrogenesis can be assessed by e.g., observation of production of esoinophilic ground substance, safranin-O staining for glycosaminoglycan expression; hematoxylin/eosin staining, assessing cell morphology, and/or RT/PCR confirmation of collagen 2 and collagen 9 gene expression. Chondrogenesis can also be observed by growing the stem cells in a pellet, formed, e.g., by gently centrifuging stem cells in suspension (e.g., at about 80Og for about 5 minutes).
  • the pellet of stem cells begins to form a tough matrix and demonstrates a structural integrity not found in non-induced, or non- chondrogenic, cell lines, pellets of which tend to fall apart when challenged. Chondrogenesis can also be demonstrated, e.g., in such cell pellets, by staining with a stain that stains collage, e.g., Sirius Red, and/or a stain that stains glycosaminoglycans (GAGs), such as, e.g., Alcian Blue. A placental stem cell is considered to have differentiated into a chondrocytic cell when the cell displays one or more of these characteristics.
  • a stain that stains collage e.g., Sirius Red
  • GAGs glycosaminoglycans
  • a placental stem cell is considered to have differentiated into a chondrocytic cell when the cell displays one or more of these characteristics.
  • Osteogenic differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into osteocytes.
  • a preferred osteocytic medium comprises MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum, followed by Osteogenic Induction Medium (Cambrex) containing 0.1 ⁇ M dexamethasone, 0.05 mM ascorbic acid-2-phosphate, 10 mM beta glycerophosphate.
  • placental stem cells are cultured in medium (e.g., DMEM-low glucose) containing about 10 "7 to about 10 "9 M dexamethasone, about 10- 50 ⁇ M ascorbate phosphate salt (e.g., ascorbate-2-phosphate) and about 10 nM to about 10 mM ⁇ -glycerophosphate.
  • Osteogenic medium can also include serum, one or more antibiotic/antimycotic agents, transforming growth factor-beta (e.g., TGF- ⁇ l) and/or bone morphogenic protein (e.g., BMP-2, BMP-4, or a combination thereof).
  • Differentiation can be assayed using a calcium-specific stain, e.g., von Kossa staining, and RT/PCR detection of, e.g., alkaline phosphatase, osteocalcin, bone sialoprotein and/or osteopontin gene expression.
  • a placental stem cell is considered to have differentiated into an osteocytic cell when the cell displays one or more of these characteristics.
  • Differentiation of placental stem cells into insulin-producing pancreatic cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into pancreatic cells.
  • An example pancreagenic medium comprises DMEM/20% CBS, supplemented with basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml. This medium is combined with conditioned media from nestin-positive neuronal cell cultures at 50/50 v/v. KnockOut Serum Replacement can be used in lieu of CBS. Cells are cultured for 14-28 days, refeeding every 3-4 days. [0193] Differentiation can be confirmed by assaying for, e.g., insulin protein production, or insulin gene expression by RTVPCR. A placental stem cell is considered to have differentiated into a pancreatic cell when the cell displays one or more of these characteristics.
  • Myogenic (cardiogenic) differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into cardiomyocytes.
  • a preferred cardiomyocytic medium comprises DMEM/20% CBS supplemented with retinoic acid, 1 ⁇ M; basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml; and epidermal growth factor, 100 ng/ml.
  • KnockOut Serum Replacement (Invitrogen, Carlsbad, California) may be used in lieu of CBS.
  • placental stem cells are cultured in DMEM/20% CBS supplemented with 50 ng/ml Cardiotropin-1 for 24 hours.
  • placental stem cells can be cultured 10-14 days in protein-free medium for 5-7 days, then stimulated with human myocardium extract, e.g., produced by homogenizing human myocardium in 1% HEPES buffer supplemented with 1% cord blood serum.
  • human myocardium extract e.g., produced by homogenizing human myocardium in 1% HEPES buffer supplemented with 1% cord blood serum.
  • a placental stem cell is considered to have differentiated into a cardiac cell when the cell displays one or more of these characteristics.
  • Placental stem cells can be preserved, that is, placed under conditions that allow for long-term storage, or conditions that inhibit cell death by, e.g., apoptosis or necrosis.
  • Placental stem cells can be preserved using, e.g., a composition comprising an apoptosis inhibitor, necrosis inhibitor and/or an oxygen-carrying perfluorocarbon, as described in related U.S. Provisional Application No. 60/754,969, entitled "Improved Medium for Collecting Placental Stem Cells and Preserving Organs," filed on December 25, 2005.
  • the invention provides a method of preserving a population of stem cells comprising contacting said population of stem cells with a stem cell collection composition comprising an inhibitor of apoptosis and an oxygen-carrying perfluorocarbon, wherein said inhibitor of apoptosis is present in an amount and for a time sufficient to reduce or prevent apoptosis in the population of stem cells, as compared to a population of stem cells not contacted with the inhibitor of apoptosis.
  • said inhibitor of apoptosis is a caspase inhibitor.
  • said inhibitor of apoptosis is a JNK inhibitor.
  • said JNK inhibitor does not modulate differentiation or proliferation of said stem cells.
  • said stem cell collection composition comprises said inhibitor of apoptosis and said oxygen-carrying perfluorocarbon in separate phases.
  • said stem cell collection composition comprises said inhibitor of apoptosis and said oxygen-carrying perfluorocarbon in an emulsion.
  • the stem cell collection composition additionally comprises an emulsifier, e.g., lecithin.
  • said apoptosis inhibitor and said perfluorocarbon are between about O 0 C and about 25°C at the time of contacting the stem cells.
  • said apoptosis inhibitor and said perfluorocarbon are between about 2°C and 10 0 C, or between about 2°C and about 5°C, at the time of contacting the stem cells.
  • said contacting is performed during transport of said population of stem cells.
  • said contacting is performed during freezing and thawing of said population of stem cells.
  • the invention provides a method of preserving a population of placental stem cells comprising contacting said population of stem cells with an inhibitor of apoptosis and an organ-preserving compound, wherein said inhibitor of apoptosis is present in an amount and for a time sufficient to reduce or prevent apoptosis in the population of stem cells, as compared to a population of stem cells not contacted with the inhibitor of apoptosis.
  • the organ-preserving compound is UW solution (described in U.S. Patent No.
  • the stem cell collection composition additionally comprises an oxygen-carrying perfluorocarbon, either in two phases or as an emulsion.
  • placental stem cells are contacted with a stem cell collection composition comprising an apoptosis inhibitor and oxygen-carrying perfluorocarbon, organ-preserving compound, or combination thereof, during perfusion.
  • said stem cells are contacted during a process of tissue disruption, e.g., enzymatic digestion.
  • placental stem cells are contacted with said stem cell collection compound after collection by perfusion, or after collection by tissue disruption, e.g., enzymatic digestion.
  • tissue disruption e.g., enzymatic digestion
  • a stem cell, or population of stem cells is exposed to a hypoxic condition during collection, enrichment or isolation for less than six hours during said preservation, wherein a hypoxic condition is a concentration of oxygen that is less than normal blood oxygen concentration.
  • a hypoxic condition is a concentration of oxygen that is less than normal blood oxygen concentration.
  • said population of stem cells is exposed to said hypoxic condition for less than two hours during said preservation.
  • said population of stem cells is exposed to said hypoxic condition for less than one hour, or less than thirty minutes, or is not exposed to a hypoxic condition, during collection, enrichment or isolation.
  • said population of stem cells is not exposed to shear stress during collection, enrichment or isolation.
  • cryopreservation medium includes, but is not limited to, culture medium including, e.g., growth medium, or cell freezing medium, for example commercially available cell freezing medium, e.g., C2695, C2639 or C6039 (Sigma).
  • Cryopreservation medium preferably comprises DMSO (dimethylsulfoxide), at a concentration of, e.g., about 10% (v/v).
  • Cryopreservation medium may comprise additional agents, for example, methylcellulose and/or glycerol.
  • Placental stem cells are preferably cooled at about l°C/min during cryopreservation.
  • a preferred cryopreservation temperature is about -80 0 C to about -180 0 C, preferably about -125°C to about -14O 0 C.
  • Cryopreserved cells can be transferred to liquid nitrogen prior to thawing for use. In some embodiments, for example, once the ampoules have reached about -9O 0 C, they are transferred to a liquid nitrogen storage area. Cryopreservation can also be done using a controlled-rate freezer.
  • Cryopreserved cells preferably are thawed at a temperature of about 25 0 C to about 40 0 C, preferably to a temperature of about 37°C.
  • Placental stem cell populations can be used to treat any disease, disorder or condition that is amenable to treatment by administration of a population of stem cells.
  • "treat” encompasses the cure of, remediation of, improvement of, lessening of the severity of, or reduction in the time course of, a disease, disorder or condition, or any parameter or symptom thereof.
  • Placental stem cells, and populations of placental stem cells can be induced to differentiate into a particular cell type, either ex vivo or in vivo, in preparation for administration to an individual in need of stem cells, or cells differentiated from stem cells. For example, placental stem cells can be injected into a damaged organ, and for organ neogenesis and repair of injury in vivo.
  • Such injury may be due to such conditions and disorders including, but not limited to, myocardial infarction, seizure disorder, multiple sclerosis, stroke, hypotension, cardiac arrest, ischemia, inflammation, thyroiditis, age-related loss of cognitive function, radiation damage, cerebral palsy, neurodegenerative disease, Alzheimer's disease, Parkinson's disease, Leigh disease, AIDS dementia, memory loss, amyotrophic lateral sclerosis, muscular dystrophy, ischemic renal disease, brain or spinal cord trauma, heart-lung bypass, glaucoma, retinal ischemia, or retinal trauma.
  • Placental stem cells can be used to treat autoimmune conditions such as juvenile diabetes, lupus, muscular dystrophy, rheumatoid arthritis, and the like.
  • Isolated populations of placental stem cells can be used, in specific embodiments, in autologous or heterologous enzyme replacement therapy to treat specific diseases or conditions, including, but not limited to lysosomal storage diseases, such as Tay-Sachs, Niema ⁇ n-Pick, Fabry's, Gaucher's disease (e.g., glucocerbrosidase deficiency), Hunter's, and Hurler's syndromes, Maroteaux-Lamy syndrome, fucosidosis (fucosidase deficiency), Batten disease (CLN3), as well as other gangliosidoses, mucopolysaccharidoses, and glycogenoses.
  • lysosomal storage diseases such as Tay-Sachs, Niema ⁇ n-Pick, Fabry's, Gaucher's disease (e.g., glucocerbrosidase deficiency), Hunter's, and Hurler's syndromes, Maroteaux-Lamy syndrome
  • Isolated populations of placental stem cells may be used alone, or as autologous or heterologous transgene carriers in gene therapy, to correct inborn errors of metabolism, cystic fibrosis, adrenoleukodystrophy (e.g., co-A ligase deficiency), metachromatic leukodystrophy (arylsulfatase A deficiency) (e.g., symptomatic, or presymptomatic late infantile or juvenile forms), globoid cell leukodystrophy (Krabbe's disease; galactocerebrosidase deficiency), acid lipase deficiency (Wolman disease), glycogen storage disease, hypothyroidism, anemia (e.g., aplastic anemia, sickle cell anemia, etc.), Pearson syndrome, Pompe's disease, phenylketonuria (PKU), porphyrias, maple syrup urine
  • adrenoleukodystrophy e.g.
  • placental stem cells can be used to treat skeletal dysplasia.
  • placental stem cells transformed to express tissue plasminogen activator (tPA) can be administered to an individual to treat thrombus.
  • tissue plasminogen activator tPA
  • isolated populations of placental stem cells may be used in autologous or heterologous tissue regeneration or replacement therapies or protocols, including, but not limited to treatment of corneal epithelial defects, treatment of osteogenesis imperfecta, cartilage repair, facial dermabrasion, mucosal membranes, tympanic membranes, intestinal linings, neurological structures (e.g., retina, auditory neurons in basilar membrane, olfactory neurons in olfactory epithelium), burn and wound repair for traumatic injuries of the skin, or for reconstruction of other damaged or diseased organs or tissues.
  • an isolated population of placental stem cells is used in hematopoietic reconstitution in an individual that has suffered a partial or total loss of hematopoietic stem cells, e.g., individuals exposed to lethal or sub-lethal doses of radiation (whether industrial, medical or military); individuals that have undergone myeloablation as part of, e.g., cancer therapy, and the like, in the treatment of, e.g., a hematologic malignancy.
  • Placental stem cells can be used in hematopoietic reconstitution in individuals having anemia (e.g., aplastic anemia, sickle cell anemia, etc.).
  • the placental stem cells are administered to such individuals with a population of hematopoietic stem cells.
  • Isolated populations of placental-derived stem cells can be used in place of, or to supplement, bone marrow or populations of stem cells derived from bone marrow.
  • approximately 1 x 10 8 to 2 x 10 8 bone marrow mononuclear cells per kilogram of patient weight are infused for engraftment in a bone marrow transplantation (i.e., about 70 ml of marrow for a 70 kg donor).
  • 70 ml requires an intensive donation and significant loss of donor blood in the donation process.
  • An isolated population of placental stem cells for hematopoietic reconstitution can comprise, in various embodiments, about, at least, or no more than 1 x 10 5 , 5 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , 5 x 10 s , 1 x 10 9 , 5 x 10 9 , 1 x 10 10 , 5 x 10 10 , 1 x 10 n or more placental stem cells.
  • placental stem cells can be used to treat patients having a blood cancer, such as a lymphoma, leukemia (such as chronic or acute myelogenous leukemia, acute lymphocytic leukemia, Hodgkin's disease, etc.), myelodysplasia, myelodysplastic syndrome, and the like.
  • a blood cancer such as a lymphoma, leukemia (such as chronic or acute myelogenous leukemia, acute lymphocytic leukemia, Hodgkin's disease, etc.), myelodysplasia, myelodysplastic syndrome, and the like.
  • the disease, disorder or condition is chronic granulomatous disease.
  • the present invention further encompasses the treatment of an individual with a stem cell combination of the invention, wherein the individual has an anemia or disorder of the blood hemoglobin.
  • the anemia or disorder may be natural (e.g., caused by genetics or disease), or may be artificially-induced (e.g., by accidental or deliberate poisoning, chemotherapy, and the like).
  • the disease or disorder is a marrow failure syndrome ⁇ e.g., aplastic anemia, Kostmann syndrome, Diamond-Blackfan anemia, amegakaryocytic thrombocytopenia, and the like), a bone marrow disorder or a hematopoietic disease or disorder.
  • a marrow failure syndrome ⁇ e.g., aplastic anemia, Kostmann syndrome, Diamond-Blackfan anemia, amegakaryocytic thrombocytopenia, and the like
  • a bone marrow disorder a hematopoietic disease or disorder.
  • Placental stem cells can also be used to treat severe combined immunodeficiency disease, including, but not limited to, combined immunodeficiency disease ⁇ e.g., Wiskott- Aldrich syndrome, severe DiGeorge syndrome, and the like).
  • combined immunodeficiency disease e.g., Wiskott- Aldrich syndrome, severe DiGeorge syndrome, and the like.
  • the placental stem cells of the invention can be used in the manufacture of a tissue or organ in vivo.
  • the methods of the invention encompass using cells obtained from the placenta, e.g., stem cells or progenitor cells, to seed a matrix and to be cultured under the appropriate conditions to allow the cells to differentiate and populate the matrix.
  • the tissues and organs obtained by the methods of the invention can be used for a variety of purposes, including research and therapeutic purposes.
  • placental stem cells and placental stem cell populations may be used for autologous and allogenic transplants, including matched and mismatched HLA type hematopoietic transplants.
  • the host is treated to reduce immunological rejection of the donor cells, or to create immunotolerance ⁇ see, e.g., U.S. Patent Nos. 5,800,539 and 5,806,529).
  • the host is not treated to reduce immunological rejection or to create immunotolerance.
  • Placental stem cells can be used in therapeutic transplantation protocols, e.g., to augment or replace stem or progenitor cells of the liver, pancreas, kidney, lung, nervous system, muscular system, bone, bone marrow, thymus, spleen, mucosal tissue, gonads, or hair. Additionally, placental stem cells may be used instead of specific classes of progenitor cells ⁇ e.g., chondrocytes, hepatocytes, hematopoietic cells, pancreatic parenchymal cells, neuroblasts, muscle progenitor cells, etc.) in therapeutic or research protocols in which progenitor cells would typically be used.
  • progenitor cells e.g., chondrocytes, hepatocytes, hematopoietic cells, pancreatic parenchymal cells, neuroblasts, muscle progenitor cells, etc.
  • the invention provides for the use of placental stem cells, particularly CD200 + placental stem cells, as an adjunct to hair replacement therapy.
  • placental stem cells e.g., CD200 + placental stem cells
  • the number of stem cells injected can be, e.g., between about 100 and about 10,000 per injection, in a volume of about 0.1 to about 1.0 ⁇ L, though more ore fewer cells in a greater or lesser volume can also be used.
  • Administration of placental stem cells to facilitate hair regrowth can comprise a single injection or multiple injections in, e.g., a regular or a random pattern in an area in which hair regrowth is desired.
  • Known hair regrowth therapies can be used in conjunction with the placental stem cells, e.g., topical minoxidil. Hair loss that can be treated using placental stem cells can be naturally-occurring (e.g., male pattern baldness) or induced (e.g., resulting from toxic chemical exposure).
  • Placental stem cells and placental stem cell populations of the invention can be used for augmentation, repair or replacement of cartilage, tendon, or ligaments.
  • prostheses e.g., hip prostheses
  • joints e.g., knee
  • Cartilage tissue constructs can also be employed in major reconstructive surgery for different types of joints (see, e.g., Resnick & Niwayama, eds., 1988, Diagnosis of Bone and Joint Disorders, 2d ed., W. B. Saunders Co.).
  • the placental stem cells of the invention can be used to repair damage to tissues and organs resulting from, e.g., trauma, metabolic disorders, or disease.
  • the trauma can be, e.g., trauma from surgery, e.g., cosmetic surgery.
  • a patient can be administered placental stem cells, alone or combined with other stem or progenitor cell populations, to regenerate or restore tissues or organs which have been damaged as a consequence of disease.
  • the present invention provides compositions comprising placental stem cells, or biomolecules therefrom.
  • the placental stem cells of the present invention can be combined with any physiologically-acceptable or medically-acceptable compound, composition or device for use in, e.g., research or therapeutics.
  • the placental stem cell populations of the invention can be preserved, for example, cryopreserved for later use.
  • Methods for cryopreservation of cells, such as stem cells are well known in the art.
  • Placental stem cell populations can be prepared in a form that is easily administrable to an individual.
  • the invention provides a placental stem cell population that is contained within a container that is suitable for medical use.
  • a container can be, for example, a sterile plastic bag, flask, jar, or other container from which the placental stem cell population can be easily dispensed.
  • the container can be a blood bag or other plastic, medically-acceptable bag suitable for the intravenous administration of a liquid to a recipient.
  • the container is preferably one that allows for cryopreservation of the combined stem cell population.
  • the cryopreserved placental stem cell population can comprise placental stem cells derived from a single donor, or from multiple donors.
  • the placental stem cell population can be completely HLA-matched to an intended recipient, or partially or completely HLA- mismatched.
  • the invention provides a composition comprising a placental stem cell population in a container.
  • the stem cell population is cryopreserved.
  • the container is a bag, flask, or jar.
  • said bag is a sterile plastic bag.
  • said bag is suitable for, allows or facilitates intravenous administration of said placental stem cell population.
  • the bag can comprise multiple lumens or compartments that are interconnected to allow mixing of the placental stem cells and one or more other solutions, e.g., a drug, prior to, or during, administration.
  • the composition comprises one or more compounds that facilitate cryopreservation of the combined stem cell population.
  • said placental stem cell population is contained within a physiologically-acceptable aqueous solution.
  • said physiologically-acceptable aqueous solution is a 0.9% NaCl solution.
  • said placental stem cell population comprises placental cells that are HLA-matched to a recipient of said stem cell population.
  • said combined stem cell population comprises placental cells that are at least partially HLA-mismatched to a recipient of said stem cell population.
  • said placental stem cells are derived from a plurality of donors.
  • compositions for use in vivo can be formulated into pharmaceutical compositions for use in vivo.
  • Such pharmaceutical compositions comprise a population of placental stem cells, or a population of cells comprising placental stem cells, in a pharmaceutically-acceptable carrier, e.g., a saline solution or other accepted physiologically-acceptable solution for in vivo administration.
  • Pharmaceutical compositions of the invention can comprise any of the placental stem cell populations, or placental stem cell types, described elsewhere herein.
  • the pharmaceutical compositions can comprise fetal, maternal, or both fetal and maternal placental stem cells.
  • the pharmaceutical compositions of the invention can further comprise placental stem cells obtained from a single individual or placenta, or from a plurality of individuals or placentae.
  • the pharmaceutical compositions of the invention can comprise any number of placental stem cells.
  • a single unit dose of placental stem cells can comprise, in various embodiments, about, at least, or no more than 1 x 10 5 , 5 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , 5 x 10 8 , 1 x 1O 9 , 5 x 10 9 , 1 x 10 10 , 5 x 10 10 , 1 x l ⁇ " or more placental stem cells.
  • compositions of the invention comprise populations of cells that comprise 50% viable cells or more (that is, at least 50% of the cells in the population are functional or living). Preferably, at least 60% of the cells in the population are viable. More preferably, at least 70%, 80%, 90%, 95%, or 99% of the cells in the population in the pharmaceutical composition are viable.
  • compositions of the invention can comprise one or more compounds that, e.g., facilitate engraftment ⁇ e.g., anti-T-cell receptor antibodies, an immunosuppressant, or the like); stabilizers such as albumin, dextran 40, gelatin, hydroxyethyl starch, and the like.
  • the pharmaceutical composition of the invention comprises about 1.25% HSA and about 2.5% dextran.
  • Other injectable formulations, suitable for the administration of cellular products, may be used.
  • the composition of the invention comprises placental stem cells that are substantially, or completely, non-maternal in origin.
  • the invention provides in one embodiment a composition comprising a population of placental stem cells that are CD200 + and HLA-G + ; CD73 + , CD105 + , and CD200 + ; CD200 + and OCT-4 + ; CD73 + , CD105 + and HLA-G + ; CD73 + and CDl 05 + and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said population of placental stem cell when said population of placental cells is cultured under conditions that allow the formation of an embryoid-like body; or OCT-4 + and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said population of placental stem cell when said population of placental cells is cultured under conditions that allow the formation of an embryoid-like body; or a combination of the foregoing, wherein at least 70%, 80%, 90%, 95% or 99% of said placental stem cells are non-maternal in origin.
  • the placental stem cells of the invention can be used to produce conditioned medium, that is, medium comprising one or more biomolecules secreted or excreted by the stem cells.
  • the conditioned medium comprises medium in which placental stem cells have grown for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 or more days.
  • the conditioned medium comprises medium in which placental stem cells have grown to at least 30%, 40%, 50%, 60%, 70%, 80%, 90% confluence, or up to 100% confluence.
  • Such conditioned medium can be used to support the culture of a separate population of placental stem cells, or stem cells of another kind.
  • the conditioned medium comprises medium in which placental stem cells have been differentiated into an adult cell type.
  • the conditioned medium of the invention comprises medium in which placental stem cells and non-placental stem cells have been cultured.
  • the invention further comprises matrices, hydrogels, scaffolds, and the like that comprise a placental stem cell, or a population of placental stem cells.
  • Placental stem cells of the invention can be seeded onto a natural matrix, e.g., a placental biomaterial such as an amniotic membrane material.
  • a placental biomaterial such as an amniotic membrane material.
  • an amniotic membrane material can be, e.g., amniotic membrane dissected directly from a mammalian placenta; fixed or heat-treated amniotic membrane, substantially dry (i.e., ⁇ 20% H 2 O) amniotic membrane, chorionic membrane, substantially dry chorionic membrane, substantially dry amniotic and chorionic membrane, and the like.
  • Preferred placental biomaterials on which placental stem cells can be seeded are described in Hariri, U.S. Application Publication No. 2004/0048796.
  • Placental stem cells of the invention can be suspended in a hydrogel solution suitable for, e.g., injection.
  • Suitable hydrogels for such compositions include self-assembling peptides, such as RAD 16.
  • a hydrogel solution comprising the cells can be allowed to harden, for instance in a mold, to form a matrix having cells dispersed therein for implantation. Placental stem cells in such a matrix can also be cultured so that the cells are mitotically expanded prior to implantation.
  • the hydrogel is, e.g., an organic polymer (natural or synthetic) that is cross-linked via covalent, ionic, or hydrogen bonds to create a three-dimensional open-lattice structure that entraps water molecules to form a gel.
  • Hydrogel-forming materials include polysaccharides such as alginate and salts thereof, peptides, polyphosphazines, and polyacrylates, which are crosslinked ionically, or block polymers such as polyethylene oxide-polypropylene glycol block copolymers which are crosslinked by temperature or pH, respectively.
  • the hydrogel or matrix of the invention is biodegradable.
  • the formulation comprises an in situ polymerizable gel (see., e.g., U.S. Patent Application Publication 2002/0022676; Anseth et al., J. Control Release, 78(1 -3): 199-209 (2002); Wang et ah, Biomaterials, 24(22):3969-80
  • the polymers are at least partially soluble in aqueous solutions, such as water, buffered salt solutions, or aqueous alcohol solutions, that have charged side groups, or a monovalent ionic salt thereof.
  • aqueous solutions such as water, buffered salt solutions, or aqueous alcohol solutions
  • polymers having acidic side groups that can be reacted with cations are poly(phosphazenes), poly(acrylic acids), poly(methacrylic acids), copolymers of acrylic acid and methacrylic acid, poly( vinyl acetate), and sulfonated polymers, such as sulfonated polystyrene.
  • Copolymers having acidic side groups formed by reaction of acrylic or methacrylic acid and vinyl ether monomers or polymers can also be used.
  • acidic groups are carboxylic acid groups, sulfonic acid groups, halogenated (preferably fluorinated) alcohol groups, phenolic OH groups, and acidic OH groups.
  • the placental stem cells of the invention or co-cultures thereof can be seeded onto a three-dimensional framework or scaffold and implanted in vivo.
  • a three-dimensional framework or scaffold can be implanted in combination with any one or more growth factors, cells, drugs or other components that stimulate tissue formation or otherwise enhance or improve the practice of the invention.
  • Nonwoven mats can be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (e.g., PGA/PLA)
  • Foams composed of, e.g., poly( ⁇ - caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, formed by processes such as freeze-drying, or lyophilization (see, e.g., U.S. Pat. No. 6,355,699), can also be used as scaffolds.
  • Placental stem cells of the invention can also be seeded onto, or contacted with, a physiologically-acceptable ceramic material including, but not limited to, mono-, di-, tri-, alpha-tri-, beta-tri-, and tetra-calcium phosphate, hydroxyapatite, fluoroapatites, calcium sulfates, calcium fluorides, calcium oxides, calcium carbonates, magnesium calcium phosphates, biologically active glasses such as BIOGLASS ® , and mixtures thereof.
  • a physiologically-acceptable ceramic material including, but not limited to, mono-, di-, tri-, alpha-tri-, beta-tri-, and tetra-calcium phosphate, hydroxyapatite, fluoroapatites, calcium sulfates, calcium fluorides, calcium oxides, calcium carbonates, magnesium calcium phosphates, biologically active glasses such as BIOGLASS ® , and mixtures thereof.
  • Porous biocompatible ceramic materials currently commercially available include SURGIBONE ® (CanMedica Corp., Canada), ENDOBON ® (Merck Biomaterial France, France), CEROS ® (Mathys, AG 3 Bettlach, Switzerland), and mineralized collagen bone grafting products such as HEALOSTM (DePuy, Inc., Raynham, MA) and VITOSS ® , RHAK ⁇ SSTM, and CORTOSS ® (Orthovita, Malvern, Pa.).
  • the framework can be a mixture, blend or composite of natural and/or synthetic materials.
  • placental stem cells can be seeded onto, or contacted with, a felt, which can be, e.g., composed of a multifilament yarn made from a bioabsorbable material such as PGA, PLA, PCL copolymers or blends, or hyaluronic acid.
  • the placental stem cells of the invention can, in another embodiment, be seeded onto foam scaffolds that may be composite structures. Such foam scaffolds can be molded into a useful shape, such as that of a portion of a specific structure in the body to be repaired, replaced or augmented.
  • the framework is treated, e.g., with 0.1M acetic acid followed by incubation in polylysine, PBS, and/or collagen, prior to inoculation of the cells of the invention in order to enhance cell attachment.
  • External surfaces of a matrix may be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma-coating the matrix, or addition of one or more proteins (e.g., collagens, elastic fibers, reticular fibers), glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc.), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, and the like.
  • proteins e.g., collagens, elastic fibers, reticular fibers
  • the scaffold comprises, or is treated with, materials that render it non-thrombogenic. These treatments and materials may also promote and sustain endothelial growth, migration, and extracellular matrix deposition. Examples of these materials and treatments include but are not limited to natural materials such as basement membrane proteins such as laminin and Type IV collagen, synthetic materials such as EPTFE, and segmented polyurethaneurea silicones, such as PURSPANTM (The Polymer Technology Group, Inc., Berkeley, Calif.).
  • the scaffold can also comprise anti-thrombotic agents such as heparin; the scaffolds can also be treated to alter the surface charge (e.g., coating with plasma) prior to seeding with placental stem cells.
  • Mammalian placental cells can be conditionally immortalized by transfection with any suitable vector containing a growth-promoting gene, that is, a gene encoding a protein that, under appropriate conditions, promotes growth of the transfected cell, such that the production and/or activity of the growth-promoting protein is regulatable by an external factor.
  • a growth-promoting gene is an oncogene such as, but not limited to, v-myc, N-myc, c-myc, p53, SV40 large T antigen, polyoma large T antigen, EIa adenovirus or E7 protein of human papillomavirus.
  • External regulation of the growth-promoting protein can be achieved by placing the growth-promoting gene under the control of an externally-regulatable promoter, e.g., a promoter the activity of which can be controlled by, for example, modifying the temperature of the transfected cells or the composition of the medium in contact with the cells, in one embodiment, a tetracycline (tet)-controlled gene expression system can be employed (see Gossen et al, Proc. Natl. Acad. Sci. USA 89:5547-5551, 1992; Hoshimaru et al, Proc. Natl. Acad. Sci. USA 93:1518-1523, 1996).
  • an externally-regulatable promoter e.g., a promoter the activity of which can be controlled by, for example, modifying the temperature of the transfected cells or the composition of the medium in contact with the cells
  • tet tetracycline
  • tTA tet-controlled transactivator
  • tTA is a fusion protein of the repressor (tetR) of the transposon-10-derived tet resistance operon of Escherichia coli and the acidic domain of VP16 of herpes simplex virus.
  • the vector further contains a gene encoding a selectable marker, e.g., a protein that confers drug resistance.
  • a selectable marker e.g., a protein that confers drug resistance.
  • the bacterial neomycin resistance gene (neo ⁇ ) is one such marker that may be employed within the present invention.
  • Cells carrying neo ⁇ may be selected by means known to those of ordinary skill in the art, such as the addition of, e.g., 100-200 ⁇ g/mL G418 to the growth medium.
  • Transfection can be achieved by any of a variety of means known to those of ordinary skill in the art including, but not limited to, retroviral infection.
  • a cell culture may be transfected by incubation with a mixture of conditioned medium collected from the producer cell line for the vector and DMEM/F12 containing N2 supplements.
  • a placental cell culture prepared as described above may be infected after, e.g., five days in vitro by incubation for about 20 hours in one volume of conditioned medium and two volumes of DMEM/F12 containing N2 supplements.
  • Transfected cells carrying a selectable marker may then be selected as described above.
  • the substrate is a polyornithine/laminin substrate, consisting of tissue culture plastic coated with polyornithine (10 ⁇ g/mL) and/or laminin (10 ⁇ g/mL), a polylysine/laminin substrate or a surface treated with fibronectin.
  • Cultures are then fed every 3-4 days with growth medium, which may or may not be supplemented with one or more proliferation-enhancing factors. Proliferation-enhancing factors may be added to the growth medium when cultures are less than 50% confluent.
  • conditionally-immortalized placental stem cell lines can be passaged using standard techniques, such as by trypsinization, when 80-95% confluent. Up to approximately the twentieth passage, it is, in some embodiments, beneficial to maintain selection (by, for example, the addition of G418 for cells containing a neomycin resistance gene). Cells may also be frozen in liquid nitrogen for long-term storage.
  • Clonal cell lines can be isolated from a conditionally-immortalized human placental stem cell line prepared as described above. In general, such clonal cell lines may be isolated using standard techniques, such as by limit dilution or using cloning rings, and expanded. Clonal cell lines may generally be fed and passaged as described above. [0247] Conditionally-immortalized human placental stem cell lines, which may, but need not, be clonal, may generally be induced to differentiate by suppressing the production and/or activity of the growth-promoting protein under culture conditions that facilitate differentiation.
  • the conditions e.g., temperature or composition of medium
  • differentiation can be achieved by the addition of tetracycline to suppress transcription of the growth- promoting gene.
  • 1 ⁇ g/mL tetracycline for 4-5 days is sufficient to initiate differentiation.
  • additional agents may be included in the growth medium.
  • the placental stem cells for the present invention can be used in assays to determine the influence of culture conditions, environmental factors, molecules (e.g., biomolecules, small inorganic molecules, etc.) and the like on stem ceil proliferation, expansion, and/or differentiation, compared to placental stem cells not exposed to such conditions.
  • the placental stem cells of the present invention are assayed for changes in proliferation, expansion or differentiation upon contact with a molecule.
  • the invention provides a method of identifying a compound that modulates the proliferation of a plurality of placental stem cells, comprising contacting said plurality of stem cells with said compound under conditions that allow proliferation, wherein if said compound causes a detectable change in proliferation of said plurality of stem cells compared to a plurality of stem cells not contacted with said compound, said compound is identified as a compound that modulates proliferation of placental stem cells.
  • said compound is identified as an inhibitor of proliferation.
  • said compound is identified as an enhancer of proliferation.
  • the invention provides a method of identifying a compound that modulates the expansion of a plurality of placental stem cells, comprising contacting said plurality of stem cells with said compound under conditions that allow expansion, wherein if said compound causes a detectable change in expansion of said plurality of stem cells compared to a plurality of stem cells not contacted with said compound, said compound is identified as a compound that modulates expansion of placental stem cells.
  • said compound is identified as an inhibitor of expansion.
  • said compound is identified as an enhancer of expansion.
  • the invention provides a method of identifying a compound that modulates the differentiation of a placental stem cell, comprising contacting said stem cells with said compound under conditions that allow differentiation, wherein if said compound causes a detectable change in differentiation of said stem cells compared to a stem cell not contacted with said compound, said compound is identified as a compound that modulates proliferation of placental stem cells.
  • said compound is identified as an inhibitor of differentiation.
  • said compound is identified as an enhancer of differentiation.
  • Placental stem cells are obtained from a post-partum mammalian placenta either by perfusion or by physical disruption, e.g., enzymatic digestion.
  • the cells are cultured in a culture medium comprising 60% DMEM-LG (Gibco), 40% MCDB-201 (Sigma), 2% fetal calf serum (FCS) (Hyclone Laboratories), Ix insulin-transferrin-selenium (ITS), Ix lenolenic- acid-bovine-serum-albumin (LA-BSA) 5 10 "9 M dexamethasone (Sigma), 10 "4 M ascorbic acid 2-phosphate (Sigma), epidermal growth factor (EGF) 10ng/ml (R&D Systems), platelet derived-growth factor (PDGF-BB) 10ng/ml (R&D Systems), and IOOU penicillin/100OU streptomycin.
  • DMEM-LG Gabco
  • FCS fetal calf serum
  • ITS Ix insulin-transferrin-selenium
  • LA-BSA Ix lenolenic- acid-bovine-serum-albumin
  • the culture flask in which the cells are cultured is prepared as follows. T75 flasks are coated with fibronectin (FN), by adding 5 ml PBS containing 5ng/ml human FN (Sigma F0895) to the flask. The flasks with FN solution are left at 37°C for 30 min. The FN solution is then removed prior to cell culture. There is no need to dry the flasks following treatment. Alternatively, the flasks are left in contact with the FN solution at 4°C overnight or longer; prior to culture, the flasks are warmed and the FN solution is removed.
  • FN fibronectin
  • Cultures of placental stem cells from placental perfusate are established as follows. Cells from a Ficoll gradient are seeded in FN-coated T75 flasks, prepared as above, at 50- 100x10 6 cells/flask in 15 ml culture medium. Typically, 5 to 10 flasks are seeded. The flasks are incubated at 37 0 C for 12-18 hrs to allow the attachment of adherent cells. 10 ml of warm PBS is added to each flask to remove cells in suspension, and mixed gently. 15 rnL of the medium is then removed and replaced with 15 ml fresh culture medium. All medium is changed 3-4 days after the start of culture. Subsequent culture medium changes are performed, during which 50% or 7.5 ml of the medium is removed.
  • the culture is checked under a microscope to examine the growth of the adherent cell colonies.
  • adherent cells are harvested by trypsin digestion. Cells harvested from these primary cultures are designated passage 0 (zero).
  • Placental Stem Cells Isolated By Physical Disruption and Enzymatic Digestion [0256] Placental stem cell cultures are established from digested placental tissue as follows. The perfused placenta is placed on a sterile paper sheet with the maternal side up. Approximately 0.5 cm of the surface layer on maternal side of placenta is scraped off with a blade, and the blade is used to remove a placental tissue block measuring approximately 1 x 2 x 1 cm. This placenta tissue is then minced into approximately lmm 3 pieces.
  • tissue/cell pellet is resuspended in 130 mL culture medium, and the cells are seeded at 13ml per fibronectin- coated T-75 flask. Cells are incubated at 37°C with a humidified atmosphere with 5% CO 2 . Placental Stem Cells are optionally cryopreserved at this stage.
  • Cryopreserved cells are quickly thawed in a 37 0 C water bath. Placental stem cells are immediately removed from the cryovial with 10ml warm medium and transferred to a 15ml sterile tube. The cells are centrifuged at 40Og for 10 minutes at room temperature. The cells are gently resuspended in 10ml of warm culture medium by pipetting, and viable cell counts are determined by Trypan blue exclusion. Cells are then seeded at about 6000-7000 cells per cm 2 onto FN-coated flasks, prepared as above (approximately 5x10 5 cells per T-75 flask). The cells are incubated at 37°C, 5% CO 2 and 90% humidity.
  • the cells After counting the cells and determining viability as above, the cells are centrifuged at 1000 RPM for 5 minutes at room temperature. Cells are passaged by gently resuspending the cell pellet from one T-75 flask with culture medium, and evenly plating the cells onto two FN-coated T-75 flasks.
  • exemplary populations of adherent placental stem cells are identified that express markers CD105, CD33, CD73, CD29, CD44, CDlO, and CD90. These populations of cells typically does not express CD34, CD45, CDl 17 or CD133. Some, but not all cultures of these placental stem cells expressed HLA-ABC and/or HLA-DR.
  • Placental stem cells were obtained from the following sources: (1) placental perfusate (from perfusion of the placental vasculature); and enzymatic digestions of (2) amnion, (3) chorion, (4) amnion-chorion plate, and (5) umbilical cord.
  • placental perfusate from perfusion of the placental vasculature
  • the various placental tissues were cleaned in sterile PBS (Gibco-Invitrogen Corporation, Carlsbad, CA) and placed on separate sterile Petri dishes.
  • the various tissues were minced using a sterile surgical scalpel and placed into 50 mL Falcon Conical tubes.
  • the minced tissues were digested with IX Collagenase (Sigma- Aldrich, St. Louis, MO) for 20 minutes in a 37 0 C water bath, centrifuged, and then digested with 0.25% Trypsin-EDTA (Gibco-Invitrogen Corp) for 10 minutes in a 37 0 C water bath.
  • the various tissues were centrifuged after digestion and rinsed once with sterile PBS (Gibco-Invitrogen Corp).
  • the reconstituted cells were then filtered twice, once with 100 ⁇ m cell strainers and once with 30 ⁇ m separation filters, to remove any residual extracellular matrix or cellular debris.
  • Cells that were HLA ABC7CD457CD347CD133 "1" were selected for characterization. Cells having this phenotype were identified, quantified, and characterized by two of Becton- Dickinson flow cytometers, the FACSCalibur and the FACS Aria (Becton-Dickinson, San Jose, CA, USA). The various placental cells were stained, at a ratio of about 10 ⁇ L of antibody per 1 million cells, for 30 minutes at room temperature on a shaker.
  • the cells were rinsed once to remove unbound antibodies and were fixed overnight with 4% paraformaldehyde (USB, Cleveland, OH) at 4 0 C. The following day, the cells were rinsed twice, filtered through a 30 ⁇ m separation filter, and were run on the flow cytometer(s). W
  • placental cells from perfusate, amnion, or chorion, prior to any culture, was stained with 7-Amino-Actinomycin D (7AAD; BD Biosciences Pharmingen) and monoclonal antibodies specific for the phenotype of interest.
  • the cells were stained at a ratio of 10 ⁇ L of antibody per 1 million cells, and were incubated for 30 minutes at room temperature on a shaker. These cells were then positively sorted for live cells expressing the phenotype of interest on the BD FACS Aria and plated into culture. Sorted (population of interest) and "All" (non-sorted) placental cell populations were plated for comparisons.
  • the cells were plated onto a fibronectin (Sigma-Aldrich) coated 96 well plate at the cell densities listed in Table 1 (cells/cm 2 ). The cell density, and whether the cell type was plated in duplicate or triplicate, was determined and governed by the number of cells expressing the phenotype of interest.
  • fibronectin Sigma-Aldrich
  • Complete medium (60% DMEM-LG (Gibco) and 40% MCDB-201 (Sigma); 2% fetal calf serum (Hyclone Labs.); Ix insulin-transferrin-selenium (ITS); Ix Iinoleic acid-bovine serum albumin (LA-BSA); 10 "9 M dexamethasone (Sigma); 10 "4 M ascorbic acid 2-phosphate (Sigma); epidermal growth factor 10 ng/mL (R&D Systems); and platelet-derived growth factor (PDGF-BB) 10 ng/mL (R&D Systems)) was added to each well of the 96 well plate and the plate was placed in a 5% CO 2 /37 C incubator. On day 7, 100 ⁇ L of complete medium was added to each of the wells. The 96 well plate was monitored for about two weeks and a final assessment of the culture was completed on day 12. This is very early in the placental stem cell culture, and represents passage 0 cells.
  • FACSCalibur data was analyzed in Flow Jo (Tree star, Inc) using standard gating techniques.
  • the BD FACS Aria data was analyzed using the FACSDiva software (Becton- Dickinson).
  • the FACS Aria data was analyzed using doublet discrimination gating to minimize doublets, as well as, standard gating techniques. All results were compiled in Microsoft Excel and all values, herein, are represented as average ⁇ standard deviation (number, standard error of mean).
  • Post-digestion viability was assessed using the manual trypan blue exclusion method (FIG 1).
  • the average viability of cells obtained from the majority of the digested tissue was around 70%.
  • the BD FACS Aria data also identified amnion, perfusate, and chorion as providing higher numbers of HLA ABC " /CD457CD347CD133 + cells than the remaining sources.
  • HLA ABC7CD457CD347CD133 + cells were identified and quantified from each cell source, its cells were further analyzed and characterized for their expression of cell surface markers HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200, and CD 105.
  • Perfusate-derived cells were consistently positive for HLA-G, CD33, CDl 17, CDlO, CD44, CD200, CD90, CD38, CD105, and CD13 (FIG. 4).
  • Amnion-derived cells were consistently positive for HLA-G, CD33, CDl 17, CDlO, CD44, CD200, CD90, CD38, CD105, and CD13 (FIG 5).
  • Umbilical cord-derived cells were consistently positive for HLA-G, CD33, CD90, CD38, CD 105, and CDl 3, while the expression of CDl 17, CDlO, CD44, and CD200 varied (FIG. 8).
  • Non-sorted perfusate-derived cells plated at a cell density of 93,800/cm 2 , resulted in mostly small, round, non-adherent cells with several adherent cells located around the well peripheries.
  • Sorted amnion-derived cells plated at a cell density of 6,300/cm 2 , resulted in small, round, non-adherent cells.
  • Non-sorted amnion-derived cells, plated at a cell density of 6,300/cm 2 resulted in small, round, non-adherent cells.
  • Non-sorted amnion-derived cells plated at a cell density of 62,500/cm 2 resulted in small, round, non-adherent cells.
  • Sorted chorion-derived cells plated at a cell density of 6,300/cm 2 , resulted in small, round, non-adherent cells.
  • Non-sorted chorion-derived cells plated at a cell density of 6,300/cm 2 , resulted in small, round, non-adherent cells.
  • Non-sorted chorion-derived cells plated at a cell density of 62,500/cm 2 resulted in small, round, non-adherent cells.
  • This Example demonstrates an exemplary cell surface marker profile of placental stem cells.
  • Placental stem cells or umbilical cord stem cells, obtained by enzymatic digestion, in culture medium were washed once by adding 2 mL 2% FBS-PBS and centrifuging at 40Og for 5 minutes. The supernatant was decanted, and the pellet was resuspended in 100-200 ⁇ L 2% FBS-PBS.
  • Control tubes were prepared as follows:
  • control and sample tubes were incubated in the dark at room temperature for 30 minutes. After incubation, the tubes were washed by adding 2mL 2% FBS-PBS and centrifuging at 40Og for 5 minutes. The supernatant was decanted, and the pellet was resuspended in 100-200 ⁇ L 2% FBS-PBS and acquire on flow cytometer. All other antibodies were used following this procedure. [0289J Matched placental stem cells from amniotic membrane and umbilical cord stem cells were analyzed using fluorescently-labeled antibodies and flow cytometry to identify cell surface markers that were present or absent.
  • CD 105 proliferation related endothelial specific marker
  • CD200 marker associated with regulatory function
  • CD34 expressed on endothelial cells and on hematopoietic stem cells
  • CDlO stem cell/precursor cell marker
  • cytokeratin K epidermal marker
  • CD44 cell migration, lymphocyte homing, hematopoeisis
  • CD45 lineage marker
  • CD 133 marker for hematopoietic progenitor cells
  • CDl 17 stem cell factor (c-Kit)
  • CD90 expressed on primitive hematopoietic stem cells in normal bone marrow, cord blood and fetal liver cells
  • HLA ABC pan MHC I, antigen presentation, immunogenicity
  • ⁇ -2 -microglobulin associates with MHC I, antigen presentation, immunogenicity
  • HLA DR 5 DQ 5 DP pan MHC II, antigen presentation, immunogenicity
  • CD80/86 co-stimuls
  • ALDEFLUOR® Assay Kit from Stem Cell
  • the assay uses ALDEFLUOR®, a fluorescent ALDH substrate (Aldagen, Inc.,
  • ALDEFLUOR® reagent is provided in a stable, inactive form.
  • the ALDEFLUOR® was activated by dissolving the dry compound in dimethylsulfoxide (DMSO) and adding 2N HCl, and was added immediately to the cells.
  • DMSO dimethylsulfoxide
  • a control tube was also established by combing the cells with ALDEFLUOR® plus DEAB, a specific inhibitor of ALDH.
  • Cells analyzed included four umbilical cord stem cell lines and three placental stem cell lines from amnion-chorion plate, a bone marrow-derived mesenchymal stem cell line
  • BM-MSC 7 an adipose-derived stem cell line (ADSC), a human villous trophoblast cell line
  • HVT HVT
  • CD34 + stem cells purified from cord blood.
  • ALDEFLUOR® substrate was activated by adding 25 ⁇ l of DMSO to the dry
  • ALDEFLUOR® Reagent and let stand for 1 minute at RT. 25 ⁇ l of 2N HCL was added and mixed well. This mixture was incubated for 15 min at RT. 360 ⁇ l of ALDEFLUOR® Assay
  • Buffer was added to the vial and mixed.
  • the resulting mixture was stored at 2-8°C during use.
  • Placental stem cell lines demonstrated ALDH activity of from about 3% to about 25% (3.53%, 8.76% and 25.26%).
  • Umbilical cord stem cell lines demonstrated ALDH activity of from about 16% to about 20% (16.59%, 17.01%, 18.44% and 19.83%).
  • BM- MSC and HVT were negative and 1.5% respectively for ALDH, but the adipose derived MSC is close to 30% ALDH + .
  • the positive control CD34 + cells purified from umbilical cord blood were, as expected, highly positive (75%) for ALDH.
  • This Example demonstrates one method of collecting placental stem cells by perfusion.
  • a post-partum placenta is obtained within 24 hours after birth.
  • the umbilical cord is clamped with an umbilical cord clamp approximately 3 to 4 inches about the placental disk, and the cord is cut above the clamp.
  • the umbilical cord is either discarded, or processed to recover, e.g., umbilical cord stem cells, and/or to process the umbilical cord membrane for the production of a biomaterial.
  • Excess amniotic membrane and chorion is cut from the placenta, leaving approximately 1 A inch around the edge of the placenta. The trimmed material is discarded.
  • the amniotic membrane is separated from the chorion using blunt dissection with the fingers.
  • the amniotic membrane is cut around the base of the umbilical cord with scissors, and detached from the placental disk.
  • the amniotic membrane can be discarded, or processed, e.g., to obtain stem cells by enzymatic digestion, or to produce, e.g., an amniotic membrane biomaterial.
  • the fetal side of the remaining placental material is cleaned of all visible blood clots and residual blood using sterile gauze, and is then sterilized by wiping with an iodine swab than with an alcohol swab.
  • the umbilical cord is then clamped crosswise with a sterile hemostat beneath the umbilical cord clamp, and the hemostat is rotated away, pulling the cord over the clamp to create a fold.
  • the cord is then partially cut below the hemostat to expose a cross-section of the cord supported by the clamp.
  • the cord is clamped with a sterile hemostat.
  • the cord is then placed on sterile gauze and held with the hemostat to provide tension.
  • the cord is then cut straight across directly below the hemostat, and the edge of the cord near the vessel is re-clamped.
  • a small volume of sterile injection grade 0.9% NaCl solution to check for leaks If no leaks are present, the pump speed is increased, and about 750 mL of the injection grade 0.9% NaCl solution is pumped through the placental vasculature. Perfusion can be aided by gently massaging the placental disk from the outer edges to the cord. When a collection bag is full, the bag is removed from the coupler connecting the tubing to the bag, and a new bag is connected to the tube.
  • each bag is placed inside a plasma extractor without disturbing the pellet of cells. The supernatant within the bags is then removed and discarded. The bag is then gently massaged to resuspend the cells in the remaining supernatant.
  • a sterile 1 mL syringe about 300-500 ⁇ L of cells is withdrawn from the collection bag, via a sampling site coupler, and transferred to a 1.5 mL centrifuge tube. The weight and volume of the remaining perfusate are determined, and 1/3 volume of hetastarch is added to the perfusate and mixed thoroughly. The number of cells per mL is determined. Red blood ceils are removed from the perfusate using a plasma extractor.
  • Placental cells are then immediately cultured to isolate placental stem cells, or are cryopreserved for later use.
  • Neuronal differentiation of placental stem cells can also be accomplished as follows:
  • Placental stem cells are grown for 24 hr in preinduction medium consisting of DMEM/20% FBS and 1 mM beta-mercaptoethanol.
  • Neuronal induction medium consisting of DMEM and 1-10 mM betamercaptoethanol is added to the cells.
  • induction media consisting of DMEM/2% DMSO/200 ⁇ M butylated hydroxyanisole may be used.
  • morphologic and molecular changes may occur as early as 60 minutes after exposure to serum-free media and betamercaptoethanol.
  • RT/PCR may be used to assess the expression of e.g., nerve growth factor receptor and neurofilament heavy chain genes.
  • AIM adipo-induction medium
  • AIMl comprised MesenCult Basal Medium for human Mesenchymal Stem Cells (StemCell Technologies) supplemented with Mesenchymal Stem Cell Adipogenic Supplements (StemCell Technologies).
  • AIM2 comprised DMEM/MCDB-201 with 2% FCS and LA-BSA (1%). About 1.25 x 10 5 placental stem cells were grown in 5 mL AIMl or AIM2 in T-25 flasks. The cells were cultured in incubators for 7-21 days. The cells developed oil droplet vacuoles in the cytoplasm, as confirmed by oil-red staining, suggesting the differentiation of the stem cells into adipocytes.
  • Adipogenic differentiation of placental stem cells can also be accomplished as follows:
  • Placental stem cells are grown in MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum. 2. Three cycles of induction/maintenance are used. Each cycle consists of feeding the placental stem cells with Adipogenesis Induction Medium (Cambrex) and culturing the cells for 3 days (at 37°C, 5% CO 2 ), followed by 1-3 days of culture in Adipogenesis Maintenance Medium (Cambrex).
  • An alternate induction medium that can be used contains 1 ⁇ M dexamethasone, 0.2 mM indomethacin, 0.01 mg/ml insulin, 0.5 mM IBMX, DMEM-high glucose, FBS 5 and antibiotics.
  • the cells are cultured for an additional 7 days in adipogenesis maintenance medium, replacing the medium every 2-3 days.
  • a hallmark of adipogenesis is the development of multiple intracytoplasmic lipid vesicles that can be easily observed using the lipophilic stain oil red O. Expression of lipase and/or fatty acid binding protein genes is confirmed by RT/PCR in placental stem cells that have begun to differentiate into adipocytes.
  • Osteogenic medium was prepared from 185 mL Cambrex Differentiation Basal Medium - Osteogenic and SingleQuots (one each of dexamethasone, 1-glutamine, ascorbate, pen/strep, MCGS 5 and ⁇ -glycerophosphate). Placental stem cells from perfusate were plated, at about 3 x 10 3 cells per cm 2 of tissue culture surface area in 0.2-0.3 mL MSCGM per cm 2 tissue culture area. Typically, all cells adhered to the culture surface for 4-24 hours in MSCGM at 37°C in 5% CO 2 . Osteogenic differentiation was induced by replacing the medium with Osteogenic Differentiation medium.
  • Osteogenic differentiation can also be accomplished as follows:
  • Adherent cultures of placental stem cells are cultured in MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum.
  • Osteogenic differentiation is induced by replacing MSCGM with Osteogenic Induction Medium (Cambrex) containing 0.1 ⁇ M dexamethasone, 0.05 mM ascorbic acid-2-phosphate, 10 mM beta glycerophosphate.
  • Pancreatic differentiation is accomplished as follows:
  • Placental stem cells are cultured in DMEM/20% CBS, supplemented with basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml. KnockOut Serum Replacement may be used in lieu of CBS.
  • Conditioned media from nestin-positive neuronal cell cultures is added to media at a 50/50 concentration.
  • Differentiation is characterized by assaying for insulin protein or insulin gene expression by RTYPCR.
  • Placental stem cells are cultured in DMEM/20% CBS, supplemented with retinoic acid, 1 ⁇ M; basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml; and epidermal growth factor, 100 ng/ml.
  • KnockOut Serum Replacement (Invitrogen, Carlsbad, California) may be used in lieu of CBS.
  • placental stem cells are cultured in DMEM/20% CBS supplemented with 50 ng/ml Cardiotropin-1 for 24 hours.
  • placental stem cells are maintained in protein-free media for 5-7 days, then stimulated with human myocardium extract (escalating dose analysis).
  • Myocardium extract is produced by homogenizing I gm human myocardium in
  • Cells are cultured for 10-14 days, refeeding every 3-4 days.
  • Placental stem cells are maintained in MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum.
  • Placental stem cells are aliquoted into a sterile polypropylene tube. The cells are centrifuged (150 x g for 5 minutes), and washed twice in Incomplete Chondrogenesis Medium (Cambrex).
  • the cell pellets are fed every 2-3 days with freshly prepared complete chondrogenesis medium.
  • Pellets are maintained suspended in medium by daily agitation using a low speed vortex.
  • Chondrogenesis is characterized by e.g., observation of production of esoinophilic ground substance, assessing cell morphology, an/or RT/PCR confirmation of collagen 2 and/or collagen 9 gene expression and/or the production of cartilage matrix acid mucopolysaccharides, as confirmed by Alcian blue cytochemical staining.
  • Cartilage is an avascular, alymphatic tissue that lacks a nerve supply. Cartilage has a low chondrocyte density ( ⁇ 5%), however these cells are surprisingly efficient at maintaining the extracellular matrix around them.
  • articular cartilage which facilitates joint lubrication in joints
  • fibrocartilage which provides shock absorption in, e.g., meniscus and intervertebral disc
  • elastic cartilage which provides anatomical structure in, e.g., nose and ears.
  • AU three types of cartilage are similar in biochemical structure.
  • Joint pain is a major cause of disability and provides an unmet need of relief in the area of orthopedics.
  • Primary osteoarthritis (which can cause joint degeneration), and trauma are two common causes of pain.
  • Approximately 9% of the U.S. population has osteoarthritis of hip or knee, and more than 2 million knee surgeries are performed yearly.
  • Current treatments are more geared towards treatment of symptoms rather than repairing the cartilage.
  • Natural repair occurs when f ⁇ broblast-like cells invade the area and fill it with fibrous tissue which is neither as resilient or elastic as the normal tissue, hence causing more damage.
  • Treatment options historically included tissue grafts, subchondral drilling, or total joint replacement.
  • CARTICEL® an autologous chondrocyte injection
  • SYNVISC® and ORTHOVISC® which are hyaluronic acid injections for temporary pain relief
  • CHONDROGENTM an injection of adult mesenchymal stem cells for meniscus repair.
  • Placental and umbilical cord stem cells were isolated and purified from full term human placenta by enzymatic digestion.
  • Human MSC cells and HDF cells were purchased from Cambrex, and MC3T3 cells were purchased from American Type Culture Collection. All cell lines used were centrifuged into pellets in polypropylene centrifuge tubes at 800 RPM for 5 minutes and grown in both chondrogenic induction media (Cambrex) and non- inducing basal MSC media (Cambrex). Pellets were harvested and histologically analyzed at 7, 14, 21 and 28 days by staining for glycosaminoglycans (GAGs) with Alcian Blue, and/or for collagens with Sirius Red. Collagen type was further assessed with immunostaining. RNA analysis for cartilage-specific genes was performed at 7 and 14 days. [0323] Results
  • Experiment 1 Chondrogenesis studies were designed to achieve three main objectives: (1) to demonstrate that placental and umbilical cord stem cells can differentiate and form cartilage tissue; (2) to demonstrate that placental and umbilical cord stem cells can differentiate functionally into chondrocytes; and (3) to validate results obtained with the stem cells by evaluating control cell lines.
  • objective 1 in a preliminary study, one placental stem cell line was cultured in chondrogenic induction medium in the form of cell pellets, either with or without bone morphogenic protein (BMP) at a final concentration of 500 ng/mL. Pellets were assessed for evidence of chondrogenic induction every week for 4 weeks. Results indicated that the pellets do increase in size over time.
  • BMP bone morphogenic protein
  • BMP + and BMP " samples were also histologically analyzed for GAG's, an indicator of cartilage tissue, by staining with Alcian Blue.
  • BMP + cells generally appeared more metabolically active with pale vacuoles whereas BMP " cells were smaller with dense-stained nuclei and less cytoplasm (reflects low metabolic activity).
  • BMP + cells had stained heavily blue, while BMP " had stained only faintly.
  • both BMP + and BMP " cells were roughly equivalently stained with Alcian Blue.
  • cell density decreased over time, and matrix overtook the pellet.
  • the MC3T3 negative cell line did not demonstrate any presence of GAG when stained with Alcian Blue.
  • pellets from placental stem cells or umbilical cord stem cells were much larger than the MSC controls. Control pellets in non-induction media started to fall apart by Day 11, and were much smaller at 28 days than pellets developed by cells cultured in chondrogenic induction medium. Visually, there were no differences between pellets formed by placental stem cells or umbilical cord.
  • the UC67249 stem cell line which was initiated in dexamethasone-free media, formed larger pellets. Negative control MC3T3 cells did not form pellets; however, HDFs did form pellets.
  • pellets from all test groups were then subjected to histological analysis for GAG's and collagen.
  • pellets formed by the stem cells under inducing conditions were much larger and stayed intact better than pellets formed under non-inducing conditions.
  • Pellets formed under inducing conditions showed production of GAGs and increasing collagen content over time, and as early as seven days, while pellets formed under non-inducing conditions showed little to no collagen production, as evidenced by weak Alcian Blue staining.
  • the placental stem cells and umbilical cord stem cells appeared, by visual inspection, to produce tougher, larger pellets, and appeared to be producing more collagen over time, than the hMSCs.
  • Non-induced placental stem cells produced much less type II collagen, if any, compared to the induced stem cells. Over the 28-day period, cell density decreased as matrix production increased, a characteristic of cartilage tissue. [0329] These studies confirm that placental and umbilical cord stem cells can be differentiated along a chondrogenic pathway, and can easily be induced to form cartilage tissue. Initial observations indicate that such stem cells are preferable to MSCs for the formation of cartilage tissue.
  • Placental adherent stem cells in culture are trypsinized at 37°C for about 5 minutes, and loosened from the culture dish by tapping. 10% FBS is added to the culture to stop trypsinization.
  • the cells are diluted to about 1 x 10 4 cells per mL in about 5 mL of medium. Drops (either a single drop or drops from a multi-channel micropipette are placed on the inside of the lid of a 100 mL Petri dish. The lid is carefully inverted and placed on top of the bottom of the dish, which contains about 25 ml of sterile PBS to maintain the moisture content in the dish atmosphere. Cells are grown for 6-7 days.
  • This Example demonstrates a scaled up isolation of placental stem cells by enzymatic digestion.
  • placental tissue (amnion and chorion) is obtained, macerated, and digested using equal volumes of collagenase A (1 mg/ml) (Sigma) and Trypsin-EDTA (0.25%) (Gibco-BRL) in a total volume of about 30 ml for about 30 minutes at 37°C. Cells liberated by the digestion are washed 3X with culture medium, distributed into four T-225 flasks and cultured as described in Example 1. Placental stem cell yield is between about 4 x 10 8 and 5 x 10 s cells per 1Og starting material.
  • Placental tissue is dissected and digested, followed by primary and expansion cultures to achieve an expanded cell product that produces many cell doses.
  • Cells are stored in a two-tiered cell bank and are distributed as a frozen cell product. All cell doses derived from a single donor placenta are defined as a lot, and one placenta lot is processed at a time using sterile technique in a dedicated room and Class 100 laminar flow hood.
  • the cell product is defined as being CD105 + , CD200 + 5 CDlO + , and CD34 ⁇ , having a normal karyotype and no or substantially no maternal cell content.
  • Tissue Dissection and Digestion A placenta is obtained less than 24 hours after expulsion. Placental tissue is obtained from amnion, a combination of amnion and chorion, or chorion. The tissue is minced into small pieces, about 1 mm in size. Minced tissue is digested in lmg/ml Collagenase IA for 1 hour at 37°C followed by Trypsin-EDTA for 30 minutes at 37°C. After three washes in 5% FBS in PBS, the tissue is resuspended in culture medium.
  • Primary Culture The purpose of primary culture is to establish cells from digested placental tissue.
  • the digested tissue is suspended in culture medium and placed into Corning T-flasks, which are incubated in a humidified chamber maintained at 37°C with 5% CO 2 .
  • Half of the medium is replenished after 5 days of culture.
  • High-density colonies of cells form by 2 weeks of culture. Colonies are harvested with Trypsin-EDTA, which is then quenched with 2% FBS in PBS. Cells are centrifuged and resuspended in culture medium for seeding expansion cultures. These cells are defined as Passage 0 cells having doubled 0 times.
  • Expansion Culture Cells harvested from primary culture, harvested from expansion culture, or thawed from the cell bank are used to seed expansion cultures.
  • Cell Factories NUNCTM
  • NUNCTM Cell Factories
  • Cell seeds are counted on a hemacytometer with trypan blue, and cell number, viability, passage number, and the cumulative number of doublings are recorded.
  • Cells are suspended in culture medium to about 2.3 X lO 4 cells/ml and 110 ml/tray are seeded in the Cell Factories.
  • culture medium is removed and replaced with fresh medium, followed by another treatment with 5% CO 2 in air.
  • cells reach approximately 10 5 cells/cm 2
  • cells are harvested with Trypsin-EDTA, followed by quenching with 2% FBS in PBS. Cell are then centrifuged and resuspended in culture medium.
  • Cryopreservation Cells to be frozen down are harvested from culture with Trypsin- EDTA, quenched with 2% FBS in PBS, and counted on a hemacytometer. After centrifugation.
  • cells are resuspended with 10% DMSO in FBS to a concentration of about 1 million cells/ml for cells to be used for assembly of a cell bank, and 10 million cells/ml for individual frozen cell doses.
  • the cell solution is transferred to a freezing container, which is placed in an isopropyl alcohol bath in a — 80 0 C freezer. The following day, cells are transferred to liquid nitrogen.
  • a "lot” is defined as all cell doses derived from a single donor placenta. Cells maintained normal growth, karyotype, and cell surface maker phenotype for over 8 passages and 30 doublings during expansion culture. Given this limitation, doses comprise cells from 5 passages and about 20 doublings. To generate a supply of equivalent cells, a single lot is expanded in culture and is stored in a two-tiered cell bank and frozen doses. In particular, cells harvested from the primary culture, which are defined as Passage 0 cells having undergone 0 doublings, are used to initiate an expansion culture. After the first passage, approximately 4 doublings occur, and cells are frozen in a Master Cell Bank (MCB). Vials from the MCB are used to seed additional expansion cultures.
  • MCB Master Cell Bank
  • WCB Working Cell Bank
  • Frozen containers of cells are placed into a sealed plastic bag and immersed in a 37°C water bath. Containers are gently swirled until all of the contents are melted except for a small piece of ice. Containers are removed from the sealed plastic bag and a 1OX volume of culture medium is slowly added to the cells with gentle mixing. A sample is counted on the hemacytometer and seeded into expansion cultures. 6.9.4 Thawing Cells for Injection
  • Frozen containers of cells are transferred to the administration site in a dry nitrogen shipper. Prior to administration, containers are placed into a sealed plastic bag and immersed in a 37 0 C water bath. Containers are gently swirled until all of the contents are melted except for a small piece of ice. Containers are removed from the sealed plastic bag and an equal volume of 2.5% HS A/5% Dextran is added. Cells are injected with no further washing.
  • a maternal blood sample accompanies all donor placentas.
  • the sample is screened for Hepatitis B core antibody and surface antigen, Hepatitis C Virus antibody and nucleic acid, and HIV I and II antibody and nucleic acid.
  • Placental processing and primary culture begins prior to the receipt of test results, but continues only for placentas associated with maternal blood samples testing negative for all viruses. A lot is rejected if the donor tests positive for any pathogen.
  • the tests described in Table 3 are performed on the MCB, the WCB, and a sample of the cell dose material derived from a vial of the WCB. A lot is released only when all specifications are met.
  • Table 3 Cell testing and specifications
  • the cell product is below the upper limit of 5EU/kg/dose for recipients over 40kg in body weight.
  • Cells are placed in 1% paraformaldehyde (PFA) in PBS for 20 minutes and stored in a refrigerator until stained (up to a week). Cells are washed with 2% FBS, 0.05% sodium azide in PBS (Staining Buffer) and then resuspended in staining buffer. Cells are stained with the following antibody conjugates: CD105-FITC, CD200-PE, CD34-PECy7, CDlO-APC. Cells are also stained with isotype controls. After 30 minute incubation, the cells are washed and resuspended with Staining Buffer, followed by analysis on a flow cytometer. Cells having an increased fluorescence compared to isotype controls are counted as positive for a marker.
  • PFA paraformaldehyde
  • Placental stem cell- or umbilical cord stem cell-specific genes encodes a number of cytoskeleton and cell-to-cell adhesion proteins associated with epithelial cells and an immunoglobulin-like surface protein, CD200, implicated in maternal-fetal immune tolerance.
  • Placental stem cells and umbilical cord stem cells will be referred to collectively hereinafter in this Example as AC/UC stem cells.
  • BM (Cat* PT-2501) and DF (Cat# CC-2511) were purchased from Cambrex.
  • AC and UC originated from passage 0 tissue culture flasks. AC and UC in the flasks were obtained by digestion from a donor placenta designated 2063919. T-75 culture flasks were seeded at 6000 cells/cm 2 and cells were passaged when they became confluent. Population doublings W
  • FIG. 10 shows growth profiles for the four cell lines in the study; circles indicate which cultures were harvested for RNA isolation. In total twelve samples were collected. BM, AC(6), and UC(6) were harvested after three population doublings; these samples were regarded as being in culture for a "short" period of time. A short-term DF sample was not collected. Intermediate length cultures, 11 to 14 doublings, were collected for all cell types. Long-term cultures were collected from all cell lines at about 35 population doublings or just prior to senescence, whichever came first. Senescence occurred before 15 doublings for BM and at 25 doublings for DF. The purchased BM and DF cells were expanded many times prior to gene analysis, and cannot be considered early-stage.
  • BM grown for three doublings are deemed a short-term culture.
  • BM-11 is operationally referred to as an intermediate length culture, but because senescence occurred at 14 doublings, BM-11 is most likely a long-term culture biologically.
  • Microarray analysis identifies patterns of gene expression, and hierarchical clustering
  • HC attempts to find similarities in the context of two dimensions - genes in the first dimension and different conditions (different RNA samples) in the second.
  • the GeneChips used in this experiment contained over 22,000 probe sets (referred to as the "all genes list"), but many of these sets interrogate genes that are not expressed in any condition.
  • genes not expressed or expressed at low levels (raw values below 250) in all samples were eliminated to yield a list of 8,215 genes.
  • Gene expression patterns of the 8215 genes were displayed using the line graph view in GeneSpring (FIG. 11).
  • the x-axis shows the twelve experimental conditions and the y- axis shows the normalized probe set expression values on a log scale.
  • the y-axis covers a 10,000-fold range, and genes that are not expressed or expressed at very low levels are set to a value of 0.01. By default the normalized value is set to 1.
  • Each line represents a single gene (actually a probe set, some genes have multiple probe sets) and runs across all twelve conditions as a single color. Colors depict relative expression levels, as described for the heatmaps, but the coloring pattern is determined by selecting one condition.
  • AC-03 is the selected condition in FIG. 11.
  • Genes up-regulated relative to the normalized value are displayed by the software as red, and those that are down-regulated, are displayed as blue.
  • the obvious upward and downward pointing spikes in AC-03 through UC-11 indicate that many genes are differentially expressed across these conditions.
  • the striking similarity in the color patterns between AC-03 and UC-03 show that many of the same genes are up or down- regulated in these two samples.
  • Horizontal line segments indicate that a gene's expression level is unchanged across a number of conditions. This is most notable by comparing UC-36, UC-38, and UC-38-T. There are no obvious spikes, but there is a subtle trend in that a number of red lines between UC-36 and UC-38-T are below the normalized value of 1.
  • FIG. 12 shows genes differentially over-expressed, by six-fold or more relative to the baseline, in AC-03. The majority of genes up-regulated in AC-03 are also up-regulated in UC-03, and more divergent in BM and DF.
  • ADARBl adenosine deaminase RNA- RNA processing, central nervous system speciflc, Bl (REDl homolog development rat)
  • B4GALT6 UDP-Gal:betaGlcNAc beta 1,4- carbohydrate metabolism, integral to galactosyltransferase, membrane, may function in intercellular polypeptide 6 recognition and/or adhesion
  • the experimental design compared cells cultured for short, medium, and long periods of time in culture.
  • each culture period has a characteristic set of differentially expressed genes.
  • AC-03 and UC-03 two hundred up-regulated genes regress to the mean after eight population doublings.
  • this early stage gene expression pattern resembles the expression profile of AC and UC while in the natural placental environment. In the placenta these cells are not actively dividing, they are metabolizing nutrients, signaling between themselves, and securing their location by remodeling the extracellular surroundings.
  • Gene expression by the intermediate length cultures is defined by rapid cell division and genes differentially expressed at this time are quite different from those differentially expressed during the early phase. Many of the genes up-regulated in AC-1 1 and UC-1 1, along with BM-03 and DF- 14, are involved in chromosome replication and cell division. Based on gene expression, BM-03 appears biologically to be a mid-term culture. In this middle stage cell type-specific gene expression is overshadowed by cellular proliferation. In addition, almost every gene over expressed in the short-term AC or UC cultures is down- regulated in the middle and later stage conditions. 143 genes were up-regulated > five-fold during this highly proliferative phase, constituting approximately 1.7% of the expressed genes.
  • the long-term cultures represent the final or senescent phase. In this phase, cells have exhausted their ability to divide, and, especially for AC and UC, the absolute number of differentially expressed genes is noticeably reduced. This may be the result of cells being fully adapted to their culture environment and a consequently reduced burden to biosynthesize. Surprisingly, late BM and DF cultures do not display this same behavior; a large number of genes are differentially expressed in BM-11 and DF-24 relative to AC and UC and the normalized value of 1. AC and UC are distinguishable from BM and DF most notably in the long-term cultures.
  • the placental stem cell-specific gene list described here is diverse. COL4A1 and COL4A2 are coordinately regulated, and KRT 18 and KRT8 also appear to be co-expressed. Eight of the genes encode proteins involved in cell to cell contact, three of which (DSC3, DSG2, and PKP2) are localized to desmosomes, intercellular contact points anchored to intermediate filament cytoskeleton proteins such as keratin 18 and keratin 8. Tight cell-to- cell contact is characteristic of epithelial and endothelial cells and not typically associated with fibroblasts. Table 3 lists 16 genes, of the 46 total, characteristic to epithelial cells. Placental stem cells are generally described as fibroblast-like small spindle-shaped cells.
  • CD200 This morphology is typically distinct from BM and DF, especially at lower cell densities. Also of note is the expression pattern of CD200, which is present in AC/UC stem cell and absent in all BM and DF samples. Moreover, CD200 has been shown to be associated with immune tolerance in the placenta during fetal development (see, e.g., Clark et ah, Am. J. Reprod. Immunol. 50(3):l 87-195 (2003)).
  • This subset of genes of 46 genes constitutes a set of molecular biomarkers that distinguishes AC/UC stem cells from bone marrow-derived mesenchymal stem cells or fibroblasts.

Abstract

The present invention provides placental stem cells and placental stem cell populations, and methods of culturing, proliferating and expanding the same. The invention also provides methods of differentiating the placental stem cells. The invention further provides methods of using the placental stem cells in assays and for transplanting.

Description

PLACENTAL STEMCELL POPULATIONS
[0001] This application claims benefit of U.S. Provisional Application No. 60/754,968, filed December 29, 2005; and claims benefit of U.S. Provisional Application No. 60/846,641, filed September 22, 2006.
1. FIELD OF THE INVENTION
[0002] The present invention provides isolated placental stem cells, populations of placental stem cells, compositions comprising the stem cells, and methods of obtaining the stem cells.
2. BACKGROUND OF THE INVENTION
[0003] Human stem cells are totipotential or pluripotential precursor cells capable of generating a variety of mature human cell lineages. Evidence exists that demonstrates that stem cells can be employed to repopulate many, if not all, tissues and restore physiologic and anatomic functionality.
[0004] Many different types of mammalian stem cells have been characterized. See, e.g., Caplan et al., U.S. Patent No. 5,486,359 (human mesenchymal stem cells); Boyse et al, U.S. Patent No. 5,004,681 (fetal and neonatal hematopoietic stem and progenitor cells); Boyse et al, U.S. 5,192,553 (same); Beltrami et al., Cell 114(6):763-766 (2003) (cardiac stem cells); Forbes etal., J. Pathol. 197(4):510-518 (2002) (hepatic stem cells). Umbilical cord blood, and total nucleated cells derived from cord blood, have been used in transplants to restore, partially or fully, hematopoietic function in patients who have undergone ablative therapy.
3. SUMMARY OF THE INVENTION
[0005] The present invention provides isolated placental stem cells, populations of placental stem cells, compositions comprising the stem cells, and methods of obtaining the stem cells. [0006] The invention first provides isolated stem cells, and cell populations comprising such stem cells, wherein the stem cells are present in, and isolatable from placental tissue {e.g., amnion, chorion, placental cotyledons, etc.) The placental stem cells exhibit one or more characteristics of a stem cell {e.g., exhibit markers associated with stem cells, replicate at least 10-20 times in culture in an undifferentiated state, differentiate into adult cells representative of the three germ layers, etc.), and can adhere to a tissue culture substrate {e.g., tissue culture plastic such as the surface of a tissue culture dish or multiwell plate). [0007] In one embodiment, the invention provides an isolated placental stem cell that is CQ200+ or HLA-G+. In a specific embodiment, said cell is CD200+ and HLA-G+. In a specific embodiment, said stem cell is CD73+ and CD105+. In another specific embodiment, said stem cell is CD34", CD38" or CD45". In another specific embodiment, said stem cell is CD34", CD38" and CD45~ In another specific embodiment, said stem cell is CD34", CD38", CD45~, CD73+ and CD105+. In another specific embodiment, said stem cell facilitates the formation of one or more embryoid-like bodies from a population of isolated placental cells comprising placental stem cells when said population is cultured under conditions that allow formation of embryoid-like bodies.
[0008] In another embodiment, the invention provides a population of isolated placental cells comprising, e.g., that is enriched for, CD200+, HLA-G+ stem cells. In various embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% or more of said isolated placental cells are CD200+, HLA-G+ stem cells. In a specific embodiment of the above populations, said stem cells are CD73+ and CD105+. In another specific embodiment, said stem cells are CD34~, CD38~ or CD45". In a more specific embodiment, said stem cells are CD34", CD3S", CD45~, CD73+ and CD105+. In other specific embodiments, said population has been expanded, e.g., passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times. In another specific embodiment, said population forms one or more embryoid-like bodies when cultured under conditions that allow formation of embryoid-like bodies.
[0009J In another embodiment, the invention provides an isolated stem cell that is CD73+, CD105+, and CD200+. In a specific embodiment, said stem cell is HLA-G+. In another specific embodiment, said stem cell is CD34", CD38~ or CD45~. In another specific embodiment, said stem cell is CD34~, CD38" and CD45". In a more specific embodiment, said stem cell is CD34~, CD38~, CD45~, and HLA-G+. In another specific embodiment, said stem cell facilitates development of one or more embryoid-like bodies from a population of isolated placental cells comprising the stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies.
[0010] In another embodiment, the invention provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73+, CD105+, CD200+ stem cells. In various embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD73+, CDl 05+, CD200+ stem cells. In a specific embodiment of said populations, said stem cells are HLA-G+. In another specific embodiment, said stem cells are CD34 , CD38" or CD45". In another specific embodiment, said stem cells are CD34~, CD38" and CD45". In a more specific embodiment, said stem cells are CD34", CD38", CD45~, and HLA-G+. In other specific embodiments, said population has been expanded, for example, passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times. In another specific embodiment, said population forms one or more embryoid-like bodies in culture under conditions that allow formation of embryoid-like bodies.
[0011] The invention also provides an isolated stem cell that is CD200+ and OCT-4+. In a specific embodiment, the stem cell is CD73+ and CD105+. In another specific embodiment, said stem cell is HLA-G+. In another specific embodiment, said stem cell is CD34~, CD38~ or CD45". In another specific embodiment, said stem cell is CD34~, CD38" and CD45~. In a more specific embodiment, said stem cell is CD34", CD38", CD45", CD73+, CDl 05+ and HLA-G+. In another specific embodiment, said stem cell facilitates the formation of one or more embryoid-like bodies from a population of isolated placental cells comprising placental stem cells when said population is cultured under conditions that allow formation of embryoid-like bodies.
[0012] In another embodiment, the invention provides a population of isolated cells comprising, e.g., that is enriched for, CD200+, OCT-4+ stem cells. In various embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD200+, OCT- 4+ stem cells. In a specific embodiment of the above populations, said stem cells are CD73+ and CD105+. In another specific embodiment, said stem cells are HLA-G+. In another specific embodiment, said stem cells are CD34", CD38~ and CD45~\ In a more specific embodiment, said stem cells are CD34", CD38" CD45", CD73+, CD105+ and HLA-G+. In other specific embodiments, said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times. In another specific embodiment, said population forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
[0013] In another embodiment, the invention provides an isolated stem cell that is CD73+ and CDl 05+ and which facilitates the formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies. In a specific embodiment, said stem cell is CD34 , CD38 or CD45 . In another specific embodiment, said stem cell is CD34", CD38" and CD45~. In another specific embodiment, said stem cell is OCT4+. In a more specific embodiment, said stem cell is OCT4+, CD34", CD38~ and CD45". [0014] The invention further provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73+, CD105+ stem cells, wherein said population forms one or more embryoid-like bodies under conditions that allow formation of embryoid-like bodies. In various embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD73+, CD105+ stem cells. In a specific embodiment of the above populations, said stem cells are CD34~, CD38~ or CD45~. In another specific embodiment, said stem cells are CD34", CD38~ and CD45~. In another specific embodiment, said stem cells are OCT-4+. In a more specific embodiment, said stem cells are OCT-4+, CD34~, CD38~ and CD45". In other specific embodiments, said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
[0015] The invention further provides an isolated stem cell that is CD73+, CDlOS+ and HLA- G+. In a specific embodiment, said stem cell is CD34~, CD38~ or CD45~. In another specific embodiment, said stem cell is CD34~, CD38~ and CD45~. In another specific embodiment, said stem cell is OCT-4+. In another specific embodiment, said stem cell is CD200+. In a more specific embodiment, said stem cell is CD34", CD38", CD45", OCT-4+ and CD200+. In another specific embodiment, said stem cell facilitates the formation of one or more embryoid-like bodies from a population of isolated placental cells comprising placental stem cells in culture under conditions that allow formation of embryoid-like bodies. [0016] The invention further provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73+, CD105+ and HLA-G+ stem cells. In various embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are CD73+, CD105+ and HLA-G+ stem cells. In a specific embodiment of the above populations, said stem cells are CD34~, CD38~ or CD45". In another specific embodiment, said stem cells are CD34~, CD38~ and CD45". In another specific embodiment, said stem cells are OCT-4+. In another specific embodiment, said stem cells are CD200+. In a more specific embodiment, said stem cells are CD34", CD38", CD45", OCT-4+ and CD200+. In another specific embodiment, said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times. In another specific embodiment, said population forms embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
[0017] The invention further provides an isolated stem cell that is OCT-4+ and which facilitates formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when cultured under conditions that allow formation of embryoid-like bodies. In a specific embodiment, said stem cell is CD73+ and CDl 05+. In another specific embodiment, said stem cell is CD34~~, CD38~, or CD45". In another specific embodiment, said stem cell is CD200+. In a more specific embodiment, said stem cell is CD73+, CDl 05+, CD200+, CD34", CD38", and CD45~.
[0018] The invention also provides a population of isolated cells comprising, e.g., that is enriched for, OCT-4+ placental stem cells, wherein said population forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid- like bodies. In various embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50% at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of said isolated placental cells are OCT4+ placental stem cells. In a specific embodiment of the above populations, said stem cells are CD73+ and CD105+. In another specific embodiment, said stem cells are CD34", CD38~, or CD45~. In another specific embodiment, said stem cells are CD200+. In a more specific embodiment, said stem cells are CD73+, CD105"1", CD200+, CD34"~, CD38~, and CD45". In another specific embodiment, said population has been expanded, for example, passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times.
[0019] The invention further provides an isolated population of the placental stem cells described herein that is produced according to a method comprising perfusing a mammalian placenta that has been drained of cord blood and perfused to remove residual blood; perfusing said placenta with a perfusion solution; and collecting said perfusion solution, wherein said perfusion solution after perfusion comprises a population of placental cells that comprises placental stem cells; and isolating a plurality of said placental stem cells from said population of cells. In a specific embodiment, the perfusion solution is passed through both the umbilical vein and umbilical arteries and collected after it exudes from the placenta. In another specific embodiment, the perfusion solution is passed through the umbilical vein and collected from the umbilical arteries, or passed through the umbilical arteries and collected from the umbilical vein.
[0020] The invention further provides an isolated population of the placental stem cells described herein that is produced according to a method comprising digesting placental tissue with a tissue-disrupting enzyme to obtain a population of placental cells comprising placental stem cells, and isolating a plurality of placental stem cells from the remainder of said placental cells. In specific embodiments, said placental tissue is a whole placenta, an amniotic membrane, chorion, a combination of amnion and chorion, or a combination of any of the foregoing. In other specific embodiment, the tissue-disrupting enzyme is trypsin or collagenase.
[0021] In more specific embodiments, the invention provides any of the isolated stem cells above, wherein said stem cell expresses one or more genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, ILl 8, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PJP2, RTNl5 SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A, and wherein said bone marrow derived stem cell has undergone a number of passages in culture equivalent to the number of passages said placental stem cell has undergone. Sequences corresponding to these genes are found on Affymetrix GENECHIP® arrays. These genes can also be found at GenBank accession nos. NM_001615 (ACTG2), BC065545 (ADARBl), (NM_181847 (AMIGO2), AY358590 (ARTS-I), BC074884 (B4GALT6), BC008396 (BCHE), BC020196 (Cl Iorf9), BC031103 (CD200), NM_001845 (COL4A1), NM_001846 (COL4A2), BC052289 (CPA4), BC094758 (DMD), AF293359 (DSC3), NM_001943 (DSG2), AF338241 (ELOVL2), AY336105 (F2RL1), NM_018215 (FUl 0781), AY416799 (GATA6), BC075798 (GPR126), NM_016235 (GPRC5B), AF340038 (ICAMl), BC000844 (IER3), BC066339 (IGFBP7), BC013142 (ILIA), BT019749 (IL6), BC007461 (ILl 8), (BC072017) KRT18, BC075839 (KRT8), BC060825 (LIPG), BC065240 (LRAP), BCO 10444 (MATN2), BCOl 1908 (MEST), BC068455 (NFE2L3), NM_014840 (NUAKl), AB006755 (PCDH7), NM_014476 (PDLIM3), BC 126199 (PKP-2), BC090862 (RTNl), BC002538 (SERPINB9), BC023312 (ST3GAL6), BC001201 (ST6GALNAC5), BC126160 or BC065328 (SLC12A8), BC025697 (TCF21), BC096235 (TGFB2), BC005046 (VTN), and BC005001 (ZC3H12A) as of December 2006. [0022] In a more specific embodiment, said stem cell expresses ACTG2, ADARBl, AMIG02, ARTS-I, B4GALT6, BCHE, Cl lor©, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJ10781, GAT A6, GPRl 26, GPRC5B, HLA-G, ICAMl , IER3, IGFBP7, ILIA, IL6, IL 18, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl5 PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12AS, TCF21, TGFB2, VTN, and ZC3H12A at a detectably higher level than a bone marrow-derived mesenchymal stem cell.
[0023] In more specific embodiments, the invention also provides any of the populations of isolated stem cells above, wherein said stem cells express one or more genes at a detectably higher level than a population of bone marrow-derived mesenchymal stem cells, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl , AMIG02, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CPA4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, ILl 8, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A, and wherein said population of bone marrow derived stem cells has undergone a number of passages in culture equivalent to the number of passages said placental stem cell has undregone, and wherein said population of bone marrow-derived mesenchymal stem cells has a number of cells equivalent to said population of isolated stem cells. In a more specific embodiment, the population of isolated stem cells expresses ACTG2, ADARBl5 AMIGO2, ARTS-I, B4GALT6, BCHE, Cl lorfP, CD200, COL4A1 , COL4A2, CPA4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G5 ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A at a detectably higher level than said population of isolated bone marrow-derived mesenchymal stem cells.
[0024] In more specific embodiments of methods of selecting cell populations, the invention also provides methods of selecting one of the above-mentioned cell populations, comprising selecting cells that express one or more genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJ 10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILlA5 IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLCl 2 A8, TCF21, TGFB2, VTN, and ZC3H12A, and wherein said bone marrow derived stem cell has undergone a number of passages in culture equivalent to the number of passages said placental stem cell has undergone. In a W
more specific embodiment, said selecting comprises selecting cells that express ACTG2, ADARBl5 AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl5 IER3, IGFBP7, ILIA, IL6, ILl 8, KRTl 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl5 PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLCl 2 A8, TCF21, TGFB2, VTN and ZC3H12A at a detectably higher level than a bone marrow-derived mesenchymal stem cell.
[0025] The invention also provides compositions that comprise one or more of the stem cells of the invention, wherein the stem cell has been isolated from the placenta. Thus, the invention further provides a composition comprising a stem cell, wherein said stem cell is CD200+ and HLA-G+. In a specific embodiment, said stem cell is CD73+ and CD105+. In another specific embodiment, said stem cell is CD34", CD38~ or CD45~. In another specific embodiment, said stem cell is CD34~, CD38~ and CD45". In a more specific embodiment, said stem cell is CD34" CD38~, CD45", CD73+, CD105+, CD200+ and HLA-G+. [0026] In another embodiment, the invention provides a composition comprising a stem cell, wherein said stem cell is CD73+, CDlOS+ and CD200+. In a specific embodiment, said stem cell is HLA-G+. In another specific embodiment, said stem cell is CD34~, CD38~ or CD45". In another specific embodiment, said stem cell is CD34~, CD38~ and CD45~. In another specific embodiment, said stem cell is CD34", CD38~, CD45" and HLA-G+. [0027] In another embodiment, the invention provides a composition comprising a stem cell, wherein said stem cell is CD200+ and OCT-4+. In a specific embodiment, said stem cell is CD73+ and CD105+. In another specific embodiment, said stem cell is HLA-G+. In another specific embodiment, said stem cell is CD34~, CD38~or CD45~. In another specific embodiment, said stem cell is CD34", CD38~ and CD45~. In another specific embodiment, said stem cell is CD34', CD38", CD45", CD73+, CD105+, and HLA-G+. [0028] In another embodiment, the invention provides a composition comprising a stem cell that is CD73+ and CD105+, wherein said stem cell facilitates formation of an embryoid-like body in a population of isolated placental cells comprising said stem cell under conditions that allow the formation of an embryoid-like body. In a specific embodiment, said stem cell is CD34", CD38~ or CD45~. In another specific embodiment, said stem cell is OCT-4+. In another specific embodiment, said stem cell is CD200+. In another specific embodiment, said stem cell is OCT-4+, CD200+, CD34", CD38" and CD45".
[0029] In yet another embodiment, the invention provides a composition comprising a stem cell that is CD73+, CDl 05+ and HLA-G+. In a specific embodiment, said stem cell is CD34~, CD38" or CD45 . In another specific embodiment, said stem cell is OCT-4+. In another specific embodiment, said stem cell is CD200+. In another specific embodiment, said stem cell is OCT-4+, CD200+, CD34", CD38~ and CD45".
[0030] In another embodiment, the invention provides a composition comprising a stem cell that is OCT-4"1", wherein said stem cell facilitates formation of an embryoid-Iike body in a population of isolated placental cells comprising said stem cell under conditions that allow the formation of an embryoid-like body. In a specific embodiment, said stem cell is CD73+ and CD105+. In another specific embodiment, said stem cell is CD34", CD38" and CD45~. In another specific embodiment, said stem cell is CD200 . In another specific embodiment, said stem cell is CD73+, CDl 05+, CD200+, CD34", CD38" and CD45". [0031] In more specific embodiments of the above compositions, said stem cell expresses one or more genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell, wherein said one or more genes are selected from the group consisting of ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1 , COL4A2, CPA4, DMD, DSC3, DSG2, ELO VL2, F2RL1, FLJl 0781, GATA6, GPRl 26, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, ILl 8, KRTI 8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A, and wherein said bone marrow derived stem cell has undergone a number of passages in culture equivalent to the number of passages said placental stem cell has undergone. In a more specific embodiment of the above compositions, said stem cells express ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD5 DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRTI8, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A at a detectably higher level than a population of isolated bone marrow-derived mesenchymal stem cell, wherein said population of stem cells and said population of bone marrow-derived mesenchymal cells have equivalent numbers of cells.
[0032] In another specific embodiment, any of the foregoing compositions comprises a matrix. In a more specific embodiment, said matrix is a three-dimensional scaffold. In another more specific embodiment, said matrix comprises collagen, gelatin, laminin, fibronectin, pectin, ornithine, or vitronectin. In another more specific embodiment, the matrix is an amniotic membrane or an amniotic membrane-derived biomaterial. In another more specific embodiment, said matrix comprises an extracellular membrane protein. In another more specific embodiment, said matrix comprises a synthetic compound. In another more specific embodiment, said matrix comprises a bioactive compound. In another more specific embodiment, said bioactive compound is a growth factor, cytokine, antibody, or organic molecule of less than 5,000 daltons.
[0033] In another embodiment, the invention further provides a composition comprising medium conditioned by any of the foregoing stem cells, or any of the foregoing stem cell populations. In a specific embodiment, any such composition comprises a stem cell that is not derived from a placenta. In a more specific embodiment, said stem cell is an embryonic stem cell. In another more specific embodiment, said stem cell is a mesenchymal stem cell. In another more specific embodiment, said stem cell is a bone marrow-derived stem cell. In another more specific embodiment, said stem cell is a hematopoietic progenitor cell. In another more specific embodiment, said stem cell is a somatic stem cell. In an even more specific embodiment, said somatic stem cell is a neural stem cell, a hepatic stem cell, a pancreatic stem cell, an endothelial stem cell, a cardiac stem cell, or a muscle stem cell. [0034] The invention also provides methods for producing populations of stem cells derived from mammalian placenta. In one embodiment, for example, the invention provides a method of producing a cell population comprising selecting cells that (a) adhere to a substrate, and (b) express CD200 and HLA-G; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, and (b) express CD73, CDl 05, and CD200; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate and (b) express CD200 and OCT-4; and isolating said cells from other cells to form a cell population. In yet another embodiment, the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, (b) express CD73 and CDl 05, and (c) facilitate the formation of one or more embryoid-like bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, and (b) express CD73, CDl 05 and HLA-G; and isolating said cells from other cells to form a cell population. The invention also provides a method of producing a cell population, comprising selecting cells that (a) adhere to a substrate, (b) express OCT-4, and (c) facilitate the formation of one or more embryoid-like bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies; and isolating said cells from other cells to form a cell population. In a specific embodiment of any of the foregoing methods, said substrate comprises fibronectin. In another specific embodiment, the methods comprise selecting cells that express ABC-p. In another specific embodiment, the methods comprise selecting cells exhibiting at least one characteristic specific to a mesenchymal stem cell. In a more specific embodiment, said characteristic specific to a mesenchymal stem cell is expression of CD29, expression of CD44, expression of CD90, or expression of a combination of the foregoing. In another specific embodiment of the methods, said selecting is accomplished using an antibody. In another specific embodiment, said selecting is accomplished using flow cytometry. In another specific embodiment, said selecting is accomplished using magnetic beads. In another specific embodiment, said selecting is accomplished by fluorescence-activated cell sorting. In another specific embodiment of the above methods, said cell population is expanded.
[0035] The invention also provides a method of producing a stem cell line, comprising transforming a stem cell with a DNA sequence that encodes a growth-promoting protein; and exposing said stem cell to conditions that promote production of said growth-promoting protein. In a specific embodiment, said growth-promoting protein is v-myc, N-myc, c-myc, p53, SV40 large T antigen, polyoma large T antigen, EIa adenovirus or human papillomavirus E7 protein. In a more specific embodiment, said DNA sequence is regulatable. In more specific embodiment, said DNA sequence is regulatable by tetracycline. In another specific embodiment, said growth-promoting protein has a regulatable activity. In another specific embodiment, said growth-promoting protein is a temperature-sensitive mutant.
[0036] The invention further provides cryopreserved stem cell populations. For example, the invention provides a population of CD200+, HLA-G+ stem cells, wherein said cells have been cryopreserved, and wherein said population is contained within a container. The invention also provides a population of CD73+, CD105"1", CD200+ stem cells, wherein said stem cells have been cryopreserved, and wherein said population is contained within a container. The invention also provides a population of CD200+, OCT-4+ stem cells, wherein said stem cells have been cryopreserved, and wherein said population is contained within a container. The invention also provides a population of CD73+, CD105+ stem cells, wherein said cells have been cryopreserved, and wherein said population is contained within a container, and wherein said stem cells facilitate the formation of one or more embryo id-like bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies. The invention further provides a population of CD73+, CD105+, HLA-G+ stem cells, wherein said cells have been cryopreserved, and wherein said population is contained within a container. The invention also provides a population of OCT-4+ stem cells, wherein said cells have been cryopreserved, wherein said population is contained within a container, and wherein said stem cells facilitate the formation of one or more embryoid-iike bodies when cultured with a population of placental cells under conditions that allow for the formation of embryoid-like bodies. In a specific embodiment of any of the foregoing cryopreserved populations, said container is a bag. In various specific embodiments, said population comprises about, at least, or at most 1 x 106 said stem cells, 5 x 106 said stem cells, 1 x 107 said stem cells, 5 x 107 said stem cells, 1 x 108 said stem cells, 5 x 108 said stem cells, 1 x 109 said stem cells, 5 x 109 said stem cells, or 1 x 1010 said stem cells. In other specific embodiments of any of the foregoing cryopreserved populations, said stem cells have been passaged about, at least, or no more than 5 times, no more than 10 times, no more than 15 times, or no more than 20 times. In another specific embodiment of any of the foregoing cryopreserved populations, said stem cells have been expanded within said container.
3.1 DEFINITIONS [0037] As used herein, the term "SH2" refers to an antibody that binds an epitope on the marker CD 105. Thus, cells that are referred to as SH2+ are CD105+. [0038] As used herein, the terms "SH3" and SH4" refer to antibodies that bind epitopes present on the marker CD73. Thus, cells that are referred to as SH3+ and/or SH4+ are CD73+. [0039] As used herein, the term "isolated stem cell" means a stem cell that is substantially separated from other, non-stem cells of the tissue, e.g., placenta, from which the stem cell is derived. A stem cell is "isolated" if at least 50%, 60%, 70%, 80%, 90%, 95%, or at least 99% of the non-stem cells with which the stem cell is naturally associated, or stem cells displaying a different marker profile, are removed from the stem cell, e.g., during collection and/or culture of the stem cell.
[0040] As used herein, the term "population of isolated cells" means a population of cells that is substantially separated from other cells of the tissue, e.g., placenta, from which the population of cells is derived. A stem cell is "isolated" if at least 50%, 60%, 70%, 80%, 90%, 95%, or at least 99% of the cells with which the population of cells, or cells from which the population of cells is derived, is naturally associated, i.e., stem cells displaying a different marker profile, are removed from the stem cell, e.g., during collection and/or culture of the stem cell.
[0041] As used herein, the term "placental stem cell" refers to a stem cell or progenitor cell that is derived from a mammalian placenta, regardless of morphology, cell surface markers, or the number of passages after a primary culture. The term "placental stem cell" as used herein does not, however, refer to a trophoblast. A cell is considered a "stem cell" if the cell retains at least one attribute of a stem cell, e.g., a marker or gene expression profile associated with one or more types of stem cells; the ability to replicate at least 10-40 times in culture, the ability to differentiate into cells of all three germ layers; the lack of adult (i.e., differentiated) cell characteristics, or the like. The terms "placental stem cell" and "placenta- derived stem cell" may be used interchangeably.
[0042] As used herein, a stem cell is "positive" for a particular marker when that marker is detectable above background. For example, a placental stem cell is positive for, e.g., CD73 because CD73 is detectable on placental stem cells in an amount detectably greater than background (in comparison to, e.g., an isotype control). A cell is also positive for a marker when that marker can be used to distinguish the cell from at least one other cell type, or can be used to select or isolate the cell when present or expressed by the cell. In the context of, e.g., antibody-mediated detection, "positive," as an indication a particular cell surface marker is present, means that the marker is detectable using an antibody, e.g., a fluorescently-labeled antibody, specific for that marker; "positive" also means that a cell bears that marker in a amount that produces a signal, e.g., in a cytometer, that is detectably above background. For example, a cell is "CD200+" where the cell is detectably labeled with an antibody specific to CD200, and the signal from the antibody is detectably higher than a control (e.g., background). Conversely, "negative" in the same context means that the cell surface marker is not detectable using an antibody specific for that marker compared to background. For example, a cell is "CD34~" where the cell is not detectably labeled with an antibody specific to CD34. Unless otherwise noted herein, cluster of differentiation ("CD") markers are detected using antibodies. OCT-4 is determined to be present, and a cell is "OCT-4+" if OCT-4 is detectable using RT-PCR. 4. BRIEF DESCRIPTION OF THE FIGURES
[0043] FIG. 1: Viability of placental stem cells from perfusion (A), amnion (B), chorion (C), amnion-chorion plate (D) or umbilical cord (E). Numbers on X-axis designate placenta from which stem cells were obtained.
[0044] FIG. 2: Percent HLA ABC7CD457CD347CD133+ cells from perfusion (A), amnion
(B), chorion (C), amnion-chorion plate (D) or umbilical cord (E) as determined by
FACSCalibur. Numbers on X-axis designate placenta from which stem cells were obtained.
[0045] FIG. 3: Percent HLA ABC7CD457CD347CD133+ cells from perfusion (A), amnion
(B), chorion (C), amnion-chorion plate (D) or umbilical cord (E), as determined by FACS
Aria. Numbers on X-axis designate placenta from which stem cells were obtained.
[0046] FIG. 4: HLA-G5 CDlO5 CDl 3, CD33, CD38, CD44, CD90, CD 105, CDl 17, CD200 expression in stem cells derived from placental perfusate.
[0047] FIG. 5: HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from amnion.
[0048] FIG. 6: HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from chorion.
[0049] FIG. 7: HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from amnion-chorion plate.
[0050] FIG. 8: HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200 expression in stem cells derived from umbilical cord.
[0051] FIG. 9: Average expression of HLA-G, CDlO, CD 13, CD33, CD38, CD44, CD90,
CD 105, CDl 17, CD200 expression in stem cells derived from perfusion (A), amnion (B), chorion (C), amnion-chorion plate (D) or umbilical cord (E).
[0052] FIG. 10: Culture time courses for amnion/chorion (AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (NHDF) cell lines used in this study. All cultures were grown and propagated using the same seeding and passage densities. Circles indicate which cultures were used for RNA isolation. Late cultures were harvested just prior to senescence. Two UC cultures were harvested at 38 doublings (UC-38) to compare the effect of trypsinization on gene expression. AU other cultures were lysed directly in their culture flasks prior to RNA isolation.
[0053] FIG. 11 : Line plot of relative expression levels of 8215 genes in amnion/chorion
(AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (DF) cells. The number associated with each cell line designation on the X-axis indicates the number of days the cell line was cultured prior to evaluation of gene expression levels. The chart was generated from RNA expression data analyzed by GeneSpring software. AC-03 was used as the selected condition.
[0054] FIG. 12: Subset of the all genes list showing genes over-expressed > 6-fold in AC-03 for amnion/chorion (AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (DF) cells. The number associated with each cell line designation on the X-axis indicates the number of days the cell line was cultured prior to evaluation of gene expression levels. The chart was generated from RNA expression data analyzed by GeneSpring software. AC-03 was used as the selected condition. [0055] FIG. 13: Placental stem cell-specific or umbilical cord stem cell-specific genes found by fold change filtering for amnion/chorion (AC), umbilical cord (UC), bone marrow-derived stem cell (BM-MSC) and human dermal fibroblast (DF) cells. The number associated with each cell line designation on the X-axis indicates the number of days the cell line was cultured prior to evaluation of gene expression levels. The chart was generated from RNA expression data analyzed by GeneSpring software. AC-03 was used as the selected condition.
5. DETAILED DESCRIPTION OF THE INVENTION
5.1 PLACENTAL STEM CELLS AND PLACENTAL STEM CELL POPULATIONS
[0056] Placental stem cells are stem cells, obtainable from a placenta or part thereof, that adhere to a tissue culture substrate and have the capacity to differentiate into non-placental cell types. Placental stem cells can be either fetal or maternal in origin (that is, can have the genotype of either the fetus or mother, respectively). Preferably, the placental stem cells and placental stem cell populations of the invention are fetal in origin. Populations of placental stem cells, or populations of cells comprising placental stem cells, can comprise placental stem cells that are solely fetal or maternal in origin, or can comprise a mixed population of placental stem cells of both fetal and maternal origin. The placental stem cells, and populations of cells comprising the placental stem cells, can be identified and selected by the morphological, marker, and culture characteristic discussed below.
5.1.1 Physical and Morphological Characteristics
[0057] The placental stem cells of the present invention, when cultured in primary cultures or in cell culture, adhere to the tissue culture substrate, e.g., tissue culture container surface (e.g., tissue culture plastic). Placental stem cells in culture assume a generally fibroblastoid, stellate appearance, with a number of cyotplasmic processes extending from the central cell body. The placental stem cells are, however, morphologically differentiable from fibroblasts cultured under the same conditions, as the placental stem cells exhibit a greater number of such processes than do fibroblasts. Morphologically, placental stem cells are also differentiable from hematopoietic stem cells, which generally assume a more rounded, or cobblestone, morphology in culture.
5.1.2 Cell Surface. Molecular and Genetic Markers
[0058] Placental stem cells of the present invention, and populations of placental stem cells, express a plurality of markers that can be used to identify and/or isolate the stem cells, or populations of cells that comprise the stem cells. The placental stem cells, and stem cell populations of the invention (that is, two or more placental stem cells) include stem cells and stem cell-containing cell populations obtained directly from the placenta, or any part thereof (e.g., amnion, chorion, placental cotyledons, and the like). Placental stem cell populations also includes populations of (that is, two or more) placental stem cells in culture, and a population in a container, e.g., a bag. Placental stem cells are not, however, trophoblasts. [0059] The placental stem cells of the invention generally express the markers CD73, CDl 05, CD200, HLA-G, and/or OCT-4, and do not express CD34, CD38, or CD45. Placental stem cells can also express HLA-ABC (MHC-I) and HLA-DR. These markers can be used to identify placental stem cells, and to distinguish placental stem cells from other stem cell types. Because the placental stem cells can express CD73 and CD 105, they can have mesenchymal stem cell-like characteristics. However, because the placental stem cells can express CD200 and HLA-G, a fetal-specific marker, they can be distinguished from mesenchymal stem cells, e.g., bone marrow-derived mesenchymal stem cells, which express neither CD200 nor HLA-G. In the same manner, the lack of expression of CD34, CD38 and/or CD45 identifies the placental stem cells as non-hematopoietic stem cells. [0060] Thus, in one embodiment, the invention provides an isolated stem cell that is CD200+ or HLA-G+. In a specific embodiment, said stem cell is a placental stem cell. In a specific embodiment, the stem cell is CD200+ and HLA-G+. In a specific embodiment, said stem cell is CD73+ and CD105+. In another specific embodiment, said stem cell is CD34~, CD38~ or CD45~. In another specific embodiment, said stem cell is CD34", CD38~ and CD45". In another specific embodiment, said stem cell is CD34", CD38~, CD45", CD73+ and CDlOS+. In another specific embodiment, said CD200+ or HLA-G+ stem cell facilitates the formation of ernbryoid-like bodies in a population of placental cells comprising the stem cells, under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said placental stem cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these markers.
[0061] In another embodiment, the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD200+ or HLA-G+ placental cell, whereby said cell is a placental stem cell. In a specific embodiment, said selecting comprises selecting a placental cell that is both CD200+ and HLA-G+. In a specific embodiment, said selecting comprises selecting a placental cell that is also CD73+ and CD105+. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34", CD38~ or CD45". In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34~, CD38~ and CD45~. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34~, CD38~, CD45~, CD73+ and CD105+. In another specific embodiment, said selecting comprises selecting a placental cell that also facilitates the formation of embryoid-like bodies in a population of placental cells comprising the stem cells, under conditions that allow the formation of embryoid-like bodies.
[0062] In another embodiment, the invention provides an isolated population of cells comprising, e.g., that is enriched for, CD200+, HLA-G+ stem cells. In a specific embodiment, said population is a population of placental cells. In various embodiments, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at least about 60% of said cells are CD200+, HLA-G+ stem cells. Preferably, at least about 70% of said cells are CD200+, HLA-G+ stem cells. More preferably, at least about 90%, 95%, or 99% of said cells are CD200+, HLA-G+ stem cells. In a specific embodiment of the isolated populations, said stem cells are also CD73+ and CDl 05+. In another specific embodiment, said stem cells are also CD34~, CD38~ or CD45~. In a more specific embodiment, said stem cells are also CD34", CD38~, CD45" CD73+ and CDl 05+. In another embodiment, said isolated population produces one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these markers. W
[0063] In another embodiment, the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said cells are CD200+, HLA-G+ stem cells. In a specific embodiment, said selecting comprises selecting stem cells that are also CD73+ and CD105+. In another specific embodiment, said selecting comprises selecting stem cells that are also CD34", CD38~ or CD45". In another specific embodiment, said selecting comprises selecting stem cells that are also CD34", CD38", CD45", CD73+ and CD105+. In another specific embodiment, said selecting also comprises selecting a population of placental stem cells that forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies.
[0064] In another embodiment, the invention provides an isolated stem cell that is CD73+, CD105+, and CD200+. In an specific embodiment, said isolated stem cell is an isolated placental stem cell. In another specific embodiment, said stem cell is HLA-G+. In another specific embodiment, said stem cell is CD34", CD38~ or CD45". In another specific embodiment, said stem cell is CD34", CD38" and CD45~. In a more specific embodiment, said stem cell is CD34", CD38", CD45", and HLA-G+. In another specific embodiment, the isolated CD73+, CDl 05+, and CD200+ stem cell facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said placental stern cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these markers.
[0065] In another embodiment, the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD73+, CD105+, and CD200+ placental cell, whereby said cell is a placental stem cell. In a specific embodiment, said selecting comprises selecting a placental cell that is also HLA-G+. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34", CD38" or CD45~. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34", CD38~ and CD45" In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34", CD38", CD45", and HLA-G+. In another specific embodiment, said selecting additionally comprises selecting a CD73+, CD105+, and CD200+ stem cell that facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell, when the population is cultured under conditions that facilitate formation of embryoid-like bodies. [0066] In another embodiment, the invention provides an isolated population of cells comprising, e.g., that is enriched for, CD73+, CDl 05+, CD200+ stem cells. In a specific embodiment, said stem cells are placental stem cells. In various embodiments, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at " least about 60% of said cells are CD73+, CDlOS+, CD200* stem cells. In another embodiment, at least about 70% of said cells in said population of cells are CD73+, CD105+, CD200+ stem cells. In another embodiment, at least about 90%, 95% or 99% of said cells in said population of cells are CD73+, CD105"1", CD200+ stem cells. In a specific embodiment of said populations, said stem cells are HLA-G+. In another specific embodiment, said stem cells are CD34", CD38" or CD45". In another specific embodiment, said stem cells are CD34", CD38" and CD45~. In a more specific embodiment, said stem cells are CD34~, CD38~, CD45~, and HLA-G+. In another specific embodiment, said population of cells produces one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
[0067] In another embodiment, the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said cells are CD73+, CD105+, CD200+ stem cells. In a specific embodiment, said selecting comprises selecting stem cells that are also HLA-G+. In another specific embodiment, said selecting comprises selecting stem cells that are also CD34", CD38~ or CD45". In another specific embodiment, said selecting comprises selecting stem cells that are also CD34~, CD38" and CD45". In another specific embodiment, said selecting comprises selecting stem cells that are also CD34", CD38", CD45", and HLA-G+. In another specific embodiment, said selecting additionally comprises selecting a population of placental cells that produces one or more embryoid-like bodies when the population is cultured under conditions that allow the formation of embryoid-like bodies. [0068] The invention also provides an isolated stem cell that is CD200+ and OCT-4+. In a specific embodiment, the stem cell is CD73+ and CD105+. In a specific embodiment, the stem cell is a placental stem cell. In another specific embodiment, said stem cell is HLA-G+. In another specific embodiment, said stem cell is CD34", CD38~ or CD45". In another specific embodiment, said stem cell is CD34", CD38~ and CD45~. In a more specific embodiment, said stem cell is CD34~, CD38~, CD45~ CD73+, CDl 05+ and HLA-G+. In another specific embodiment, the stem cell facilitates the production of one or more embryoid-like bodies by a population of placental cells that comprises the stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said placental stem cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these markers.
[0069] In another embodiment, the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD200+ and OCT-4+ placental cell, whereby said cell is a placental stem cell. In a specific embodiment, said selecting comprises selecting a placental cell that is also HLA-G+. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34~, CD38" or CD45". In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34", CD38" and CD45". In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34" CD38", CD45", CD73+, CD105+ and HLA-G+. In another specific embodiment, said selecting comprises selecting a placental stem cell that also facilitates the production of one or more embryoid-like bodies by a population of placental cells that comprises the stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies.
[0070] The invention also provides an isolated population of cells comprising, e.g., that is enriched for, CD200"1*, OCT-4+ stem cells. In various embodiments, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at least about 60% of said cells are CD200+, OCT-4+ stem cells. In another embodiment, at least about 70% of said cells are said CD200+, OCT-4+ stem cells. In another embodiment, at least about 90%, 95%, or 99% of said cells are said CD200+, OCT-4+ stem cells. In a specific embodiment of the isolated populations, said stem cells are CD73+ and CD105+. In another specific embodiment, said stem cells are HLA-G+. In another specific embodiment, said stem cells are CD34~, CD38~ and CD45". In a more specific embodiment, said stem cells are CD34~ CD38", CD45" CD73+, CD105+ and HLA-G+. In another specific embodiment, the population produces one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
[0071] In another embodiment, the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said cells are CD200+, OCT-4+ stem cells. In a specific embodiment, said selecting comprises selecting stem cells that are also CD73+ and CDlOS+. In another specific embodiment, said selecting comprises selecting stem cells that are also HLA-G+. In another specific embodiment, said selecting comprises selecting stem cells that are also CD34", CD38" and CD45". In another specific embodiment, said stem cells are also CD34~, CD38", CD45", CD73+, CDlOS+ and HLA-G+.
[0072] The invention further provides an isolated stem cell that is CD73+, CD105+ and HLA- G+. In a specific embodiment, the stem cell is a placental stem cell. In another specific embodiment, said stem cell is CD34~, CD38~ or CD45~. In another specific embodiment, said stem cell is CD34", CD38~ and CD45~". In another specific embodiment, said stem cell is OCT-4+. In another specific embodiment, said stem cell is CD200+. In a more specific embodiment, said stem cell is CD34", CD38~, CD45", OCT-4+ and CD200+. In another specific embodiment, said stem cell facilitates the formation of embryoid-like bodies in a population of placental ceils comprising said stem cell, when the population is cultured under conditions that allow the formation of embryoid-like bodies. In another specific embodiment, said placental stem cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these characteristics.
[0073] In another embodiment, the invention also provides a method of selecting a placental stem cell from a plurality of placental cells, comprising selecting a CD73+, CDl 05+ and HLA-G+ placental cell, whereby said cell is a placental stem cell. In a specific embodiment, said selecting comprises selecting a placental cell that is also CD34~, CD38~ or CD45". In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34", CD38" and CD45". In another specific embodiment, said selecting comprises selecting a placental cell that is also OCT-4+. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD200+. In another specific embodiment, said selecting comprises selecting a placental cell that is also CD34~, CD38", CD45", OCT-4+ and CD200+. In another specific embodiment, said selecting comprises selecting a placental cell that also facilitates the formation of one or more embryoid-like bodies in a population of placental cells that comprises said stem cell, when said population is culture under conditions that allow the formation of embryoid-like bodies.
[0074] The invention also provides an isolated population of cells comprising, e.g., that is enriched for, CD73+, CD105+ and HLA-G+ stem cells.' In a specific embodiment, said stem cells are placental stem cells. In various embodiments, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, or at least about 60% of said cells are CD73+, CD105+ and HLA-G+ stem cells. In another embodiment, at least about 70% of said cells are CD73+, CD105+ and HLA-G+. In another embodiment, at least about 90%, 95% or 99% of said cells are CD73+, CDl 05+ and HLA-G+ stem cells. In a specific embodiment of the above populations, said stem cells are CD34", CD38~ or CD45". In another specific embodiment, said stem cells are CD34", CD38~ and CD45~. In another specific embodiment, said stem cells are OCT-4+. In another specific embodiment, said stem cells are CD200+. In a more specific embodiment, said stem cells are CD34~, CD38", CD45", OCT-4+ and CD200+. In another specific embodiment, said population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
[0075] In another embodiment, the invention also provides a method of selecting a placental stem cell population from a plurality of placental cells, comprising selecting a population of placental cells wherein a majority of said cells are CD73+, CD105+ and HLA-G+. In a specific embodiment, said majority of cells are also CD34~, CD38~ and/or CD45". In another specific embodiment, said majority of cells are also CD200+. In another specific embodiment, said majority of cells are also CD34", CD38~, CD45", OCT-4+ and CD200+. [0076] In another embodiment, the invention provides an isolated stem cell that is CD73+ and CD105+ and which facilitates the formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies. In a specific embodiment, said stem cell is CD34~, CD38" or CD45~. In another specific embodiment, said stem cell is CD34~, CD38~ and CD45~. In another specific embodiment, said stem cell is OCT4+. In a more specific embodiment, said stem cell is OCT4+, CD34", CD38" and CD45". In another specific embodiment, said placental stem cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these characteristics. [00771 The invention further provides a population of isolated placental cells comprising, e.g., that is enriched for, CD73+, CDl 05+ stem cells, wherein said population forms one or more embryoid-like bodies under conditions that allow formation of embryoid-like bodies. In various embodiments, at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said isolated placental cells are CD73+, CD105+ stem cells. In a specific embodiment of the above populations, said stem cells are CD34~, CD38~ or CD45". In another specific embodiment, said stem cells are CD34~, CD38~ and CD45". In another specific embodiment, said stem cells are OCT-4+. In a more specific embodiment, said stem cells are OCT-4+, CD34", CD38" and CD45". In other specific embodiments, said population has been expanded, for example, has been passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times. In another specific embodiment, said population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
[0078] The invention further provides an isolated stem cell that is OCT-4+ and which facilitates formation of one or more embryoid-like bodies in a population of isolated placental cells comprising said stem cell when cultured under conditions that allow formation of embryoid-like bodies. In a specific embodiment, said stem cell is CD73+ and CD105+. In another specific embodiment, said stem ceil is CD34~, CD38", or CD45". In another specific embodiment, said stem cell is CD200+. In a more specific embodiment, said stem cell is CD73+, CD105+, CD200+, CD34" CD38", and CD45" In another specific embodiment, said placental stem cell is isolated away from placental cells that are not stem cells. In another specific embodiment, said placental stem cell is isolated away from placental stem cells that do not display these characteristics.
[0079J The invention also provides a population of isolated cells comprising, e.g., that is enriched for, OCT-4+ stem cells, wherein said population forms one or more embryoid-like bodies when cultured under conditions that allow the formation of embryoid-like bodies. In various embodiments, at least 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50% at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of said isolated placental cells are OCT4+ stem cells. In a specific embodiment of the above populations, said stem cells are CD73+ and CDl 05+. In another specific embodiment, said stem cells are CD34", CD38~, or CD45". In another specific embodiment, said stem cells are CD200+. In a more specific embodiment, said stem cells are CD73+, CD105+, CD200"1", CD34", CD38~ and CD45" In another specific embodiment, said population has been expanded, for example, passaged at least once, at least three times, at least five times, at least 10 times, at least 15 times, or at least 20 times. In another specific embodiment, said population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics.
[0080] In another embodiment, the invention also provides an isolated placental stem cell that is CDlO+, CD34", CDl 05+, and CD200+. The invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO+, CD34", CD105+, CD200+. In a specific embodiment of the above embodiments, said stem cells are additionally CD90+ and CD45~. In a specific embodiment, said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics. In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least about 99% of said cells in said isolated population of placental stem cells, are non-maternal in origin. [0081] In another embodiment, the invention provides an isolated placental stem cell that is HLA-A5B5C5 CD45", CDl 33" and CD34". The invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are HLA- A,B,C~, CD45", CD 133" and CD34~. In a specific embodiment, said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said population of placental stem cells is isolated away from placental stem cells that do not display these characteristics. In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least about 99% of said cells in said isolated population of placental stem cells, are non-maternal in origin. In another embodiment, the invention provides a method of obtaining a placental stem cell that is HLA-A5BJC-, CD45 , CD 133" and CD34" comprising isolating said cell from placental perfusate. [0082] In another embodiment, the invention provides an isolated placental stem cell that is CDlO+, CD13+, CD33+, CD45", CDl 17" and CD133" The invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO+, CD13+, CD33+, CD45" CDl IT and CD133". In a specific embodiment, said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least about 99% of said cells in said isolated population of placental stem cells, are non-maternal in origin. In another specific embodiment, said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics. In another embodiment, the invention provides a method of obtaining a placental stem cell that is CDlO+, CD13+, CD33+, CD45", CDI lT and CD133" comprising isolating said cell from placental perfusate.
[0083] In another embodiment, the invention provides an isolated placental stem cell that is CDlO", CD33~, CD44+, CD45", and CDl IT. The invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO", CD33", CD44+, CD45~, and CDl 17". In a specific embodiment, said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least 99% of said cells in said isolated population of placental stem cells, are non-maternal in origin. In another specific embodiment, said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics. In another embodiment, the invention provides a method of obtaining a placental stem cell that is CDlO", CD33", CD44+, CD45", CDl 17" comprising isolating said cell from placental perfusate. [0084] In another embodiment, the invention provides an isolated placental stem cell that is CDlO", CD 13", CD33", CD45"~, and CDl 17". The invention further provides an isolated population of placental stem cells, wherein at least about 70%, at least about 80%, at least about 90%, at least about 95% or at least about 99% of said placental stem cells are CDlO", CD13", CD33", CD45", and CDl 17". In a specific embodiment, said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least 99% of said cells in said isolated population of placental stem cells, are non-maternal in origin. In another specific embodiment, said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics. In another embodiment, the invention provides a method of obtaining a placental stem cell that is CDlO", CD 13", CD33", CD45", and CDl 17+ comprising isolating said cell from placental perfusate. [0085] In another embodiment, the invention provides an isolated placental stem cell that is HLA A,B,C~, CD45", CD34", CD133", positive for CDlO, CD13, CD38, CD44, CD90, CD 105, CD200 and/or HLA-G, and/or negative for CDl 17. The invention further provides an isolated population of placental stem cells, wherein said stem cells are HLA A,B,C, CD45" CD34", CDl 33", and at least about 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or about 99% of the stem cells in the population are positive for CDlO, CD 13, CD38, CD44, CD90, CD 105, CD200 and/or HLA- G, and/or negative for CD 117. In a specific embodiment, said stem cell or population of placental stem cells is isolated away from placental cells that are not stem cells. In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about 90%, at least about 95%, or at least about 99%, of said cells in said isolated population of placental stem cells, are non-maternal in origin. In another specific embodiment, said stem cell or population of placental stem cells is isolated away from placental stem cells that do not display these characteristics. In another embodiment, the invention provides a method of obtaining a placental stem cell that is HLA A5B, C~, CD45", CD34" CD133" and positive for CDlO, CD13, CD38, CD44, CD90, CD105, CD200 and/or HLA-G, and/or negative for CDl 17, comprising isolating said cell from placental perfusate.
[0086] In another embodiment, the invention provides a placental stem cell that is CD200+ and CDlO+, as determined by antibody binding, and CDl 17", as determined by both antibody binding and RT-PCR. In another embodiment, the invention provides a placental stem cell that is CDlO+, CD29" CD54+, CD200+, HLA-G+, HLA class T and β-2-microglobulin". In another embodiment, the invention provides placental stem cells, wherein the expression of at least one marker is at least two-fold higher than for a mesenchymal stem cell (e.g., a bone marrow-derived mesenchymal stem cell). In another specific embodiment, said isolated placental stem cell is non-maternal in origin. In another specific embodiment, at least about W
90%, at least about 95%, or at least 99%, of said cells in said isolated population of placental stem cells, are non-maternal in origin.
[0087J In another embodiment, the invention provides an isolated population of placental stem cells, wherein a plurality of said placental stem cells are positive for aldehyde dehydrogenase (ALDH), as assessed by an aldehyde dehydrogenase activity assay. Such assays are known in the art (see, e.g., Bostian and Berts, Biochem. J., 173, 787, (1978)). In a specific embodiment, said ALDH assay uses ALDEFLUOR® (Aldagen, Inc., Ashland, Oregon) as a marker of aldehyde dehydrogenase activity. In a specific embodiment, said plurality is between about 3% and about 25% of cells in said population of cells. In another embodiment, the invention provides a population of umbilical cord stem cells, wherein a plurality of said umbilical cord stem cells are positive for aldehyde dehydrogenase, as assessed by an aldehyde dehydrogenase activity assay that uses ALDEFLUOR® as an indicator of aldehyde dehydrogenase activity. In a specific embodiment, said plurality is between about 3% and about 25% of cells in said population of cells. In another embodiment, said population of placental stem cells or umbilical cord stem cells shows at least three-fold, or at least five-fold, higher ALDH activity than a population of bone marrow-derived mesenchymal stem cells having the same number of cells and cultured under the same conditions.
[0088] The invention provides any of the above placental stem cells, or populations of placental stem cells, wherein the stem cell or population of placental stem cells has been passaged at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 times, or more, or expanded for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 population doublings, or more.
[0089] In a specific embodiment of any of the above placental cells or cell populations, the karyotype of the cells, or at least about 95% or about 99% of the cells in said population, is normal. In another specific embodiment of any of the above placental cells or cell populations, the cells, or cells in the population of cells, are non-maternal in origin. [0090] Isolated placental stem cells, or isolated populations of placental stem cells, bearing any of the above combinations of markers, can be combined in any ratio. The invention also provides for the isolation of, or enrichment for, any two or more of the above placental stem ceil populations to form a placental stem cell population. For example, the invention provides an isolated population of placental stem cells comprising a first population of placental stem cells defined by one of the marker combinations described above and a second population of placental stem cells defined by another of the marker combinations described above, wherein said first and second populations are combined in a ratio of about 1 :99, 2:98, 3:97, 4:96, 5:95, 10:90, 20:80, 30:70, 40:60, 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98:2, or about 99:1. In like fashion, any three, four, five or more of the above-described placental stem cells or placental stem cell populations can be combined. [0091] The invention further provides placental stem cells that are obtained by disruption of placental tissue, with or without enzymatic digestion, followed by culture {see Section 5.2.3) or perfusion {see Section 5.2.4). For example, the invention provides an isolated population of placental stem cells that is produced according to a method comprising perfusing a mammalian placenta that has been drained of cord blood and perfused to remove residual blood; perfusing said placenta with a perfusion solution; and collecting said perfusion solution, wherein said perfusion solution after perfusion comprises a population of placental cells that comprises placental stem cells; and isolating a plurality of said placental stem cells from said population of cells. In a specific embodiment, the perfusion solution is passed through both the umbilical vein and umbilical arteries and collected after it exudes from the placenta. Populations of placental stem cells produced by this method typically comprise a mixture of fetal and maternal cells. In another specific embodiment, the perfusion solution is passed through the umbilical vein and collected from the umbilical arteries, or passed through the umbilical arteries and collected from the umbilical vein. Populations of placental stem cells produced by this method typically are substantially exclusively fetal in origin; that is, e.g., greater than 90%, 95%, 99%, or 99.5% of the placental stem cells in the population are fetal in origin.
[0092] In various embodiments, the placental stem cells, contained within a population of cells obtained from perfusion of a placenta, are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% of said population of placental cells. In another specific embodiment, the placental stem cells collected by perfusion comprise fetal and maternal cells. In another specific embodiment, the placental stem cells collected by perfusion are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% fetal cells.
[0093] In another specific embodiment, the invention provides a composition comprising a population of isolated placental stem cells collected by perfusion, wherein said composition comprises at least a portion of the perfusion solution used to collect the placental stem cells. [0094] The invention further provides an isolated population of the placental stem cells described herein that is produced according to a method comprising digesting placental tissue with a tissue-disrupting enzyme to obtain a population of placental cells comprising placental stem cells, and isolating a plurality of placental stem cells from the remainder of said placental cells. The whole, or any part of, the placenta can be digested to obtain placental stem cells. In specific embodiments, for example, said placental tissue is a whole placenta, an amniotic membrane, chorion, a combination of amnion and chorion, or a combination of any of the foregoing. In other specific embodiment, the tissue-disrupting enzyme is trypsin or collagenase. In various embodiments, the placental stem cells, contained within a population of cells obtained from digesting a placenta, are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% of said population of placental cells. [0095] Gene profiling confirms that isolated placental stem cells, and populations of isolated placental stem cells, are distinguishable from other cells, e.g., mesenchymal stem cells, e.g., bone marrow-derived stem cells. The placental stem cells described herein, can be distinguished from mesenchymal stem cells on the basis of the expression of one or more genes, the expression of which is specific to placental stem cells or umbilical cord stem cells in comparison to bone marrow-derived mesenchymal stem cells. In particular, placental stem cells can be distinguished from mesenchymal stem cells on the basis of the expression of one or more gene, the expression of which is significantly higher (that is, at least twofold higher) in placental stem cells than in mesenchymal stem cells, wherein the one or more gene is(are) ACTG2, ADARBl, AMIGO2, ARTS-I, B4GALT6, BCHE, Cl lorf9, CD200, COL4A1, COL4A2, CPA4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, HLA-G, ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PKP2, RTNl, SERPINB9^ ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, ZC3H12A, or a combination of any of the foregoing, wherein the expression of these genes is higher in placental stem cells or umbilical cord stem cells than in bone marrow-derived stem cells, when the stem cells are grown under equivalent conditions. In a specific embodiment, the placental stem cell-specific or umbilical cord stem cell-specific gene is CD200. [0096] The level of expression of these genes can be used to confirm the identity of a population of placental cells, to identify a population of cells as comprising at least a plurality of placental stem cells, or the like. The population of placental stem cells, the identity of which is confirmed, can be clonal, e.g., a population of placental stem cells expanded form a single placental stem cell, or a mixed population of stem cells, e.g., a population of cells comprising solely placental stem cells that are expanded from multiple placental stem cells, or a population of cells comprising placental stem cells and at least one other type of cell. [0097] The level of expression of these genes can be used to select populations of placental stem cells. For example, a population of cells, e.g., clonally-expanded cells, is selected if the expression of one or more of these genes is significantly higher in a sample from the population of cells than in an equivalent population of mesenchymal stem cells. Such selecting can be of a population from a plurality of placental stem cells populations, from a plurality of cell populations, the identity of which is not known, etc. [0098] Placental stem cells can be selected on the basis of the level of expression of one or more such genes as compared to the level of expression in said one or more genes in a mesenchymal stem cell control. In one embodiment, the level of expression of said one or more genes in a sample comprising an equivalent number of mesenchymal stem cells is used as a control. In another embodiment, the control, for placental stem cells tested under certain conditions, is a numeric value representing the level of expression of said one or more genes in mesenchymal stem cells under said conditions.
[0099} The placental stem cells of the invention display the above characteristics (e.g., combinations of cell surface markers and/or gene expression profiles) in primary culture, or during proliferation in medium comprising 60% DMEM-LG (Gibco), 40% MCDB- 201 (Sigma), 2% fetal calf serum (FCS) (Hyclone Laboratories), Ix insulin-transferrin- selenium (ITS), Ix lenolenic-acid-bovine-serum-albumin (LA-BSA), 10"9M dexamethasone (Sigma), 10"4M ascorbic acid 2-phosphate (Sigma), epidermal growth factor (EGF) 10ng/ml (R&D Systems), platelet derived-growth factor (PDGF-BB) lOng/ml (R&D Systems), and IOOU penicillin/100OU streptomycin.
[0100] The isolated populations of placental stem cells described above, and populations of placental stem cells generally, can comprise about, at least, or no more than, 1 x 105, 5 x 10s, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more placental stem cells.
5.1.3 Growth in Culture
[0101] The growth of the placental stem cells described herein , as for any mammalian cell, depends in part upon the particular medium selected for growth. Under optimum conditions, placental stem cells typically double in number in 3-5 days. During culture, the placental stem cells of the invention adhere to a substrate in culture, e.g. the surface of a tissue culture container {e.g., tissue culture dish plastic, fibronectin-coated plastic, and the like) and form a monolayer.
[0102] Populations of isolated placental cells that comprise the placental stem cells of the invention, when cultured under appropriate conditions, form embryoid-like bodies, that is, three-dimensional clusters of cells grow atop the adherent stem cell layer. Cells within the embryoid-like bodies express markers associated with very early stem cells, e.g., OCT-4, Nanog, SSEA3 and SSEA4. Cells within the embryoid-like bodies are typically not adherent to the culture substrate, as are the placental stem cells described herein, but remain attached to the adherent cells during culture. Embryoid-like body cells are dependent upon the adherent placental stem cells for viability, as embryoid-like bodies do not form in the absence of the adherent stem cells. The adherent placental stem cells thus facilitate the growth of one or more embryoid-like bodies in a population of placental cells that comprise the adherent placental stem cells. Without wishing to be bound by theory, the cells of the embryoid-like bodies are thought to grow on the adherent placental stem cells much as embryonic stem cells grow on a feeder layer of cells. Mesenchymal stem cells, e.g., bone marrow-derived mesenchymal stem cells, do not develop embryoid-like bodies in culture.
5.2 METHODS OF OBTAINING PLACENTAL STEM CELLS
5.2.1 Stem Cell Collection Composition
[0103] The present invention further provides methods of collecting and isolating placental stem cells. Generally, stem cells are obtained from a mammalian placenta using a physiologically-acceptable solution, e.g., a stem cell collection composition. A stem cell collection composition is described in detail in related U.S. Provisional Application No. 60/754,969, entitled "Improved Medium for Collecting Placental Stem Cells and Preserving Organs," filed on December 29, 2005.
[0104] The stem cell collection composition can comprise any physiologically-acceptable solution suitable for the collection and/or culture of stem cells, for example, a saline solution (e.g., phosphate-buffered saline, Kreb's solution, modified Kreb's solution, Eagle's solution, 0.9% NaCl. etc.), a culture medium (e.g., DMEM, H.DMEM, etc.), and the like. [0105] The stem cell collection composition can comprise one or more components that tend to preserve placental stem cells, that is, prevent the placental stem cells from dying, or delay the death of the placental stem cells, reduce the number of placental stem cells in a population of cells that die, or the like, from the time of collection to the time of culturing. Such components can be, e.g., an apoptosis inhibitor (e.g., a caspase inhibitor or JNK inhibitor); a vasodilator (e.g., magnesium sulfate, an antihypertensive drug, atrial natriuretic peptide (ANP), adrenocorticotropin, corticotropin-releasing hormone, sodium nitroprusside, hydralazine, adenosine triphosphate, adenosine, indomethacin or magnesium sulfate, a phosphodiesterase inhibitor, etc.); a necrosis inhibitor (e.g., 2-(lH-Indol-3-yl)-3-pentylamino- maleimide, pyrrolidine dithiocarbamate, or clonazepam); a TNF-α inhibitor; and/or an oxygen-carrying perfluorocarbon (e.g., perfluorooctyl bromide, perfluorodecyl bromide, etc.). [0106] The stem cell collection composition can comprise one or more tissue-degrading enzymes, e.g., a metalloprotease, a serine protease, a neutral protease, an RNase, or a DNase, or the like. Such enzymes include, but are not limited to, collagenases (e.g., collagenase I, II, III or IV, a collagenase from Clostridium histolyticum, etc.); dispase, thermolysin, elastase, trypsin, LIBERASE, hyaluronidase, and the like.
[0107] The stem cell collection composition can comprise a bacteriocidally or bacteriostatically effective amount of an antibiotic. In certain non-limiting embodiments, the antibiotic is a macrolide (e.g., tobramycin), a cephalosporin (e.g., cephalexin, cephradine, cefuroxime, cefprozil, cefaclor, cefixime or cefadroxil), a clarithromycin, an erythromycin, a penicillin (e.g., penicillin V) or a quinolone (e.g., ofloxacin, ciprofloxacin or norfloxacin), a tetracycline, a streptomycin, etc. In a particular embodiment, the antibiotic is active against Gram(+) and/or Gram(— ) bacteria, e.g., Pseudomonas aeruginosa, Staphylococcus aureus, and the like.
[0108] The stem cell collection composition can also comprise one or more of the following compounds: adenosine (about 1 mM to about 50 mM); D-glucose (about 20 mM to about 100 mM); magnesium ions (about 1 mM to about 50 mM); a macromolecule of molecular weight greater than 20,000 daltons, in one embodiment, present in an amount sufficient to maintain endothelial integrity and cellular viability (e.g., a synthetic or naturally occurring colloid, a polysaccharide such as dextran or a polyethylene glycol present at about 25 g/1 to about 100 g/1, or about 40 g/1 to about 60 g/1); an antioxidant (e.g., butylated hydroxyanisole, butylated hydroxytoluene, glutathione, vitamin C or vitamin E present at about 25 μM to about 100 μM); a reducing agent (e.g., N-acetylcysteine present at about 0.1 mM to about 5 mM); an agent that prevents calcium entry into cells (e.g., verapamil present at about 2 μM to about 25 μM); nitroglycerin (e.g., about 0.05 g/L to about 0.2 g/L); an anticoagulant, in one embodiment, present in an amount sufficient to help prevent clotting of residual blood (e.g., heparin or hirudin present at a concentration of about 1000 units/1 to about 100,000 units/1); or an amiloride containing compound (e.g., amiloride, ethyl isopropyl amiloride, hexamethylene amiloride, dimethyl amiloride or isobutyl amiloride present at about 1.0 μM to about 5 μM). 5.2.2 Collection and Handling of Placenta
[0109] Generally, a human placenta is recovered shortly after its expulsion after birth. In a preferred embodiment, the placenta is recovered from a patient after informed consent and after a complete medical history of the patient is taken and is associated with the placenta. Preferably, the medical history continues after delivery. Such a medical history can be used to coordinate subsequent use of the placenta or the stem cells harvested therefrom. For example, human placental stem cells can be used, in light of the medical history, for personalized medicine for the infant associated with the placenta, or for parents, siblings or other relatives of the infant.
[0110] Prior to recovery of placental stem cells, the umbilical cord blood and placental blood are removed. In certain embodiments, after delivery, the cord blood in the placenta is recovered. The placenta can be subjected to a conventional cord blood recovery process. Typically a needle or cannula is used, with the aid of gravity, to exsanguinate the placenta (see, e.g., Anderson, U.S. Patent No. 5,372,581; Hessel etal, U.S. Patent No. 5,415,665). The needle or cannula is usually placed in the umbilical vein and the placenta can be gently massaged to aid in draining cord blood from the placenta. Such cord blood recovery may be performed commercially, e.g., LifeBank USA, Cedar Knolls, N. J., ViaCord, Cord Blood Registry and Cryocell. Preferably, the placenta is gravity drained without further manipulation so as to minimize tissue disruption during cord blood recovery. [0111] Typically, a placenta is transported from the delivery or birthing room to another location, e.g., a laboratory, for recovery of cord blood and collection of stem cells by, e.g., perfusion or tissue dissociation. The placenta is preferably transported in a sterile, thermally insulated transport device (maintaining the temperature of the placenta between 20-280C), for example, by placing the placenta, with clamped proximal umbilical cord, in a sterile zip-lock plastic bag, which is then placed in an insulated container. In another embodiment, the placenta is transported in a cord blood collection kit substantially as described in pending United States Patent No. 7,147,626. Preferably, the placenta is delivered to the laboratory four to twenty-four hours following delivery. In certain embodiments, the proximal umbilical cord is clamped, preferably within 4-5 cm (centimeter) of the insertion into the placental disc prior to cord blood recovery. In other embodiments, the proximal umbilical cord is clamped after cord blood recovery but prior to further processing of the placenta. [0112] The placenta, prior to stem cell collection, can be stored under sterile conditions and at either room temperature or at a temperature of 5 to 25°C (centigrade). The placenta may be stored for a period of for a period of four to twenty- four hours, up to forty-eight hours, or longer than forty eight hours, prior to perfusing the placenta to remove any residual cord blood. In one embodiment, the placenta is harvested from between about zero hours to about two hours post-expulsion. The placenta is preferably stored in an anticoagulant solution at a temperature of 5 to 25°C (centigrade). Suitable anticoagulant solutions are well known in the art. For example, a solution of heparin or warfarin sodium can be used. In a preferred embodiment, the anticoagulant solution comprises a solution of heparin (e.g., 1% w/w in 1 : 1000 solution). The exsanguinated placenta is preferably stored for no more than 36 hours before placental stem cells are collected.
[0113] The mammalian placenta or a part thereof, once collected and prepared generally as above, can be treated in any art-known manner, e.g., can be perfused or disrupted, e.g., digested with one or more tissue-disrupting enzymes, to obtain stem cells.
5.2.3 Physical Disruption and Enzymatic Digestion of Placental Tissue [0114] In one embodiment, stem cells are collected from a mammalian placenta by physical disruption of part of all of the organ. For example, the placenta, or a portion thereof, may be, e.g., crushed, sheared, minced, diced, chopped, macerated or the like. The tissue can then be cultured to obtain a population of stem cells. Typically, the placental tissue is disrupted using, e.g., in, a stem cell collection composition (see Section 5.2.1 and below). [0115] The placenta can be dissected into components prior to physical disruption and/or enzymatic digestion and stem cell recovery. Placental stem cells can be obtained from all or a portion of the amniotic membrane, chorion, umbilical cord, placental cotyledons, or any combination thereof, including from a whole placenta. Preferably, placental stem cells are obtained from placental tissue comprising amnion and chorion. Typically, placental stem cells can be obtained by disruption of a small block of placental tissue, e.g., a. block of placental tissue that is about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900 or about 1000 cubic millimeters in volume. Any method of physical disruption can be used, provided that the method of disruption leaves a plurality, more preferably a majority, and more preferably at least 60%, 70%, 80%, 90%, 95%, 98%, or 99% of the cells in said organ viable, as determined by, e.g., trypan blue exclusion.
[0116] Stem cells can generally be collected from a placenta, or portion thereof, at any time within about the first three days post-expulsion, but preferably between about 8 hours and about 18 hours post-expulsion. [0117] In a specific embodiment, the disrupted tissue is cultured in tissue culture medium suitable for the proliferation of placental stem cells {see, e.g., Section 5.3, below, describing the culture of placental stem cells).
[0118] In another specific embodiment, stem cells are collected by physical disruption of placental tissue, wherein the physical disruption includes enzymatic digestion, which can be accomplished by use of one or more tissue-digesting enzymes. The placenta, or a portion thereof, may also be physically disrupted and digested with one or more enzymes, and the resulting material then immersed in, or mixed into, a stem cell collection composition. [0119] A preferred stem cell collection composition comprises one or more tissue-disruptive enzyme(s). Enzymatic digestion preferably uses a combination of enzymes, e.g., a combination of a matrix metalloprotease and a neutral protease, for example, a combination of collagenase and dispase. In one embodiment, enzymatic digestion of placental tissue uses a combination of a matrix metalloprotease, a neutral protease, and a mucolytic enzyme for digestion of hyaluronic acid, such as a combination of collagenase, dispase, and hyaluronidase or a combination of LIBERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) and hyaluronidase. Other enzymes that can be used to disrupt placenta tissue include papain, deoxyribonucleases, serine proteases, such as trypsin, chymotrypsin, or elastase. Serine proteases may be inhibited by alpha 2 microglobulin in serum and therefore the medium used for digestion is usually serum-free. EDTA and DNase are commonly used in enzyme digestion procedures to increase the efficiency of cell recovery. The digestate is preferably diluted so as to avoid trapping stem cells within the viscous digest. [0120] Any combination of tissue digestion enzymes can be used. Typical concentrations for tissue digestion enzymes include, e.g., 50-200 U/mL for collagenase I and collagenase IV, 1- 10 U/mL for dispase, and 10-100 U/mL for elastase. Proteases can be used in combination, that is, two or more proteases in the same digestion reaction, or can be used sequentially in order to liberate placental stem cells. For example, in one embodiment, a placenta, or part thereof, is digested first with an appropriate amount of collagenase I at about 1 to about 2 mg/ml for, e.g., 30 minutes, followed by digestion with trypsin, at a concentration of about 0.25%, for, e.g., 10 minutes, at 37°C. Serine proteases are preferably used consecutively following use of other enzymes.
[0121] In another embodiment, the tissue can further be disrupted by the addition of a chelator, e.g., ethylene glycol bis(2-aminoethyl ether)-N,N,N'N'-tetraacetic acid (EGTA) or ethylenediaminetetraacetic acid (EDTA) to the stem cell collection composition comprising the stem cells, or to a solution in which the tissue is disrupted and/or digested prior to isolation of the stem cells with the stem cell collection composition. [0122] In one embodiment, a digestion can proceed as follows. Approximately a gram of placental tissue is obtained and minced. The tissue is digested in 10 mL of a solution comprising about 1 mg/mL collagenase IA and about 0.25% trypsin at 37°C in a shaker at about 100 RPM. The digestate is washed three times with culture medium, and the washed cells are seeded into 2 T-75 flasks. The. cells are then isolated by differential adherence, and characterized for, e.g., viability, cell surface markers, differentiation, and the like. [0123] It will be appreciated that where an entire placenta, or portion of a placenta comprising both fetal and maternal cells (for example, where the portion of the placenta comprises the chorion or cotyledons), the placental stem cells collected will comprise a mix of placental stem cells derived from both fetal and maternal sources. Where a portion of the placenta that comprises no, or a negligible number of, maternal cells (for example, amnion), the placental stem cells collected will comprise almost exclusively fetal placental stem cells. [0124] Stem cells can be isolated from disrupted tissue by differential trypsinization (see Section 5.2.5, below) followed by culture in one or more new culture containers in fresh proliferation medium, optionally followed by a second differential trypsinization step.
5.2.4 Placental Perfusion
[0125] Placental stem cells can also be obtained by perfusion of the mammalian placenta. Methods of perfusing mammalian placenta to obtain stem cells are disclosed, e.g., in Hariri, U.S. Application Publication No. 2002/0123141, and in related U.S. Provisional Application No. 60/754,969, entitled "Improved Medium for Collecting Placental Stem Cells and Preserving Organs," filed on December 29, 2005.
[0126] Placental stem cells can be collected by perfusion, e.g., through the placental vasculature, using, e.g., a stem cell collection composition as a perfusion solution. In one embodiment, a mammalian placenta is perfused by passage of perfusion solution through either or both of the umbilical artery and umbilical vein. The flow of perfusion solution through the placenta may be accomplished using, e.g., gravity flow into the placenta. Preferably, the perfusion solution is forced through the placenta using a pump, e.g., a peristaltic pump. The umbilical vein can be, e.g., cannulated with a cannula, e.g., a TEFLON® or plastic cannula, that is connected to a sterile connection apparatus, such as sterile tubing. The sterile connection apparatus is connected to a perfusion manifold. [0127] In preparation for perfusion, the placenta is preferably oriented (e.g., suspended) in such a manner that the umbilical artery and umbilical vein are located at the highest point of the placenta. The placenta can be perfused by passage of a perfusion fluid through the placental vasculature and surrounding tissue. The placenta can also be perfused by passage of a perfusion fluid into the umbilical vein and collection from the umbilical arteries, or passage of a perfusion fluid into the umbilical arteries and collection from the umbilical vein. [0128J In one embodiment, for example, the umbilical artery and the umbilical vein are connected simultaneously, e.g., to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution. The perfusion solution is passed into the umbilical vein and artery. The perfusion solution exudes from and/or passes through the walls of the blood vessels into the surrounding tissues of the placenta, and is collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation. The perfusion solution may also be introduced through the umbilical cord opening and allowed to flow or percolate out of openings in the wall of the placenta which interfaced with the maternal uterine wall. Placental cells that are collected by this method, which can be referred to as a "pan" method, are typically a mixture of fetal and maternal cells. [0129] In another embodiment, the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or is passed through the umbilical artery and collected from the umbilical veins. Placental cells collected by this method, which can be referred to as a "closed circuit" method, are typically almost exclusively fetal. [0130] It will be appreciated that perfusion using the pan method, that is, whereby perfusate is collected after it has exuded from the maternal side of the placenta, results in a mix of fetal and maternal cells. As a result, the cells collected by this method comprise a mixed population of placental stem cells of both fetal and maternal origin. In contrast, perfusion solely through the placental vasculature in the closed circuit method, whereby perfusion fluid is passed through one or two placental vessels and is collected solely through the remaining vessel(s), results in the collection of a population of placental stem cells almost exclusively of fetal origin.
[0131] The closed circuit perfusion method can, in one embodiment, be performed as follows. A post-partum placenta is obtained within about 48 hours after birth. The umbilical cord is clamped and cut above the clamp. The umbilical cord can be discarded, or can processed to recover, e.g., umbilical cord stem cells, and/or to process the umbilical cord membrane for the production of a biomaterial. The amniotic membrane can be retained during perfusion, or can be separated from the chorion, e.g., using blunt dissection with the fingers. If the amniotic membrane is separated from the chorion prior to perfusion, it can be, e.g., discarded, or processed, e.g., to obtain stem cells by enzymatic digestion, or to produce, e.g., an amniotic membrane biomaterial, e.g., the biomaterial described in U.S. Application Publication No. 2004/0048796. After cleaning the placenta of all visible blood clots and residual blood, e.g., using sterile gauze, the umbilical cord vessels are exposed, e.g., by partially cutting the umbilical cord membrane to expose a cross-section of the cord. The vessels are identified, and opened, e.g., by advancing a closed alligator clamp through the cut end of each vessel. The apparatus, e.g., plastic tubing connected to a perfusion device or peristaltic pump, is then inserted into each of the placental arteries. The pump can be any pump suitable for the purpose, e.g., a peristaltic pump. Plastic tubing, connected to a sterile collection reservoir, e.g., a blood bag such as a 250 mL collection bag, is then inserted into the placental vein. Alternatively, the tubing connected to the pump is inserted into the placental vein, and tubes to a collection reservoir(s) are inserted into one or both of the placental arteries. The placenta is then perfused with a volume of perfusion solution, e.g., about 750 ml of perfusion solution. Cells in the perfusate are then collected, e.g., by centrifugation.
[0132] In one embodiment, the proximal umbilical cord is clamped during perfusion, and more preferably, is clamped within 4-5 cm (centimeter) of the cord's insertion into the placental disc.
[0133] The first collection of perfusion fluid from a mammalian placenta during the exsanguination process is generally colored with residual red blood cells of the cord blood and/or placental blood. The perfusion fluid becomes more colorless as perfusion proceeds and the residual cord blood cells are washed out of the placenta. Generally from 30 to 100 ml (milliliter) of perfusion fluid is adequate to initially exsanguinate the placenta, but more or less perfusion fluid may be used depending on the observed results. [0134] The volume of perfusion liquid used to collect placental stem cells may vary depending upon the number of stem cells to be collected, the size of the placenta, the number of collections to be made from a single placenta, etc. In various embodiments, the volume of perfusion liquid may be from 50 mL to 5000 mL, 50 mL to 4000 mL, 50 mL to 3000 mL, 100 mL to 2000 mL, 250 mL to 2000 mL, 500 mL to 2000 mL, or 750 mL to 2000 mL. Typically, the placenta is perfused with 700-800 mL of perfusion liquid following exsanguination.
[0135] The placenta can be perfused a plurality of times over the course of several hours or several days. Where the placenta is to be perfused a plurality of times, it may be maintained or cultured under aseptic conditions in a container or other suitable vessel, and perfused with the stem cell collection composition, or a standard perfusion solution (e.g., a normal saline solution such as phosphate buffered saline ("PBS")) with or without an anticoagulant (e.g., heparin, warfarin sodium, coumarin, bishydroxycoumarin), and/or with or without an antimicrobial agent (e.g., β-mercaptoethanol (0.1 mM); antibiotics such as streptomycin (e.g., at 40-100 μg/ml), penicillin (e.g., at 40U/ml), amphotericin B (e.g., at 0.5 μg/ml). In one embodiment, an isolated placenta is maintained or cultured for a period of time without collecting the perfusate, such that the placenta is maintained or cultured for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, or 2 or 3 or more days before perfusion and collection of perfusate. The perfused placenta can be maintained for one or more additional time(s), e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12. 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and perfused a second time with, e.g., 700-800 mL perfusion fluid. The placenta can be perfused 1, 2, 3, 4, 5 or more times, for example, once every 1, 2, 3, 4, 5 or 6 hours. In a preferred embodiment, perfusion of the placenta and collection of perfusion solution, e.g., stem cell collection composition, is repeated until the number of recovered nucleated cells falls below 100 cells/ml. The perfusates at different time points can be further processed individually to recover time-dependent populations of cells, e.g., stem cells. Perfusates from different time points can also be pooled. In a preferred embodiment, stem cells are collected at a time or times between about 8 hours and about 18 hours post-expulsion.
[0136] Without wishing to be bound by any theory, after exsanguination and a sufficient time of perfusion of the placenta, placental stem cells are believed to migrate into the exsanguinated and perfused microcirculation of the placenta where, according to the methods of the invention, they are collected, preferably by washing into a collecting vessel by perfusion. Perfusing the isolated placenta not only serves to remove residual cord blood but also provide the placenta with the appropriate nutrients, including oxygen. The placenta may be cultivated and perfused with a similar solution which was used to remove the residual cord blood cells, preferably, without the addition of anticoagulant agents. [0137] Perfusion according to the methods of the invention results in the collection of significantly more placental stem cells than the number obtainable from a mammalian placenta not perfused with said solution, and not otherwise treated to obtain stem cells (e.g., by tissue disruption, e.g., enzymatic digestion). In this context, "significantly more" means at least 10% more. Perfusion according to the methods of the invention yields significantly more placental stem cells than, e.g., the number of placental stem cells obtainable from culture medium in which a placenta, or portion thereof, has been cultured. [0138] Stem cells can be isolated from placenta by perfusion with a solution comprising one or more proteases or other tissue-disruptive enzymes. In a specific embodiment, a placenta or portion thereof (e. g. , amniotic membrane, amnion and chorion, placental lobule or cotyledon, umbilical cord, or combination of any of the foregoing) is brought to 25-370C, and is incubated with one or more tissue-disruptive enzymes in 200 mL of a culture medium for 30 minutes. Cells from the perfusate are collected, brought to 4°C, and washed with a cold inhibitor mix comprising 5 mM EDTA, 2 mM dithiothreitol and 2 mM beta-mercaptoethanol. The stem cells are washed after several minutes with a cold {e.g., 40C) stem cell collection composition.
5.2.5 Isolation. Sorting, and Characterization of Placental Stem Cells [0139 J Stem cells from mammalian placenta, whether obtained by perfusion or enyzmatic digestion, can initially be purified from {i.e., be isolated from) other cells by Ficoll gradient centrifugation. Such centrifugation can follow any standard protocol for centrifugation speed, etc. In one embodiment, for example, cells collected from the placenta are recovered from perfusate by centrifugation at 5000 x g for 15 minutes at room temperature, which separates cells from, e.g., contaminating debris and platelets. In another embodiment, placental perfusate is concentrated to about 200 ml, gently layered over Ficoll, and centrifuged at about 1100 x g for 20 minutes at 22°C, and the low-density interface layer of cells is collected for further processing.
[0140] Cell pellets can be resuspended in fresh stem cell collection composition, or a medium suitable for stem cell maintenance, e.g., IMDM serum-free medium containing 2U/ml heparin and 2mM EDTA (GibcoBRL, NY). The total mononuclear cell fraction can be isolated, e.g., using Lymphoprep (Nycomed Pharma, Oslo, Norway) according to the manufacturer's recommended procedure.
[0141] As used herein, "isolating" placental stem cells means to remove at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 99% of the cells with which the stem cells are normally associated in the intact mammalian placenta. A stem cell from an organ is "isolated" when it is present in a population of cells that comprises fewer than 50% of the cells with which the stem cell is normally associated in the intact organ. [0142] Placental cells obtained by perfusion or digestion can, for example, be further, or initially, isolated by differential trypsinization using, e.g., a solution of 0.05% trypsin with 0.2% EDTA (Sigma, St. Louis MO). Differential trypsinization is possible because placental stem cells typically detach from plastic surfaces within about five minutes whereas other adherent populations typically require more than 20-30 minutes incubation. The detached placental stem cells can be harvested following trypsinization and trypsin neutralization, using, e.g., Trypsin Neutralizing Solution (TNS, Cambrex). In one embodiment of isolation of adherent cells, aliquots of, for example, about 5-10 x 10 cells are placed in each of several T-75 flasks, preferably fibronectin-coated T75 flasks. In such an embodiment, the cells can be cultured with commercially available Mesenchymal Stem Cell Growth Medium (MSCGM) (Cambrex), and placed in a tissue culture incubator (37°C, 5% CO2). After 10 to 15 days, non-adherent cells are removed from the flasks by washing with PBS. The PBS is then replaced by MSCGM. Flasks are preferably examined daily for the presence of various adherent cell types and in particular, for identification and expansion of clusters of fibroblastoid cells.
[0143] The number and type of cells collected from a mammalian placenta can be monitored, for example, by measuring changes in morphology and cell surface markers using standard cell detection techniques such as flow cytometry, cell sorting, immunocytochemistry (e.g., staining with tissue specific or cell-marker specific antibodies) fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), by examination of the morphology of cells using light or confocal microscopy, and/or by measuring changes in gene expression using techniques well known in the art, such as PCR and gene expression profiling. These techniques can be used, too, to identify cells that are positive for one or more particular markers. For example, using antibodies to CD34, one can determine, using the techniques above, whether a cell comprises a detectable amount of CD34; if so, the cell is CD34+. Likewise, if a cell produces enough OCT-4 RNA to be detectable by RT-PCR, or significantly more OCT-4 RNA than an adult cell, the cell is OCT-4+ Antibodies to cell surface markers (e.g., CD markers such as CD34) and the sequence of stem cell-specific genes, such as OCT-4, are well-known in the art.
[0144] Placental cells, particularly cells that have been isolated by Ficoll separation, differential adherence, or a combination of both, may be sorted using a fluorescence activated cell sorter (FACS). Fluorescence activated cell sorting (FACS) is a well-known method for separating particles, including cells, based on the fluorescent properties of the particles (Kamarch, 1987, Methods Enzymol, 151 :150- 165). Laser excitation of fluorescent moieties in the individual particles results in a small electrical charge allowing electromagnetic separation of positive and negative particles from a mixture. In one embodiment, cell surface marker-specific antibodies or ligands are labeled with distinct fluorescent labels. Cells are processed through the cell sorter, allowing separation of cells based on their ability to bind to the antibodies used. FACS sorted particles may be directly deposited into individual wells of 96-well or 384-well plates to facilitate separation and cloning.
[0145] In one sorting scheme, stem cells from placenta are sorted on the basis of expression of the markers CD34, CD38, CD44, CD45, CD73, CD105, OCT-4 and/or HLA-G. This can be accomplished in connection with procedures to select stem cells on the basis of their adherence properties in culture. For example, an adherence selection stem can be accomplished before or after sorting on the basis of marker expression. In one embodiment, for example, cells are sorted first on the basis of their expression of CD34; CD34" cells are retained, and cells that are CD200+HLA-G+, are separated from all other CD34~ cells. In another embodiment, cells from placenta are based on their expression of markers CD200 and/or HLA-G; for example, cells displaying either of these markers are isolated for further use. Cells that express, e.g., CD200 and/or HLA-G can, in a specific embodiment, be further sorted based on their expression of CD73 and/or CD 105, or epitopes recognized by antibodies SH2, SH3 or SH4, or lack of expression of CD34, CD38 or CD45. For example, in one embodiment, placental cells are sorted by expression, or lack thereof, of CD200, HLA- G, CD73, CDl 05, CD34, CD38 and CD45, and placental cells that are CD200+, HLA-G+, CD73+, CD105+, CD34~, CD38" and CD45' are isolated from other placental cells for further use.
[0146] With respect to antibody-mediated detection and sorting of placental stem cells, any antibody, specific for a particular marker, can be used, in combination with any fluorophore or other label suitable for the detection and sorting of cells (e.g., fluorescence-activated cell sorting). Antibody/fluorophore combinations to specific markers include, but are not limited to, fluorescein isothiocyanate (FITC) conjugated monoclonal antibodies against HLA-G (available from Serotec, Raleigh, North Carolina), CDlO (available from BD Immunocytometry Systems, San Jose, California), CD44 (available from BD Biosciences Pharmingen, San Jose, California), and CD 105 (available from R&D Systems Inc., Minneapolis, Minnesota); phycoerythrin (PE) conjugated monoclonal antibodies against CD44, CD200, CDl 17, and CD 13 (BD Biosciences Pharmingen); phycoerythrin-Cy7 (PE Cy7) conjugated monoclonal antibodies against CD33 and CDlO (BD Biosciences Pharmingen); allophycocyanin (APC) conjugated streptavidin and monoclonal antibodies against CD38 (BD Biosciences Pharmingen); and Biotinylated CD90 (BD Biosciences Pharmingen). Other antibodies that can be used include, but are not limited to, CDl 33-APC (Miltenyi), KDR-Biotin (CD309, Abeam), CytokeratinK-Fitc (Sigma or Dako), HLA ABC- Fitc (BD), HLA DRDQDP-PE (BD), β-2-microglobulin-PE (BD), CD80-PE (BD) and CD86- APC (BD).
[0147] Other antibody/label combinations that can be used include, but are not limited to, CD45-PerCP (peridin chlorophyll protein); CD44-PE; CD 19-PE; CDlO-F (fluorescein); HLA-G-F and 7-amϊno-actinomycin-D (7-AAD); HLA-ABC-F; and the like. [0148] Placental stem cells can be assayed for CDl 17 or CD 133 using, for example, phycoerythrin-Cy5 (PE Cy5) conjugated streptavidin and biotin conjugated monoclonal antibodies against CDl 17 or CD 133; however, using this system, the cells can appear to be positive for CDl 17 or CD133, respectively, because of a relatively high background. [0149] Placental stem cells can be labeled with an antibody to a single marker and detected and/sorted. Placental stem cells can also be simultaneously labeled with multiple antibodies to different markers.
[0150] In another embodiment, magnetic beads can be used to separate cells. The cells may be sorted using a magnetic activated cell sorting (MACS) technique, a method for separating particles based on their ability to bind magnetic beads (0.5-100 μm diameter). A variety of useful modifications can be performed on the magnetic microspheres, including covalent addition of antibody that specifically recognizes a particular cell surface molecule or hapten. The beads are then mixed with the cells to allow binding. Cells are then passed through a magnetic field to separate out cells having the specific cell surface marker. In one embodiment, these cells can then isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers. The cells are again passed through a magnetic field, isolating cells that bound both the antibodies. Such cells can then be diluted into separate dishes, such as microtiter dishes for clonal isolation.
[0151] Placental stem cells can also be characterized and/or sorted based on cell morphology and growth characteristics. For example, placental stem cells can be characterized as having, and/or selected on the basis of, e.g., a fibroblastoid appearance in culture. Placental stem cells can also be characterized as having, and/or be selected, on the basis of their ability to form embryoid-like bodies. In one embodiment, for example, placental cells that are fibroblastoid in shape, express CD73 and CD 105, and produce one or more embryoid-like bodies in culture are isolated from other placental cells. In another embodiment, OCT-4+ placental cells that produce one or more embryoid-like bodies in culture are isolated from other placental cells. [0152] In another embodiment, placental stem cells can be identified and characterized by a colony forming unit assay. Colony forming unit assays are commonly known in the art, such as MESEN CULT™ medium (Stem Cell Technologies, Inc., Vancouver British Columbia) [0153] Placental stem cells can be assessed for viability, proliferation potential, and longevity using standard techniques known in the art, such as trypan blue exclusion assay, fluorescein diacetate uptake assay, propidium iodide uptake assay (to assess viability); and thymidine uptake assay, MTT cell proliferation assay (to assess proliferation). Longevity may be determined by methods well known in the art, such as by determining the maximum number of population doubling in an extended culture.
[0154] Placental stem cells can also be separated from other placental cells using other techniques known in the art, e.g., selective growth of desired ceils (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation); unit gravity separation; countercurrent. distribution; electrophoresis; and the like.
5.3 CULTUREOFPLACENTAL STEMCELLS
5.3.1 Culture Media
[0155] Isolated placental stem cells, or placental stem cell population, or cells or placental tissue from which placental stem cells grow out, can be used to initiate, or seed, cell cultures. Cells are generally transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen (e.g., native or denatured), gelatin, fϊbronectin, ornithine, vitronectin, and extracellular membrane protein (e.g., MATRIGEL® (BD Discovery Labware, Bedford, Mass.)).
[0156] Placental stem cells can be cultured in any medium, and under any conditions, recognized in the art as acceptable for the culture of stem cells. Preferably, the culture medium comprises serum. Placental stem cells can be cultured in, for example, DMEM-LG (Dulbecco's Modified Essential Medium, low glucose)/MCDB 201 (chick fibroblast basal medium) containing ITS (insulin-transferrin-selenium), LA+BSA (linoleic acid-bovine serum albumin), dextrose, L-ascorbic acid, PDGF, EGF, IGF-I, and penicillin/streptomycin; DMEM-HG (high glucose) comprising 10% fetal bovine serum (FBS); DMEM-HG comprising 15% FBS; IMDM (Iscove's modified Dulbecco's medium) comprising 10% FBS, 10% horse serum, and hydrocortisone; Ml 99 comprising 10% FBS, EGF, and heparin; α- MEM (minimal essential medium) comprising 10% FBS, GLUTAMAX™ and gentamicin; DMEM comprising 10% FBS, GLUTAMAX M and gentamicin, etc. A preferred medium is DMEM-LG/MCDB-201 comprising 2% FBS, ITS, LA+BSA, dextrose, L-ascorbic acid, PDGF, EGF, and penicillin/streptomycin.
[0157] Other media in that can be used to culture placental stem cells include DMEM (high or low glucose), Eagle's basal medium, Ham's FlO medium (FlO), Ham's F-12 medium (F 12), Iscove's modified Dulbecco's medium, Mesenchymal Stem Cell Growth Medium (MSCGM)5 Liebovitz's L- 15 medium, MCDB, DMEM/F12, RPMI 1640, advanced DMEM (Gibco), DMEM/MCDB201 (Sigma), and CELL-GRO FREE.
[0158] The culture medium can be supplemented with one or more components including, for example, serum {e.g., fetal bovine serum (FBS), preferably about 2-15% (v/v); equine (horse) serum (ES); human serum (HS)); beta-mercaptoethanol (BME), preferably about 0.001% (v/v); one or more growth factors, for example, platelet-derived growth factor (PDGF), epidermal growth factor (EGF)? basic fibroblast growth factor (bFGF), insulin-like growth factor- 1 (IGF-I), leukemia inhibitory factor (LIF), vascular endothelial growth factor (VEGF), and erythropoietin (EPO); amino acids, including L-valine; and one or more antibiotic and/or antimycotic agents to control microbial contamination, such as, for example, penicillin G, streptomycin sulfate, amphotericin B, gentamicin, and nystatin, either alone or in combination.
[0159] Placental stem cells can be cultured in standard tissue culture conditions, e.g., in tissue culture dishes or multiwell plates. Placental stem cells can also be cultured using a hanging drop method. In this method, placental stem cells are suspended at about 1 x 104 cells per mL in about 5 mL of medium, and one or more drops of the medium are placed on the inside of the lid of a tissue culture container, e.g., a 100 mL Petri dish. The drops can be, e.g., single drops, or multiple drops from, e.g., a multichannel pipetter. The lid is carefully inverted and placed on top of the bottom of the dish, which contains a volume of liquid, e.g., sterile PBS sufficient to maintain the moisture content in the dish atmosphere, and the stem cells are cultured.
[0160] In one embodiment, the placental stem cells are cultured in the presence of a compound that acts to maintain an undifferentiated phenotype in the placental stem cell. In a specific embodiment, the compound is a substituted 3,4-dihydropyridimol[4,5-d]pyrimidϊne. In a more specific embodiment, the compound is a compound having the following chemical structure:
Figure imgf000047_0001
The compound can be contacted with a placental stem cell, or population of placental stem cells, at a concentration of, for example, between about 1 μM to about 10 μM.
5.3.2 Expansion and Proliferation of Placental Stem Cells
[0161] Once an isolated placental stem cell, or isolated population of stem cells (e.g., a stem cell or population of stem cells separated from at least 50% of the placental cells with which the stem cell or population of stem cells is normally associated in vivo), the stem cell or population of stem cells can be proliferated and expanded in vitro. For example, a population of placental stem cells can be cultured in tissue culture containers, e.g., dishes, flasks, multiwell plates, or the like, for a sufficient time for the stem cells to proliferate to 70-90% confluence, that is, until the stem cells and their progeny occupy 70-90% of the culturing surface area of the tissue culture container.
[0162] Placental stem cells can be seeded in culture vessels at a density that allows cell growth. For example, the cells may be seeded at low density (e.g., about 1,000 to about 5,000 cells/cm2) to high density (e.g., about 50,000 or more cells/cm2). In a preferred embodiment, the cells are cultured at about 0 to about 5 percent by volume CO2 in air. In some preferred embodiments, the cells are cultured at about 2 to about 25 percent O2 in air, preferably about 5 to about 20 percent O2 in air. The cells preferably are cultured at about 25°C to about 400C, preferably 37°C. The cells are preferably cultured in an incubator. The culture medium can be static or agitated, for example, using a bioreactor. Placental stem cells preferably are grown under low oxidative stress (e.g., with addition of glutathione, ascorbic acid, catalase, tocopherol, N-acetylcysteine, or the like).
[0163] Once 70%-90% confluence is obtained, the cells may be passaged. For example, the cells can be enzymatically treated, e.g., trypsinized, using techniques well-known in the art, to separate them from the tissue culture surface. After removing the cells by pipetting and counting the cells, about 20,000-100,000 stem cells, preferably about 50,000 stem cells, are passaged to a new culture container containing fresh culture medium. Typically, the new medium is the same type of medium from which the stem cells were removed. The invention encompasses populations of placental stem cells that have been passaged at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, or 20 times, or more.
5.3.3 Placental Stem Cell Populations
[0164] The invention provides populations of placental stem cells. Placental stem cell population can be isolated directly from one or more placentas; that is, the placental stem cell population can be a population of placental cells comprising placental stem cells obtained from, or contained within, perfusate, or obtained from, or contained within, disrupted placental tissue, e.g., placental tissue digestate (that is, the collection of cells obtained by enzymatic digestion of a placenta or part thereof). Isolated placental stem cells of the invention can also be cultured and expanded to produce placental stem cell populations. Populations of placental cells comprising placental stem cells can also be cultured and expanded to produce placental stem cell populations.
[0165] Placental stem cell populations of the invention comprise placental stem cells, for example, placental stem cells as described herein. In various embodiments, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the cells in an isolated placental stem cell population are placental stem cells. That is, a placental stem cell population can comprise, e.g., as much as 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% non-stem cells.
[0166] The invention provides methods of producing isolated placental stem cell population by, e.g., selecting placental stem cells, whether derived from enzymatic digestion or perfusion, that express particular markers and/or particular culture or morphological characteristics. In one embodiment, for example, the invention provides a method of producing a cell population comprising selecting placental cells that (a) adhere to a substrate, and (b) express CD200 and HLA-G; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population comprising identifying placental cells that express CD200 and HLA-G, and isolating said cells from other cells to form a cell population. In another embodiment, the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, and (b) express CD73, CDl 05, and CD200; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population comprising identifying placental cells that express CD73, CD105, and CD200, and isolating said cells from other cells to form a cell population. In another embodiment, the method of producing a ceil population comprises selecting placental cells that (a) adhere to a substrate and (b) express CD200 and OCT-4; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population comprising identifying placental cells that express CD200 and OCT-4, and isolating said cells from other cells to form a cell population. In another embodiment, the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, (b) express CD73 and CDl 05, and (c) facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population comprising identifying placental cells that express CD73 and CD 105, and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body, and isolating said cells from other cells to form a cell population. In another embodiment, the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, and (b) express CD73, CD 105 and HLA-G; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population comprising identifying placental cells that express CD73, CD 105 and HLA-G, and isolating said cells from other cells to form a cell population. In another embodiment, the method of producing a cell population comprises selecting placental cells that (a) adhere to a substrate, (b) express OCT-4, and (c) facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body; and isolating said cells from other cells to form a cell population. In another embodiment, the invention provides a method of producing a cell population comprising identifying placental cells that express OCT-4, and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow for the formation of an embryoid-like body, and isolating said cells from other cells to form a cell population. [0167] Such cell populations can be used to treat any of the diseases or conditions listed hereinbelow. Such cell populations can also be used to assess populations of placental stem cells, e.g., as part of a quality control method.
[0168] In any of the above embodiments, the method can additionally comprise selecting placental cells that express ABC-p (a placenta-specific ABC transporter protein; see, e.g., Allikmets et ah, Cancer Res. 58(23):5337-9 (1998)). The method can also comprise selecting cells exhibiting at least one characteristic specific to, e.g., a mesenchymal stem cell, for example, expression of CD29, expression of CD44, expression of CD90, or expression of a combination of the foregoing.
[0169] In the above embodiments, the substrate can be any surface on which culture and/or selection of Ce1IIs, e.g., placental stem cells, can be accomplished. Typically, the substrate is plastic, e.g., tissue culture dish or multiwell plate plastic. Tissue culture plastic can be coated with a biomolecule, e.g., laminin or fibronectin.
[0170] Cells, e.g., placental stem cells, can be selected for a placental stem cell population by any means known in the art of cell selection. For example, cells can be selected using an antibody or antibodies to one or more cell surface markers, for example, in flow cytometry or FACS. Selection can be accomplished using antibodies in conjunction with magnetic beads. Antibodies that are specific for certain stem cell-related markers are known in the art. For example, antibodies to OCT-4 (Abeam, Cambridge, MA), CD200 (Abeam), HLA-G (Abeam), CD73 (BD Biosciences Pharmingen, San Diego, CA), CDl 05 (Abeam; BioDesign International, Saco, ME), etc. Antibodies to other markers are also available commercially, e.g., CD34, CD38 and CD45 are available from, e.g., StemCell Technologies or BioDesign International.
[0171] The isolated placental stem cell population can comprise placental cells that are not stem cells, or cells that are not placental cells.
[0172] Isolated placental stem cell populations can be combined with one or more populations of non-stem cells or non-placental cells. For example, an isolated population of placental stem cells can be combined with blood (e.g., placental blood or umbilical cord blood), blood-derived stem cells (e.g., stem cells derived from placental blood or umbilical cord blood), umbilical cord stem cells, populations of blood-derived nucleated cells, bone marrow-derived mesenchymal cells, bone-derived stem cell populations, crude bone marrow, adult (somatic) stem cells, populations of stem cells contained within tissue, cultured stem cells, populations of fully-differentiated cells (e.g., chondrocytes, fibroblasts, amniotic cells, osteoblasts, muscle cells, cardiac cells, etc.) and the like. In a specific embodiment, the invention provides a population of stem cells comprising placental stem cells and umbilical cord stem cells. Cells in an isolated placental stem cell population can be combined with a plurality of cells of another type in ratios of about 100,000,000:1, 50,000,000:1, 20,000,000:1, 10,000,000:1, 5,000,000:1, 2,000,000:1, 1,000,000:1, 500,000:1, 200,000:1, 100,000:1, 50,000: 1, 20,000:1, 10,000:1 , 5,000:1, 2,000:1, 1,000:1, 500: 1, 200: 1, 100:1, 50:1, 20:1, 10: 1, 5:1, 2:1, 1:1 ; 1 :2; 1:5; 1 :10; 1 : 100; 1:200; 1 :500; 1 :1,000; 1 :2,000; 1 :5,000; 1 :10,000; 1 :20,000; 1 :50,000; 1:100,000; 1 :500,000; 1: 1,000,000; 1 :2,000,000; 1 :5,000,000; 1 :10,000,000; 1 :20,000,000; 1 :50,000,000; or about 1 :100,000,000, comparing numbers of total nucleated cells in each population. Cells in an isolated placental stem cell population can be combined with a plurality of cells of a plurality of cell types, as well. [0173] In one, an isolated population of placental stem cells is combined with a plurality of hematopoietic stem cells. Such hematopoietic stem cells can be, for example, contained within unprocessed placental, umbilical cord blood or peripheral blood; in total nucleated cells from placental blood, umbilical cord blood or peripheral blood; in an isolated population of CD34+ cells from placental blood, umbilical cord blood or peripheral blood; in unprocessed bone marrow; in total nucleated cells from bone marrow; in an isolated population of CD34+ cells from bone marrow, or the like.
5.4 PRODUCTION OF A PLACENTAL STEM CELL BANK [0174J Stem cells from postpartum placentas can be cultured in a number of different ways to produce a set of lots, e.g., a set of individually-administrable doses, of placental stem cells. Such lots can, for example, be obtained from stem cells from placental perfusate or from enzyme-digested placental tissue. Sets of lots of placental stem cells, obtained from a plurality of placentas, can be arranged in a bank of placental stem cells for, e.g., long-term storage. Generally, adherent stem cells are obtained from an initial culture of placental material to form a seed culture, which is expanded under controlled conditions to form populations of cells from approximately equivalent numbers of doublings. Lots are preferably derived from the tissue of a single placenta, but can be derived from the tissue of a plurality of placentas.
[0175] In one embodiment, stem cell lots are obtained as follows. Placental tissue is first disrupted, e.g., by mincing, digested with a suitable enzyme, e.g., collagenase {see Section 5.2.3, above). The placental tissue preferably comprises, e.g., the entire amnion, entire chorion, or both, from a single placenta, but can comprise only a part of either the amnion or chorion. The digested tissue is cultured, e.g., for about 1-3 weeks, preferably about 2 weeks. After removal of non-adherent cells, high-density colonies that form are collected, e.g., by trypsinization. These cells are collected and resuspended in a convenient volume of culture medium, and defined as Passage 0 cells.
[0176] Passage 0 cells are then used to seed expansion cultures. Expansion cultures can be any arrangement of separate cell culture apparatuses, e.g., a Cell Factory by NUNC™. Cells in the Passage 0 culture can be subdivided to any degree so as to seed expansion cultures with, e.g., 1 x 103, 2 x 103, 3 x 103, 4 x 103, 5 x 103, 6 x lO3, 7 x 103, 8 x 103, 9 x 103, 1 x 104, 1 x IO4, 2 x 104, 3 x 104, 4 x 104, 5 x 104, 6 x 104, 7 x 104, 8 x 104, 9 x 104, or 10 x 104 stem cells. Preferably, from about 2 x 104 to about 3 x 104 Passage O cells are used to seed each expansion culture. The number of expansion cultures can depend upon the number of Passage 0 cells, and may be greater or fewer in number depending upon the particular placenta(s) from which the stem cells are obtained.
[0177} Expansion cultures are grown until the density of cells in culture reaches a certain value, e.g., about 1 x 105 cells/cm2. Cells can either be collected and cryopreserved at this point, or passaged into new expansion cultures as described above. Cells can be passaged, e.g., 2, 3, 4 , 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 times prior to use. A record of the cumulative number of population doublings is preferably maintained during expansion culture(s). The cells from a Passage 0 culture can be expanded for 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 doublings, or up to 60 doublings. Preferably, however, the number of population doublings, prior to dividing the population of cells into individual doses, is between about 15 and about 30, preferably about 20 doublings. The cells can be culture continuously throughout the expansion process, or can be frozen at one or more points during expansion.
[0178} Cells to be used for individual doses can be frozen, e.g., cryopreserved for later use. Individual doses can comprise, e.g., about 1 million to about 100 million cells per ml, and can comprise between about 106 and about 109 cells in total.
[0179} In a specific embodiment, of the method, Passage 0 cells are cultured for a first number of doublings, e.g., approximately 4 doublings, then frozen in a first cell bank. Cells from the first cell bank are frozen and used to seed a second cell bank, the cells of which are expanded for a second number of doublings, e.g., about another eight doublings. Cells at this stage are collected and frozen and used to seed new expansion cultures that are allowed to proceed for a third number of doublings, e.g., about eight additional doublings, bringing the cumulative number of cell doublings to about 20. Cells at the intermediate points in passaging can be frozen in units of about 100,000 to about 10 million cells per ml, preferably about 1 million cells per ml for use in subsequent expansion culture. Cells at about 20 doublings can be frozen in individual doses of between about 1 million to about 100 million cells per ml for administration or use in making a stem cell-containing composition. [0180] In one embodiment, therefore, the invention provides a method of making a placental stem cell bank, comprising: expanding primary culture placental stem cells from a human post-partum placenta for a first plurality of population doublings; cryopreserving said placental stem cells to form a Master Cell Bank; expanding a plurality of placental stem cells from the Master Cell Bank for a second plurality of population doublings; cryopreserving said placental stem cells to form a Working Cell Bank; expanding a plurality of placental stem cells from the Working Cell Bank for a third plurality of population doublings; and cryopreserving said placental stem cells in individual doses, wherein said individual doses collectively compose a placental stem cell bank. In a specific embodiment, the total number of population doublings is about 20. In another specific embodiment, said first plurality of population doublings is about four population doublings; said second plurality of population doublings is about eight population doublings; and said third plurality of population doublings is about eight population doublings. In another specific embodiment, said primary culture placental stem cells comprise placental stem cells from placental perfusate. In another specific embodiment, said primary culture placental stem cells comprise placental stem cells from digested placental tissue. In another specific embodiment, said primary culture placental stem cells comprise placental stem cells from placental perfusate and from digested placental tissue. In another specific embodiment, all of said placental stem cells in said placental stem cell primary culture are from the same placenta. In another specific embodiment, the method further comprises the step of selecting CD200+ or HLA-G+ placental stem cells from said plurality of said placental stem cells from said Working Cell Bank to form individual doses. In another specific embodiment, said individual doses comprise from about 104 to about 105 placental stem cells. In another specific embodiment, said individual doses comprise from about 10s to about 106 placental stem cells. In another specific embodiment, said individual doses comprise from about 106 to about 107 placental stem cells. In another specific embodiment, said individual doses comprise from about 107 to about 10s placental stem cells.
[0181] In a preferred embodiment, the donor from which the placenta is obtained {e.g., the mother) is tested for at least one pathogen. If the mother tests positive for a tested pathogen, the entire lot from the placenta is discarded. Such testing can be performed at any time during production of placental stem cell lots, including before or after establishment of Passage 0 cells, or during expansion culture. Pathogens for which the presence is tested can include, without limitation, hepatitis A, hepatitis B, hepatitis C, hepatitis D, hepatitis E, human immunodeficiency virus (types I and II), cytomegalovirus, herpesvirus, and the like. 5.5 DIFFERENTIATION OF PLACENTAL STEM CELLS
5.5.1 Induction Of Differentiation Into Neuronal or Neurogenic Cells [0182] Neuronal differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into neurons. In an example method, a neurogenic medium comprises DMEM/20% FBS and 1 mM beta- mercaptoethanol; such medium can be replaced after culture for about 24 hours with medium consisting of DMEM and 1-10 mM betamercaptoethanol. In another embodiment, the cells are contacted with DMEM/2% DMSO/200 μM butylated hydroxyanisole. In a specific embodiment, the differentiation medium comprises serum-free DMEMIF- 12, butylated hydroxyanisole, potassium chloride, insulin, forskolin, valproic acid, and hydrocortisone. In another embodiment, neuronal differentiation is accomplished by plating placental stem cells on laminin-coated plates in Neurobasal-A medium (Invitrogen, Carlsbad CA) containing B27 supplement and L-glutamine, optionally supplemented with bFGF and/or EGF. Placental stem cells can also be induced to neural differentiation by co-culture with neural cells, or culture in neuron-conditioned medium.
[0183] Neuronal differentiation can be assessed, e.g., by detection of neuron-like morphology (e.g., bipolar cells comprising extended processes) detection of the expression of e.g., nerve growth factor receptor and neurofilament heavy chain genes by RT-PCR; or detection of electrical activity, e.g., by patch-clamp. A placental stem cell is considered to have differentiated into a neuronal cell when the cell displays one or more of these characteristics.
5.5.2 Induction Of Differentiation Into Adipogenic Cells
[0184] Adipogenic differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into adipocytes. A preferred adipogenic medium comprises MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum. In one embodiment, placental stem cells are fed Adipogenesis Induction Medium (Cambrex) and cultured for 3 days (at 37°C, 5% CO2), followed by 1-3 days of culture in Adipogenesis Maintenance Medium (Cambrex). After 3 complete cycles of induction/maintenance, the cells are cultured for an additional 7 days in adipogenesis maintenance medium, replacing the medium every 2-3 days. 10185] In another embodiment, placental stem cells are cultured in medium comprising 1 μM dexamethasone, 0.2 mM indomethacin, 0.01 mg/ml insulin, 0.5 mM IBMX, DMEM-high glucose, FBS, and antibiotics. Placental stem cells can also be induced towards adipogenesis by culture in medium comprising one or more glucocorticoids {e.g., dexamethasone, indomethasone, hydrocortisone, cortisone), insulin, a compound which elevates intracellular levels of cAMP (e.g., dibutyryl-cAMP; 8-CPT-cAMP (8-(4)chlorophenylthio)-adenosine, 3',5' cyclic monophosphate); 8-bromo-cAMP; dioctanoyl-cAMP; forskolin) and/or a compound which inhibits degradation of cAMP (e.g., a phosphodiesterase inhibitor such as isobutylmethylxanthine (IBMX), methyl isobutylxanthine, theophylline, caffeine, indomethacin).
[0186] A hallmark of adipogenesis is the development of multiple intracytoplasmic lipid vesicles that can be easily observed using the lipophilic stain oil red O. Expression of lipase and/or fatty acid binding protein genes is confirmed by RT/PCR in placental stem cells that have begun to differentiate into adipocytes. A placental stem cell is considered to have differentiated into an adipocytic cell when the cell displays one or more of these characteristics.
5.5.3 Induction Of Differentiation Into Chondrocytic Cells
[0187] Chondrogenic differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into chondrocytes. A preferred chondrocytic medium comprises MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum. In one embodiment, placental stem cells are aliquoted into a sterile polypropylene tube, centrifuged (e.g., at 150 x g for 5 minutes), and washed twice in Incomplete Chondrogenesis Medium (Cambrex). The cells are resuspended in Complete Chondrogenesis Medium (Cambrex) containing 0.01 μg/ml TGF- beta-3 at a concentration of about 1-20 x 10s cells/ml. In other embodiments, placental stem cells are contacted with exogenous growth factors, e.g., GDF-5 or transforming growth factor beta3 (TGF-beta3), with or without ascorbate. Chondrogenic medium can be supplemented with amino acids including proline and glutamine, sodium pyruvate, dexamethasone, ascorbic acid, and insulin/transferrin/selenium. Chondrogenic medium can be supplemented with sodium hydroxide and/or collagen. The placental stem cells may be cultured at high or low density. Cells are preferably cultured in the absence of serum.
[0188] Chondrogenesis can be assessed by e.g., observation of production of esoinophilic ground substance, safranin-O staining for glycosaminoglycan expression; hematoxylin/eosin staining, assessing cell morphology, and/or RT/PCR confirmation of collagen 2 and collagen 9 gene expression. Chondrogenesis can also be observed by growing the stem cells in a pellet, formed, e.g., by gently centrifuging stem cells in suspension (e.g., at about 80Og for about 5 minutes). After about 1-28 days, the pellet of stem cells begins to form a tough matrix and demonstrates a structural integrity not found in non-induced, or non- chondrogenic, cell lines, pellets of which tend to fall apart when challenged. Chondrogenesis can also be demonstrated, e.g., in such cell pellets, by staining with a stain that stains collage, e.g., Sirius Red, and/or a stain that stains glycosaminoglycans (GAGs), such as, e.g., Alcian Blue. A placental stem cell is considered to have differentiated into a chondrocytic cell when the cell displays one or more of these characteristics.
5.5.4 Induction Of Differentiation Into Osteocytic Cells
[0189J Osteogenic differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into osteocytes. A preferred osteocytic medium comprises MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum, followed by Osteogenic Induction Medium (Cambrex) containing 0.1 μM dexamethasone, 0.05 mM ascorbic acid-2-phosphate, 10 mM beta glycerophosphate. In another embodiment, placental stem cells are cultured in medium (e.g., DMEM-low glucose) containing about 10"7 to about 10"9 M dexamethasone, about 10- 50 μM ascorbate phosphate salt (e.g., ascorbate-2-phosphate) and about 10 nM to about 10 mM β-glycerophosphate. Osteogenic medium can also include serum, one or more antibiotic/antimycotic agents, transforming growth factor-beta (e.g., TGF-βl) and/or bone morphogenic protein (e.g., BMP-2, BMP-4, or a combination thereof).
[0190] Differentiation can be assayed using a calcium-specific stain, e.g., von Kossa staining, and RT/PCR detection of, e.g., alkaline phosphatase, osteocalcin, bone sialoprotein and/or osteopontin gene expression. A placental stem cell is considered to have differentiated into an osteocytic cell when the cell displays one or more of these characteristics.
5.5.5 Induction Of Differentiation Into Pancreatic Cells
[0191] Differentiation of placental stem cells into insulin-producing pancreatic cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into pancreatic cells.
[0192] An example pancreagenic medium comprises DMEM/20% CBS, supplemented with basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml. This medium is combined with conditioned media from nestin-positive neuronal cell cultures at 50/50 v/v. KnockOut Serum Replacement can be used in lieu of CBS. Cells are cultured for 14-28 days, refeeding every 3-4 days. [0193] Differentiation can be confirmed by assaying for, e.g., insulin protein production, or insulin gene expression by RTVPCR. A placental stem cell is considered to have differentiated into a pancreatic cell when the cell displays one or more of these characteristics.
5.5.6 Induction Of Differentiation Into Cardiac Cells
[0194] Myogenic (cardiogenic) differentiation of placental stem cells can be accomplished, for example, by placing placental stem cells in cell culture conditions that induce differentiation into cardiomyocytes. A preferred cardiomyocytic medium comprises DMEM/20% CBS supplemented with retinoic acid, 1 μM; basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml; and epidermal growth factor, 100 ng/ml. KnockOut Serum Replacement (Invitrogen, Carlsbad, California) may be used in lieu of CBS. Alternatively, placental stem cells are cultured in DMEM/20% CBS supplemented with 50 ng/ml Cardiotropin-1 for 24 hours. In another embodiment, placental stem cells can be cultured 10-14 days in protein-free medium for 5-7 days, then stimulated with human myocardium extract, e.g., produced by homogenizing human myocardium in 1% HEPES buffer supplemented with 1% cord blood serum.
[0195] Differentiation can be confirmed by demonstration of cardiac actin gene expression, e.g., by RTVPCR, or by visible beating of the cell. A placental stem cell is considered to have differentiated into a cardiac cell when the cell displays one or more of these characteristics.
5.6 PRESERVATION OF PLACENTAL STEM CELLS [0196] Placental stem cells can be preserved, that is, placed under conditions that allow for long-term storage, or conditions that inhibit cell death by, e.g., apoptosis or necrosis. [0197] Placental stem cells can be preserved using, e.g., a composition comprising an apoptosis inhibitor, necrosis inhibitor and/or an oxygen-carrying perfluorocarbon, as described in related U.S. Provisional Application No. 60/754,969, entitled "Improved Medium for Collecting Placental Stem Cells and Preserving Organs," filed on December 25, 2005. In one embodiment, the invention provides a method of preserving a population of stem cells comprising contacting said population of stem cells with a stem cell collection composition comprising an inhibitor of apoptosis and an oxygen-carrying perfluorocarbon, wherein said inhibitor of apoptosis is present in an amount and for a time sufficient to reduce or prevent apoptosis in the population of stem cells, as compared to a population of stem cells not contacted with the inhibitor of apoptosis. In a specific embodiment, said inhibitor of apoptosis is a caspase inhibitor. In another specific embodiment, said inhibitor of apoptosis is a JNK inhibitor. In a more specific embodiment, said JNK inhibitor does not modulate differentiation or proliferation of said stem cells. In another embodiment, said stem cell collection composition comprises said inhibitor of apoptosis and said oxygen-carrying perfluorocarbon in separate phases. In another embodiment, said stem cell collection composition comprises said inhibitor of apoptosis and said oxygen-carrying perfluorocarbon in an emulsion. In another embodiment, the stem cell collection composition additionally comprises an emulsifier, e.g., lecithin. In another embodiment, said apoptosis inhibitor and said perfluorocarbon are between about O0C and about 25°C at the time of contacting the stem cells. In another more specific embodiment, said apoptosis inhibitor and said perfluorocarbon are between about 2°C and 100C, or between about 2°C and about 5°C, at the time of contacting the stem cells. In another more specific embodiment, said contacting is performed during transport of said population of stem cells. In another more specific embodiment, said contacting is performed during freezing and thawing of said population of stem cells.
[0198] In another embodiment, the invention provides a method of preserving a population of placental stem cells comprising contacting said population of stem cells with an inhibitor of apoptosis and an organ-preserving compound, wherein said inhibitor of apoptosis is present in an amount and for a time sufficient to reduce or prevent apoptosis in the population of stem cells, as compared to a population of stem cells not contacted with the inhibitor of apoptosis. In a specific embodiment, the organ-preserving compound is UW solution (described in U.S. Patent No. 4,798,824; also known as ViaSpan; see also Southard et al., Transplantation 49(2):251-257 (1990)) or a solution described in Stern et al, U.S. Patent No. 5,552,267. In another embodiment, said organ-preserving compound is hydroxyethyl starch, lactobionic acid, raffinose, or a combination thereof. In another embodiment, the stem cell collection composition additionally comprises an oxygen-carrying perfluorocarbon, either in two phases or as an emulsion.
[0199] In another embodiment of the method, placental stem cells are contacted with a stem cell collection composition comprising an apoptosis inhibitor and oxygen-carrying perfluorocarbon, organ-preserving compound, or combination thereof, during perfusion. In another embodiment, said stem cells are contacted during a process of tissue disruption, e.g., enzymatic digestion. In another embodiment, placental stem cells are contacted with said stem cell collection compound after collection by perfusion, or after collection by tissue disruption, e.g., enzymatic digestion. [0200] Typically, during placental cell collection, enrichment and isolation, it is preferable to minimize or eliminate cell stress due to hypoxia and mechanical stress. In another embodiment of the method, therefore, a stem cell, or population of stem cells, is exposed to a hypoxic condition during collection, enrichment or isolation for less than six hours during said preservation, wherein a hypoxic condition is a concentration of oxygen that is less than normal blood oxygen concentration. In a more specific embodiment, said population of stem cells is exposed to said hypoxic condition for less than two hours during said preservation. In another more specific embodiment, said population of stem cells is exposed to said hypoxic condition for less than one hour, or less than thirty minutes, or is not exposed to a hypoxic condition, during collection, enrichment or isolation. In another specific embodiment, said population of stem cells is not exposed to shear stress during collection, enrichment or isolation.
[0201 J The placental stem cells of the invention can be cryopreserved, e.g., in cryopreservation medium in small containers, e.g., ampoules. Suitable cryopreservation medium includes, but is not limited to, culture medium including, e.g., growth medium, or cell freezing medium, for example commercially available cell freezing medium, e.g., C2695, C2639 or C6039 (Sigma). Cryopreservation medium preferably comprises DMSO (dimethylsulfoxide), at a concentration of, e.g., about 10% (v/v). Cryopreservation medium may comprise additional agents, for example, methylcellulose and/or glycerol. Placental stem cells are preferably cooled at about l°C/min during cryopreservation. A preferred cryopreservation temperature is about -800C to about -1800C, preferably about -125°C to about -14O0C. Cryopreserved cells can be transferred to liquid nitrogen prior to thawing for use. In some embodiments, for example, once the ampoules have reached about -9O0C, they are transferred to a liquid nitrogen storage area. Cryopreservation can also be done using a controlled-rate freezer. Cryopreserved cells preferably are thawed at a temperature of about 250C to about 400C, preferably to a temperature of about 37°C.
5.7 USES OF PLACENTAL STEM CELLS 5.7.1 Placental Stem Cell Populations
[0202] Placental stem cell populations can be used to treat any disease, disorder or condition that is amenable to treatment by administration of a population of stem cells. As used herein, "treat" encompasses the cure of, remediation of, improvement of, lessening of the severity of, or reduction in the time course of, a disease, disorder or condition, or any parameter or symptom thereof. [0203] Placental stem cells, and populations of placental stem cells, can be induced to differentiate into a particular cell type, either ex vivo or in vivo, in preparation for administration to an individual in need of stem cells, or cells differentiated from stem cells. For example, placental stem cells can be injected into a damaged organ, and for organ neogenesis and repair of injury in vivo. Such injury may be due to such conditions and disorders including, but not limited to, myocardial infarction, seizure disorder, multiple sclerosis, stroke, hypotension, cardiac arrest, ischemia, inflammation, thyroiditis, age-related loss of cognitive function, radiation damage, cerebral palsy, neurodegenerative disease, Alzheimer's disease, Parkinson's disease, Leigh disease, AIDS dementia, memory loss, amyotrophic lateral sclerosis, muscular dystrophy, ischemic renal disease, brain or spinal cord trauma, heart-lung bypass, glaucoma, retinal ischemia, or retinal trauma. [0204] Placental stem cells can be used to treat autoimmune conditions such as juvenile diabetes, lupus, muscular dystrophy, rheumatoid arthritis, and the like. [0205] Isolated populations of placental stem cells can be used, in specific embodiments, in autologous or heterologous enzyme replacement therapy to treat specific diseases or conditions, including, but not limited to lysosomal storage diseases, such as Tay-Sachs, Niemaπn-Pick, Fabry's, Gaucher's disease (e.g., glucocerbrosidase deficiency), Hunter's, and Hurler's syndromes, Maroteaux-Lamy syndrome, fucosidosis (fucosidase deficiency), Batten disease (CLN3), as well as other gangliosidoses, mucopolysaccharidoses, and glycogenoses. [0206] Isolated populations of placental stem cells, alone or in combination with stem or progenitor cell populations, may be used alone, or as autologous or heterologous transgene carriers in gene therapy, to correct inborn errors of metabolism, cystic fibrosis, adrenoleukodystrophy (e.g., co-A ligase deficiency), metachromatic leukodystrophy (arylsulfatase A deficiency) (e.g., symptomatic, or presymptomatic late infantile or juvenile forms), globoid cell leukodystrophy (Krabbe's disease; galactocerebrosidase deficiency), acid lipase deficiency (Wolman disease), glycogen storage disease, hypothyroidism, anemia (e.g., aplastic anemia, sickle cell anemia, etc.), Pearson syndrome, Pompe's disease, phenylketonuria (PKU), porphyrias, maple syrup urine disease, homocystinuria, mucopolysaccharidenosis, chronic granulomatous disease and tyrosinemia and Tay-Sachs disease or to treat cancer (e.g., a hematologic malignancy), tumors or other pathological conditions. The placental stem cells can be used to treat skeletal dysplasia. In one embodiment, placental stem cells transformed to express tissue plasminogen activator (tPA) can be administered to an individual to treat thrombus. [0207] In other embodiments, isolated populations of placental stem cells may be used in autologous or heterologous tissue regeneration or replacement therapies or protocols, including, but not limited to treatment of corneal epithelial defects, treatment of osteogenesis imperfecta, cartilage repair, facial dermabrasion, mucosal membranes, tympanic membranes, intestinal linings, neurological structures (e.g., retina, auditory neurons in basilar membrane, olfactory neurons in olfactory epithelium), burn and wound repair for traumatic injuries of the skin, or for reconstruction of other damaged or diseased organs or tissues. [0208] In a preferred embodiment, an isolated population of placental stem cells is used in hematopoietic reconstitution in an individual that has suffered a partial or total loss of hematopoietic stem cells, e.g., individuals exposed to lethal or sub-lethal doses of radiation (whether industrial, medical or military); individuals that have undergone myeloablation as part of, e.g., cancer therapy, and the like, in the treatment of, e.g., a hematologic malignancy. Placental stem cells can be used in hematopoietic reconstitution in individuals having anemia (e.g., aplastic anemia, sickle cell anemia, etc.). Preferably, the placental stem cells are administered to such individuals with a population of hematopoietic stem cells. Isolated populations of placental-derived stem cells can be used in place of, or to supplement, bone marrow or populations of stem cells derived from bone marrow. Typically, approximately 1 x 108 to 2 x 108 bone marrow mononuclear cells per kilogram of patient weight are infused for engraftment in a bone marrow transplantation (i.e., about 70 ml of marrow for a 70 kg donor). To obtain 70 ml requires an intensive donation and significant loss of donor blood in the donation process. An isolated population of placental stem cells for hematopoietic reconstitution can comprise, in various embodiments, about, at least, or no more than 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 10s, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 10n or more placental stem cells.
[0209] In one embodiment, therefore, placental stem cells can be used to treat patients having a blood cancer, such as a lymphoma, leukemia (such as chronic or acute myelogenous leukemia, acute lymphocytic leukemia, Hodgkin's disease, etc.), myelodysplasia, myelodysplastic syndrome, and the like. In another embodiment, the disease, disorder or condition is chronic granulomatous disease.
[0210] Because hematopoietic reconstitution can be used in the treatment of anemias, the present invention further encompasses the treatment of an individual with a stem cell combination of the invention, wherein the individual has an anemia or disorder of the blood hemoglobin. The anemia or disorder may be natural (e.g., caused by genetics or disease), or may be artificially-induced (e.g., by accidental or deliberate poisoning, chemotherapy, and the like). In another embodiment, the disease or disorder is a marrow failure syndrome {e.g., aplastic anemia, Kostmann syndrome, Diamond-Blackfan anemia, amegakaryocytic thrombocytopenia, and the like), a bone marrow disorder or a hematopoietic disease or disorder.
[0211] Placental stem cells can also be used to treat severe combined immunodeficiency disease, including, but not limited to, combined immunodeficiency disease {e.g., Wiskott- Aldrich syndrome, severe DiGeorge syndrome, and the like).
[0212] The placental stem cells of the invention, alone or in combination with other stem cell or progenitor cell populations, can be used in the manufacture of a tissue or organ in vivo. The methods of the invention encompass using cells obtained from the placenta, e.g., stem cells or progenitor cells, to seed a matrix and to be cultured under the appropriate conditions to allow the cells to differentiate and populate the matrix. The tissues and organs obtained by the methods of the invention can be used for a variety of purposes, including research and therapeutic purposes.
[0213] In a preferred embodiment of the invention, placental stem cells and placental stem cell populations may be used for autologous and allogenic transplants, including matched and mismatched HLA type hematopoietic transplants. In one embodiment of the use of placental stem cells as allogenic hematopoietic transplants, the host is treated to reduce immunological rejection of the donor cells, or to create immunotolerance {see, e.g., U.S. Patent Nos. 5,800,539 and 5,806,529). In another embodiment, the host is not treated to reduce immunological rejection or to create immunotolerance.
[0214] Placental stem cells, either alone or in combination with one or more other stem cell populations, can be used in therapeutic transplantation protocols, e.g., to augment or replace stem or progenitor cells of the liver, pancreas, kidney, lung, nervous system, muscular system, bone, bone marrow, thymus, spleen, mucosal tissue, gonads, or hair. Additionally, placental stem cells may be used instead of specific classes of progenitor cells {e.g., chondrocytes, hepatocytes, hematopoietic cells, pancreatic parenchymal cells, neuroblasts, muscle progenitor cells, etc.) in therapeutic or research protocols in which progenitor cells would typically be used.
[0215] In one embodiment, the invention provides for the use of placental stem cells, particularly CD200+ placental stem cells, as an adjunct to hair replacement therapy. For example, in one embodiment, placental stem cells, e.g., CD200+ placental stem cells, are injected subcutaneously or intradermally at a site in which hair growth or regrowth is desired. The number of stem cells injected can be, e.g., between about 100 and about 10,000 per injection, in a volume of about 0.1 to about 1.0 μL, though more ore fewer cells in a greater or lesser volume can also be used. Administration of placental stem cells to facilitate hair regrowth can comprise a single injection or multiple injections in, e.g., a regular or a random pattern in an area in which hair regrowth is desired. Known hair regrowth therapies can be used in conjunction with the placental stem cells, e.g., topical minoxidil. Hair loss that can be treated using placental stem cells can be naturally-occurring (e.g., male pattern baldness) or induced (e.g., resulting from toxic chemical exposure).
[0216] Placental stem cells and placental stem cell populations of the invention can be used for augmentation, repair or replacement of cartilage, tendon, or ligaments. For example, in certain embodiments, prostheses (e.g., hip prostheses) can be coated with replacement cartilage tissue constructs grown from placental stem cells of the invention. In other embodiments, joints (e.g., knee) can be reconstructed with cartilage tissue constructs grown from placental stem cells. Cartilage tissue constructs can also be employed in major reconstructive surgery for different types of joints (see, e.g., Resnick & Niwayama, eds., 1988, Diagnosis of Bone and Joint Disorders, 2d ed., W. B. Saunders Co.). [0217] The placental stem cells of the invention can be used to repair damage to tissues and organs resulting from, e.g., trauma, metabolic disorders, or disease. The trauma can be, e.g., trauma from surgery, e.g., cosmetic surgery. In such an embodiment, a patient can be administered placental stem cells, alone or combined with other stem or progenitor cell populations, to regenerate or restore tissues or organs which have been damaged as a consequence of disease.
5.7.2 Compositions Comprising Placental Stem Cells
[0218] The present invention provides compositions comprising placental stem cells, or biomolecules therefrom. The placental stem cells of the present invention can be combined with any physiologically-acceptable or medically-acceptable compound, composition or device for use in, e.g., research or therapeutics.
5.7.2.1 Cryopreserved Placental Stem Cells
[0219] The placental stem cell populations of the invention can be preserved, for example, cryopreserved for later use. Methods for cryopreservation of cells, such as stem cells, are well known in the art. Placental stem cell populations can be prepared in a form that is easily administrable to an individual. For example, the invention provides a placental stem cell population that is contained within a container that is suitable for medical use. Such a container can be, for example, a sterile plastic bag, flask, jar, or other container from which the placental stem cell population can be easily dispensed. For example, the container can be a blood bag or other plastic, medically-acceptable bag suitable for the intravenous administration of a liquid to a recipient. The container is preferably one that allows for cryopreservation of the combined stem cell population.
[0220] The cryopreserved placental stem cell population can comprise placental stem cells derived from a single donor, or from multiple donors. The placental stem cell population can be completely HLA-matched to an intended recipient, or partially or completely HLA- mismatched.
[0221] Thus, in one embodiment, the invention provides a composition comprising a placental stem cell population in a container. In a specific embodiment, the stem cell population is cryopreserved. In another specific embodiment, the container is a bag, flask, or jar. In more specific embodiment, said bag is a sterile plastic bag. In a more specific embodiment, said bag is suitable for, allows or facilitates intravenous administration of said placental stem cell population. The bag can comprise multiple lumens or compartments that are interconnected to allow mixing of the placental stem cells and one or more other solutions, e.g., a drug, prior to, or during, administration. In another specific embodiment, the composition comprises one or more compounds that facilitate cryopreservation of the combined stem cell population. In another specific embodiment, said placental stem cell population is contained within a physiologically-acceptable aqueous solution. In a more specific embodiment, said physiologically-acceptable aqueous solution is a 0.9% NaCl solution. In another specific embodiment, said placental stem cell population comprises placental cells that are HLA-matched to a recipient of said stem cell population. In another specific embodiment, said combined stem cell population comprises placental cells that are at least partially HLA-mismatched to a recipient of said stem cell population. In another specific embodiment, said placental stem cells are derived from a plurality of donors.
5.7.2.2 Pharmaceutical Compositions
[0222] Populations of placental stem cells, or populations of cells comprising placental stem cells, can be formulated into pharmaceutical compositions for use in vivo. Such pharmaceutical compositions comprise a population of placental stem cells, or a population of cells comprising placental stem cells, in a pharmaceutically-acceptable carrier, e.g., a saline solution or other accepted physiologically-acceptable solution for in vivo administration. Pharmaceutical compositions of the invention can comprise any of the placental stem cell populations, or placental stem cell types, described elsewhere herein. The pharmaceutical compositions can comprise fetal, maternal, or both fetal and maternal placental stem cells. The pharmaceutical compositions of the invention can further comprise placental stem cells obtained from a single individual or placenta, or from a plurality of individuals or placentae.
[0223] The pharmaceutical compositions of the invention can comprise any number of placental stem cells. For example, a single unit dose of placental stem cells can comprise, in various embodiments, about, at least, or no more than 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 1O9, 5 x 109, 1 x 1010, 5 x 1010, 1 x lθ" or more placental stem cells.
[0224] The pharmaceutical compositions of the invention comprise populations of cells that comprise 50% viable cells or more (that is, at least 50% of the cells in the population are functional or living). Preferably, at least 60% of the cells in the population are viable. More preferably, at least 70%, 80%, 90%, 95%, or 99% of the cells in the population in the pharmaceutical composition are viable.
[0225] The pharmaceutical compositions of the invention can comprise one or more compounds that, e.g., facilitate engraftment {e.g., anti-T-cell receptor antibodies, an immunosuppressant, or the like); stabilizers such as albumin, dextran 40, gelatin, hydroxyethyl starch, and the like.
[0226] When formulated as an injectable solution, in one embodiment, the pharmaceutical composition of the invention comprises about 1.25% HSA and about 2.5% dextran. Other injectable formulations, suitable for the administration of cellular products, may be used. [0227] In one embodiment, the composition of the invention comprises placental stem cells that are substantially, or completely, non-maternal in origin. For example, the invention provides in one embodiment a composition comprising a population of placental stem cells that are CD200+ and HLA-G+; CD73+, CD105+, and CD200+; CD200+ and OCT-4+; CD73+, CD105+ and HLA-G+; CD73+ and CDl 05+ and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said population of placental stem cell when said population of placental cells is cultured under conditions that allow the formation of an embryoid-like body; or OCT-4+ and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said population of placental stem cell when said population of placental cells is cultured under conditions that allow the formation of an embryoid-like body; or a combination of the foregoing, wherein at least 70%, 80%, 90%, 95% or 99% of said placental stem cells are non-maternal in origin. In a specific embodiment, the composition additionally comprises a stem cell that is not obtained from a placenta. 5.7.2.3 Placental Stem Cell Conditioned Media
[0228] The placental stem cells of the invention can be used to produce conditioned medium, that is, medium comprising one or more biomolecules secreted or excreted by the stem cells. In various embodiments, the conditioned medium comprises medium in which placental stem cells have grown for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 or more days. In other embodiments, the conditioned medium comprises medium in which placental stem cells have grown to at least 30%, 40%, 50%, 60%, 70%, 80%, 90% confluence, or up to 100% confluence. Such conditioned medium can be used to support the culture of a separate population of placental stem cells, or stem cells of another kind. In another embodiment, the conditioned medium comprises medium in which placental stem cells have been differentiated into an adult cell type. In another embodiment, the conditioned medium of the invention comprises medium in which placental stem cells and non-placental stem cells have been cultured.
5.7.2.4 Matrices Comprising Placental Stem Cells
[0229] The invention further comprises matrices, hydrogels, scaffolds, and the like that comprise a placental stem cell, or a population of placental stem cells. [0230] Placental stem cells of the invention can be seeded onto a natural matrix, e.g., a placental biomaterial such as an amniotic membrane material. Such an amniotic membrane material can be, e.g., amniotic membrane dissected directly from a mammalian placenta; fixed or heat-treated amniotic membrane, substantially dry (i.e., <20% H2O) amniotic membrane, chorionic membrane, substantially dry chorionic membrane, substantially dry amniotic and chorionic membrane, and the like. Preferred placental biomaterials on which placental stem cells can be seeded are described in Hariri, U.S. Application Publication No. 2004/0048796.
[0231] Placental stem cells of the invention can be suspended in a hydrogel solution suitable for, e.g., injection. Suitable hydrogels for such compositions include self-assembling peptides, such as RAD 16. In one embodiment, a hydrogel solution comprising the cells can be allowed to harden, for instance in a mold, to form a matrix having cells dispersed therein for implantation. Placental stem cells in such a matrix can also be cultured so that the cells are mitotically expanded prior to implantation. The hydrogel is, e.g., an organic polymer (natural or synthetic) that is cross-linked via covalent, ionic, or hydrogen bonds to create a three-dimensional open-lattice structure that entraps water molecules to form a gel. Hydrogel-forming materials include polysaccharides such as alginate and salts thereof, peptides, polyphosphazines, and polyacrylates, which are crosslinked ionically, or block polymers such as polyethylene oxide-polypropylene glycol block copolymers which are crosslinked by temperature or pH, respectively. In some embodiments, the hydrogel or matrix of the invention is biodegradable.
[0232J In some embodiments of the invention, the formulation comprises an in situ polymerizable gel (see., e.g., U.S. Patent Application Publication 2002/0022676; Anseth et al., J. Control Release, 78(1 -3): 199-209 (2002); Wang et ah, Biomaterials, 24(22):3969-80
(2003).
[0233] In some embodiments, the polymers are at least partially soluble in aqueous solutions, such as water, buffered salt solutions, or aqueous alcohol solutions, that have charged side groups, or a monovalent ionic salt thereof. Examples of polymers having acidic side groups that can be reacted with cations are poly(phosphazenes), poly(acrylic acids), poly(methacrylic acids), copolymers of acrylic acid and methacrylic acid, poly( vinyl acetate), and sulfonated polymers, such as sulfonated polystyrene. Copolymers having acidic side groups formed by reaction of acrylic or methacrylic acid and vinyl ether monomers or polymers can also be used. Examples of acidic groups are carboxylic acid groups, sulfonic acid groups, halogenated (preferably fluorinated) alcohol groups, phenolic OH groups, and acidic OH groups.
[0234] The placental stem cells of the invention or co-cultures thereof can be seeded onto a three-dimensional framework or scaffold and implanted in vivo. Such a framework can be implanted in combination with any one or more growth factors, cells, drugs or other components that stimulate tissue formation or otherwise enhance or improve the practice of the invention.
[0235] Examples of scaffolds that can be used in the present invention include nonwoven mats, porous foams, or self assembling peptides. Nonwoven mats can be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (e.g., PGA/PLA)
(VICRYL, Ethicon, Inc., Somerville, NJ.). Foams, composed of, e.g., poly(ε- caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, formed by processes such as freeze-drying, or lyophilization (see, e.g., U.S. Pat. No. 6,355,699), can also be used as scaffolds.
[0236] Placental stem cells of the invention can also be seeded onto, or contacted with, a physiologically-acceptable ceramic material including, but not limited to, mono-, di-, tri-, alpha-tri-, beta-tri-, and tetra-calcium phosphate, hydroxyapatite, fluoroapatites, calcium sulfates, calcium fluorides, calcium oxides, calcium carbonates, magnesium calcium phosphates, biologically active glasses such as BIOGLASS®, and mixtures thereof. Porous biocompatible ceramic materials currently commercially available include SURGIBONE® (CanMedica Corp., Canada), ENDOBON® (Merck Biomaterial France, France), CEROS® (Mathys, AG3 Bettlach, Switzerland), and mineralized collagen bone grafting products such as HEALOS™ (DePuy, Inc., Raynham, MA) and VITOSS®, RHAKΘSS™, and CORTOSS® (Orthovita, Malvern, Pa.). The framework can be a mixture, blend or composite of natural and/or synthetic materials.
[0237] In another embodiment, placental stem cells can be seeded onto, or contacted with, a felt, which can be, e.g., composed of a multifilament yarn made from a bioabsorbable material such as PGA, PLA, PCL copolymers or blends, or hyaluronic acid. [0238] The placental stem cells of the invention can, in another embodiment, be seeded onto foam scaffolds that may be composite structures. Such foam scaffolds can be molded into a useful shape, such as that of a portion of a specific structure in the body to be repaired, replaced or augmented. In some embodiments, the framework is treated, e.g., with 0.1M acetic acid followed by incubation in polylysine, PBS, and/or collagen, prior to inoculation of the cells of the invention in order to enhance cell attachment. External surfaces of a matrix may be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma-coating the matrix, or addition of one or more proteins (e.g., collagens, elastic fibers, reticular fibers), glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate, etc.), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, and the like.
[0239] In some embodiments, the scaffold comprises, or is treated with, materials that render it non-thrombogenic. These treatments and materials may also promote and sustain endothelial growth, migration, and extracellular matrix deposition. Examples of these materials and treatments include but are not limited to natural materials such as basement membrane proteins such as laminin and Type IV collagen, synthetic materials such as EPTFE, and segmented polyurethaneurea silicones, such as PURSPAN™ (The Polymer Technology Group, Inc., Berkeley, Calif.). The scaffold can also comprise anti-thrombotic agents such as heparin; the scaffolds can also be treated to alter the surface charge (e.g., coating with plasma) prior to seeding with placental stem cells.
5.7.3 Immortalized Placental Stem Cell Lines
[0240] Mammalian placental cells can be conditionally immortalized by transfection with any suitable vector containing a growth-promoting gene, that is, a gene encoding a protein that, under appropriate conditions, promotes growth of the transfected cell, such that the production and/or activity of the growth-promoting protein is regulatable by an external factor. In a preferred embodiment the growth-promoting gene is an oncogene such as, but not limited to, v-myc, N-myc, c-myc, p53, SV40 large T antigen, polyoma large T antigen, EIa adenovirus or E7 protein of human papillomavirus.
[0241J External regulation of the growth-promoting protein can be achieved by placing the growth-promoting gene under the control of an externally-regulatable promoter, e.g., a promoter the activity of which can be controlled by, for example, modifying the temperature of the transfected cells or the composition of the medium in contact with the cells, in one embodiment, a tetracycline (tet)-controlled gene expression system can be employed (see Gossen et al, Proc. Natl. Acad. Sci. USA 89:5547-5551, 1992; Hoshimaru et al, Proc. Natl. Acad. Sci. USA 93:1518-1523, 1996). In the absence of tet, a tet-controlled transactivator (tTA) within this vector strongly activates transcription from phcw-i, a minimal promoter from human cytomegalovirus fused to tet operator sequences. tTA is a fusion protein of the repressor (tetR) of the transposon-10-derived tet resistance operon of Escherichia coli and the acidic domain of VP16 of herpes simplex virus. Low, non-toxic concentrations of tet (e.g., 0.01-1.0 μg/mL) almost completely abolish transactivation by tTA.
[0242] In one embodiment, the vector further contains a gene encoding a selectable marker, e.g., a protein that confers drug resistance. The bacterial neomycin resistance gene (neoΛ) is one such marker that may be employed within the present invention. Cells carrying neoΛ may be selected by means known to those of ordinary skill in the art, such as the addition of, e.g., 100-200 μg/mL G418 to the growth medium.
[0243] Transfection can be achieved by any of a variety of means known to those of ordinary skill in the art including, but not limited to, retroviral infection. In general, a cell culture may be transfected by incubation with a mixture of conditioned medium collected from the producer cell line for the vector and DMEM/F12 containing N2 supplements. For example, a placental cell culture prepared as described above may be infected after, e.g., five days in vitro by incubation for about 20 hours in one volume of conditioned medium and two volumes of DMEM/F12 containing N2 supplements. Transfected cells carrying a selectable marker may then be selected as described above.
[0244] Following transfection, cultures are passaged onto a surface that permits proliferation, e.g., allows at least 30% of the cells to double in a 24 hour period. Preferably, the substrate is a polyornithine/laminin substrate, consisting of tissue culture plastic coated with polyornithine (10 μg/mL) and/or laminin (10 μg/mL), a polylysine/laminin substrate or a surface treated with fibronectin. Cultures are then fed every 3-4 days with growth medium, which may or may not be supplemented with one or more proliferation-enhancing factors. Proliferation-enhancing factors may be added to the growth medium when cultures are less than 50% confluent.
[0245] The conditionally-immortalized placental stem cell lines can be passaged using standard techniques, such as by trypsinization, when 80-95% confluent. Up to approximately the twentieth passage, it is, in some embodiments, beneficial to maintain selection (by, for example, the addition of G418 for cells containing a neomycin resistance gene). Cells may also be frozen in liquid nitrogen for long-term storage.
[0246] Clonal cell lines can be isolated from a conditionally-immortalized human placental stem cell line prepared as described above. In general, such clonal cell lines may be isolated using standard techniques, such as by limit dilution or using cloning rings, and expanded. Clonal cell lines may generally be fed and passaged as described above. [0247] Conditionally-immortalized human placental stem cell lines, which may, but need not, be clonal, may generally be induced to differentiate by suppressing the production and/or activity of the growth-promoting protein under culture conditions that facilitate differentiation. For example, if the gene encoding the growth-promoting protein is under the control of an externally-regulatable promoter, the conditions, e.g., temperature or composition of medium, may be modified to suppress transcription of the growth-promoting gene. For the tetracycline-controlled gene expression system discussed above, differentiation can be achieved by the addition of tetracycline to suppress transcription of the growth- promoting gene. In general, 1 μg/mL tetracycline for 4-5 days is sufficient to initiate differentiation. To promote further differentiation, additional agents may be included in the growth medium.
5.7.4 Assays
[0248] The placental stem cells for the present invention can be used in assays to determine the influence of culture conditions, environmental factors, molecules (e.g., biomolecules, small inorganic molecules, etc.) and the like on stem ceil proliferation, expansion, and/or differentiation, compared to placental stem cells not exposed to such conditions. [0249] In a preferred embodiment, the placental stem cells of the present invention are assayed for changes in proliferation, expansion or differentiation upon contact with a molecule. In one embodiment, for example, the invention provides a method of identifying a compound that modulates the proliferation of a plurality of placental stem cells, comprising contacting said plurality of stem cells with said compound under conditions that allow proliferation, wherein if said compound causes a detectable change in proliferation of said plurality of stem cells compared to a plurality of stem cells not contacted with said compound, said compound is identified as a compound that modulates proliferation of placental stem cells. In a specific embodiment, said compound is identified as an inhibitor of proliferation. In another specific embodiment, said compound is identified as an enhancer of proliferation.
[0250] In another embodiment, the invention provides a method of identifying a compound that modulates the expansion of a plurality of placental stem cells, comprising contacting said plurality of stem cells with said compound under conditions that allow expansion, wherein if said compound causes a detectable change in expansion of said plurality of stem cells compared to a plurality of stem cells not contacted with said compound, said compound is identified as a compound that modulates expansion of placental stem cells. In a specific embodiment, said compound is identified as an inhibitor of expansion. In another specific embodiment, said compound is identified as an enhancer of expansion. [0251] In another embodiment, the invention provides a method of identifying a compound that modulates the differentiation of a placental stem cell, comprising contacting said stem cells with said compound under conditions that allow differentiation, wherein if said compound causes a detectable change in differentiation of said stem cells compared to a stem cell not contacted with said compound, said compound is identified as a compound that modulates proliferation of placental stem cells. In a specific embodiment, said compound is identified as an inhibitor of differentiation. In another specific embodiment, said compound is identified as an enhancer of differentiation.
6. EXAMPLES
6.1 EXAMPLE 1 : CULTURE OF PLACENTAL STEM CELLS [0252] Placental stem cells are obtained from a post-partum mammalian placenta either by perfusion or by physical disruption, e.g., enzymatic digestion. The cells are cultured in a culture medium comprising 60% DMEM-LG (Gibco), 40% MCDB-201 (Sigma), 2% fetal calf serum (FCS) (Hyclone Laboratories), Ix insulin-transferrin-selenium (ITS), Ix lenolenic- acid-bovine-serum-albumin (LA-BSA)5 10"9 M dexamethasone (Sigma), 10"4M ascorbic acid 2-phosphate (Sigma), epidermal growth factor (EGF) 10ng/ml (R&D Systems), platelet derived-growth factor (PDGF-BB) 10ng/ml (R&D Systems), and IOOU penicillin/100OU streptomycin. [0253] The culture flask in which the cells are cultured is prepared as follows. T75 flasks are coated with fibronectin (FN), by adding 5 ml PBS containing 5ng/ml human FN (Sigma F0895) to the flask. The flasks with FN solution are left at 37°C for 30 min. The FN solution is then removed prior to cell culture. There is no need to dry the flasks following treatment. Alternatively, the flasks are left in contact with the FN solution at 4°C overnight or longer; prior to culture, the flasks are warmed and the FN solution is removed.
Placental Stem Cells Isolated By Perfusion
[0254] Cultures of placental stem cells from placental perfusate are established as follows. Cells from a Ficoll gradient are seeded in FN-coated T75 flasks, prepared as above, at 50- 100x106 cells/flask in 15 ml culture medium. Typically, 5 to 10 flasks are seeded. The flasks are incubated at 370C for 12-18 hrs to allow the attachment of adherent cells. 10 ml of warm PBS is added to each flask to remove cells in suspension, and mixed gently. 15 rnL of the medium is then removed and replaced with 15 ml fresh culture medium. All medium is changed 3-4 days after the start of culture. Subsequent culture medium changes are performed, during which 50% or 7.5 ml of the medium is removed.
[0255] Starting at about day 12, the culture is checked under a microscope to examine the growth of the adherent cell colonies. When cell cultures become approximately 80% confluent, typically between day 13 to day 18 after the start of culture, adherent cells are harvested by trypsin digestion. Cells harvested from these primary cultures are designated passage 0 (zero).
Placental Stem Cells Isolated By Physical Disruption and Enzymatic Digestion [0256] Placental stem cell cultures are established from digested placental tissue as follows. The perfused placenta is placed on a sterile paper sheet with the maternal side up. Approximately 0.5 cm of the surface layer on maternal side of placenta is scraped off with a blade, and the blade is used to remove a placental tissue block measuring approximately 1 x 2 x 1 cm. This placenta tissue is then minced into approximately lmm3 pieces. These pieces are collected into a 50ml Falcon tube and digested with collagenase IA (2mg/ml, Sigma) for 30 minutes, followed by trypsin-EDTA (0.25%, GIBCO BRL) for 10 minutes, at 370C in water bath. The resulting solution is centrifuged at 40Og for 10 minutes at room temperature, and the digestion solution is removed. The pellet is resuspended to approximately 10 volumes with PBS (for example, a 5 ml pellet is resuspended with 45 ml PBS), and the tubes are centrifuged at 40Og for 10 minutes at room temperature. The tissue/cell pellet is resuspended in 130 mL culture medium, and the cells are seeded at 13ml per fibronectin- coated T-75 flask. Cells are incubated at 37°C with a humidified atmosphere with 5% CO2. Placental Stem Cells are optionally cryopreserved at this stage.
Subculturing and Expansion of Placental Stem Cells
[0257] Cryopreserved cells are quickly thawed in a 370C water bath. Placental stem cells are immediately removed from the cryovial with 10ml warm medium and transferred to a 15ml sterile tube. The cells are centrifuged at 40Og for 10 minutes at room temperature. The cells are gently resuspended in 10ml of warm culture medium by pipetting, and viable cell counts are determined by Trypan blue exclusion. Cells are then seeded at about 6000-7000 cells per cm2 onto FN-coated flasks, prepared as above (approximately 5x105 cells per T-75 flask). The cells are incubated at 37°C, 5% CO2 and 90% humidity. When the cells reached 75-85% confluency, all of the spent media is aseptically removed from the flasks and discarded. 3ml of 0.25% trypsin/EDTA (w/v) solution is added to cover the cell layer, and the cells are incubated at 37°C, 5% CO2 and 90% humidity for 5 minutes. The flask is tapped once or twice to expedite cell detachment. Once >95% of the cells are rounded and detached, 7ml of warm culture medium is added to each T-75 flask, and the solution is dispersed by pipetting over the cell layer surface several times.
[0258] After counting the cells and determining viability as above, the cells are centrifuged at 1000 RPM for 5 minutes at room temperature. Cells are passaged by gently resuspending the cell pellet from one T-75 flask with culture medium, and evenly plating the cells onto two FN-coated T-75 flasks.
[0259] Using the above methods, exemplary populations of adherent placental stem cells are identified that express markers CD105, CD33, CD73, CD29, CD44, CDlO, and CD90. These populations of cells typically does not express CD34, CD45, CDl 17 or CD133. Some, but not all cultures of these placental stem cells expressed HLA-ABC and/or HLA-DR.
6.2 EXAMPLE 2: ISOLATION OF PLACENTAL STEM CELLS FROM . PLACENTAL STRUCTURES
6.2.1 Materials & Methods
6.2.1.1 Isolation of Populations of Placental Cells Comprising Placental Stem Cells
[0260] Distinct populations of placental cells were obtained from the placentas of normal, full-term pregnancies. All donors provided full written consent for the use of their placentas for research purposes. Placental stem cells were obtained from the following sources: (1) placental perfusate (from perfusion of the placental vasculature); and enzymatic digestions of (2) amnion, (3) chorion, (4) amnion-chorion plate, and (5) umbilical cord. The various placental tissues were cleaned in sterile PBS (Gibco-Invitrogen Corporation, Carlsbad, CA) and placed on separate sterile Petri dishes. The various tissues were minced using a sterile surgical scalpel and placed into 50 mL Falcon Conical tubes. The minced tissues were digested with IX Collagenase (Sigma- Aldrich, St. Louis, MO) for 20 minutes in a 370C water bath, centrifuged, and then digested with 0.25% Trypsin-EDTA (Gibco-Invitrogen Corp) for 10 minutes in a 370C water bath. The various tissues were centrifuged after digestion and rinsed once with sterile PBS (Gibco-Invitrogen Corp). The reconstituted cells were then filtered twice, once with 100 μm cell strainers and once with 30 μm separation filters, to remove any residual extracellular matrix or cellular debris.
6.2.1.2 Cellular Viability Assessment and Cell Counts
[0261] The manual trypan blue exclusion method was employed post digestion to calculate cell counts and assess cellular viability. Cells were mixed with Trypan Blue Dye (Sigma- Aldrich) at a ratio of 1 : 1, and the cells were read on hemacytometer.
6.2.1.3 Cell Surface Marker Characterization
[0262] Cells that were HLA ABC7CD457CD347CD133"1" were selected for characterization. Cells having this phenotype were identified, quantified, and characterized by two of Becton- Dickinson flow cytometers, the FACSCalibur and the FACS Aria (Becton-Dickinson, San Jose, CA, USA). The various placental cells were stained, at a ratio of about 10 μL of antibody per 1 million cells, for 30 minutes at room temperature on a shaker. The following anti-human antibodies were used: Fluorescein Isothiocyanate (FITC) conjugated monoclonal antibodies against HLA-G (Serotec, Raleigh, NC), CDlO (BD Immunocytometry Systems, San Jose, CA), CD44 (BD Biosciences Pharmingen, San Jose, CA), and CD 105 (R&D Systems Inc., Minneapolis, MN); Phycoerythrin (PE) conjugated monoclonal antibodies against CD44, CD200, CDl 17, and CD 13 (BD Biosciences Pharmingen); Phycoerythrin-Cy5 (PE Cy5) conjugated Streptavidin and monoclonal antibodies against CDl 17 (BD Biosciences Pharmingen); Phycoerythrin-Cy7 (PE Cy 7) conjugated monoclonal antibodies against CD33 and CDlO (BD Biosciences); Allophycocyanin (APC) conjugated streptavidin and monoclonal antibodies against CD38 (BD Biosciences Pharmingen); and Biotinylated CD90 (BD Biosciences Pharmingen). After incubation, the cells were rinsed once to remove unbound antibodies and were fixed overnight with 4% paraformaldehyde (USB, Cleveland, OH) at 40C. The following day, the cells were rinsed twice, filtered through a 30 μm separation filter, and were run on the flow cytometer(s). W
[0263] Samples that were stained with anti-mouse IgG antibodies (BD Biosciences Pharmingen) were used as negative controls and were used to adjust the Photo Multiplier Tubes (PMTs). Samples that were single stained with anti-human antibodies were used as positive controls and were used to adjust spectral overlaps/compensations.
6.2.1 A Cell Sorting and Culture
[0264] One set of placental cells (from perfusate, amnion, or chorion), prior to any culture, was stained with 7-Amino-Actinomycin D (7AAD; BD Biosciences Pharmingen) and monoclonal antibodies specific for the phenotype of interest. The cells were stained at a ratio of 10 μL of antibody per 1 million cells, and were incubated for 30 minutes at room temperature on a shaker. These cells were then positively sorted for live cells expressing the phenotype of interest on the BD FACS Aria and plated into culture. Sorted (population of interest) and "All" (non-sorted) placental cell populations were plated for comparisons. The cells were plated onto a fibronectin (Sigma-Aldrich) coated 96 well plate at the cell densities listed in Table 1 (cells/cm2). The cell density, and whether the cell type was plated in duplicate or triplicate, was determined and governed by the number of cells expressing the phenotype of interest.
Table 1 : Cell plating densities
Figure imgf000075_0001
[0265] Complete medium (60% DMEM-LG (Gibco) and 40% MCDB-201 (Sigma); 2% fetal calf serum (Hyclone Labs.); Ix insulin-transferrin-selenium (ITS); Ix Iinoleic acid-bovine serum albumin (LA-BSA); 10"9 M dexamethasone (Sigma); 10"4 M ascorbic acid 2-phosphate (Sigma); epidermal growth factor 10 ng/mL (R&D Systems); and platelet-derived growth factor (PDGF-BB) 10 ng/mL (R&D Systems)) was added to each well of the 96 well plate and the plate was placed in a 5% CO2/37 C incubator. On day 7, 100 μL of complete medium was added to each of the wells. The 96 well plate was monitored for about two weeks and a final assessment of the culture was completed on day 12. This is very early in the placental stem cell culture, and represents passage 0 cells.
6.2.1.5 Data Analysis
[0266] FACSCalibur data was analyzed in Flow Jo (Tree star, Inc) using standard gating techniques. The BD FACS Aria data was analyzed using the FACSDiva software (Becton- Dickinson). The FACS Aria data was analyzed using doublet discrimination gating to minimize doublets, as well as, standard gating techniques. All results were compiled in Microsoft Excel and all values, herein, are represented as average ± standard deviation (number, standard error of mean).
6.2.2 Results
6.2.2.1 Cellular Viability
[0267] Post-digestion viability was assessed using the manual trypan blue exclusion method (FIG 1). The average viability of cells obtained from the majority of the digested tissue (from amnion, chorion or amnion-chorion plate) was around 70%. Amnion had an average viability of 74.35% ±10.31% (n=6, SEM=4.21), chorion had an average viability of 78.18% ± 12.65% (n=4, SEM=6.32), amnion-chorion plate had an average viability of 69.05% ±10.80% (n=4, SEM=5.40), and umbilical cord had an average viability of 63.30% ±20.13% (n=4, SEM= 10.06). Cells from perfusion, which did not undergo digestion, retained the highest average viability, 89.98±6.39% (n=5, SEM=2.86).
6.2.2.2 Cell Quantification
[0268] The populations of placental cells and umbilical cord cells were analyzed to determine the numbers of HLA ABC7CD457CD347CD133+ cells. From the analysis of the BD FACSCalibur data, it was observed that the amnion, perfusate, and chorion contained the greatest total number of these cells, 30.72 ± 21.80 cells (n=4, SEM=I 0.90), 26.92 ± 22.56 cells (n=3, SEM=I 3.02), and 18.39 ± 6.44 cells (n=2, SEM=4.55) respectively (data not shown). The amnion-chorion plate and umbilical cord contained the least total number of cells expressing the phenotype of interest, 4.72 ± 4.16 cells (n=3, SEM=2.40) and 3.94 ± 2.58 cells (n=3, SEM=I .49) respectively (data not shown). [0269] Similarly, when the percent of total cells expressing the phenotype of interest was analyzed, it was observed that amnion and placental perfusate contained the highest percentages of cells expressing this phenotype (0.0319% ± 0.0202% (n=4, SEM=0.0101) and 0.0269% -t 0.0226% (n=3, SEM=O-0130) respectively (FIG. 2). Although umbilical cord contained a small number of cells expressing the phenotype of interest (FIG. 2), it contained the third highest percentage of cells expressing the phenotype of interest, 0.020±0.0226% (n=3, SEM=O10131) (FIG. 2). The chorion and amnion-chorion plate contained the lowest percentages of cells expressing the phenotype of interest, 0.0184±0.0064% (n=2, SEM=0.0046) and 0.0177±0.0173% (n=3, SEM=COlO) respectively (FIG. 2). [0270] Consistent with the results of the BD FACSCalibur analysis, the BD FACS Aria data also identified amnion, perfusate, and chorion as providing higher numbers of HLA ABC" /CD457CD347CD133+ cells than the remaining sources. The average total number of cells expressing the phenotype of interest among amnion, perfusate, and chorion was 126.47 ± 55.61 cells (n=15, SEM=14.36), 81.65 ± 34.64 cells (n=20, SEM=7.75), and 51.47 ± 32.41 cells (n=15, SEM=8.37), respectively (data not shown). The amnion-chorion plate and * umbilical cord contained the least total number of cells expressing the phenotype of interest, 44.89 ± 37.43 cells (n=9, SEM=12.48) and 11.00 ± 4.03 cells (n=9, SEM=I .34) respectively (data not shown).
[0271] BD FACS Aria data revealed that the perfusate and amnion produced the highest percentages of HLA ABC7CD457CD347CD133+ cells, 0.1523 ± 0.0227% (n=15, SEM=0.0059) and 0.0929 ± 0.0419% (n=20, SEM=0.0094) respectively (FIG. 3). The amnion-chorion plate contained the third highest percentage of cells expressing the phenotype of interest, 0.0632±0.0333% (n=9, SEM=0.0111) (FIG. 3). The chorion and umbilical cord contained the lowest percentages of cells expressing the phenotype of interest, 0.0623±0.0249% (n=15, SEM=0.0064) and 0.0457±0.0055% (n=9, SEM=O-OO 18) respectively (FIG. 3).
[0272] After HLA ABC7CD457CD347CD133+ cells were identified and quantified from each cell source, its cells were further analyzed and characterized for their expression of cell surface markers HLA-G, CDlO, CD13, CD33, CD38, CD44, CD90, CD105, CDl 17, CD200, and CD 105.
6.2.2.3 Placental Perfusate-Derived Cells
[0273] Perfusate-derived cells were consistently positive for HLA-G, CD33, CDl 17, CDlO, CD44, CD200, CD90, CD38, CD105, and CD13 (FIG. 4). The average expression of each marker for perfusate-derived cells was the following: 37.15% ± 38.55% (n=4, SEM= 19.28) of the cells expressed HLA-G; 36.37% ± 21.98% (n=7, SEM=8.31) of the cells expressed CD33; 39.39% ± 39.91% (n=4, SEM=19.96) of the cells expressed CDl 17; 54.97% ± 33.08% (n=4, SEM=16.54) of the cells expressed CDlO; 36.79% ± 11.42% (n=4, SEM=5.71) of the cells expressed CD44; 41.83% ± 19.42% (n=3, SEM=I 1.21) of the cells expressed CD200; 74.25% ± 26.74% (n=3, SEM=I 5.44) of the cells expressed CD90; 35.10% ± 23.10% (n=3, SEM=13.34) of the cells expressed CD38; 22.87% ± 6.87% (n=3, SEM=3.97) of the cells expressed CD105; and 25.49% ± 9.84% (n=3, SEM=5.68) of the cells expressed CD13.
6.2.2.4 Amnion-Derived Cells
[0274} Amnion-derived cells were consistently positive for HLA-G, CD33, CDl 17, CDlO, CD44, CD200, CD90, CD38, CD105, and CD13 (FIG 5). The average expression of each marker for amnion-derived was the following: 57.27% ± 41.11% (n=3, SEM=23.73) of the cells expressed HLA-G; 16.23% ± 15.81% (n=6, SEM=6.46) of the cells expressed CD33; 62.32% ± 37.89% (n=3, SEM=21.87) of the cells expressed CDl 17; 9.71% ± 13.73% (n=3, SEM=7.92) of the cells expressed CDlO; 27.03% ± 22.65% (n=3, SEM=13.08) of the cells expressed CD44; 6.42% ± 0.88% (n=2, SEM=0.62) of the cells expressed CD200; 57.61% ±22.10% (n=2, SEM=15.63) of the cells expressed CD90; 63.76% ± 4.40% (n=2, SEM=3.11) of the cells expressed CD38; 20.27% ± 5.88% (n=2, SEM=4.16) of the cells expressed CD105; and 54.37% ± 13.29% (n=2, SEM=9.40) of the cells expressed CD13.
6.2.2.5 Chorion-Derived Cells
[0275] Chorion-derived cells were consistently positive for HLA-G, CDl 17, CDlO, CD44, CD200, CD90, CD38, and CD13, while the expression of CD33, and CD105 varied (FIG. 6). The average expression of each marker for chorion cells was the following: 53.25% ± 32.87% (n=3, SEM=18.98) of the cells expressed HLA-G; 15.44% ± 11.17% (n=6, SEM=4.56) of the cells expressed CD33; 70.76% ± 11.87% (n=3, SEM=6.86) of the cells expressed CDl 17; 35.84% ± 25.96% (n=3, SEM=14.99) of the cells expressed CDlO; 28.76% ± 6.09% (n=3, SEM=3.52) of the cells expressed CD44; 29.20% ± 9.47% (n=2, SEM=6.70) of the cells expressed CD200; 54.88% ± 0.17% (n=2, SEM-0.12) of the cells expressed CD90; 68.63% ± 44.37% (n=2, SEM=31.37) of the cells expressed CD38; 23.81% ± 33.67% (n=2, SEM=23.81) of the cells expressed CDl 05; and 53.16% ± 62.70% (n=2, SEM=44.34) of the cells expressed CDl 3.
6.2.2.6 Amnion-Chorion Plate-Derived Cells
[0276] Cells from amnion-chorion plate were consistently positive for HLA-G, CD33, CDl 17, CDlO, CD44, CD200, CD90, CD38, CD 105, and CD 13 (FIG. 7). The average expression of each marker for amnion-chorion plate-derived cells was the following: 78.52% ±13.13% (n=2, SEM=9.29) of the cells expressed HLA-G; 38.33% ± 15.74% (n=5, SEM=7.04) of the cells expressed CD33; 69.56% ± 26.41% (n=2, SEM=I 8.67) of the cells expressed CDl 17; 42.44% ± 53.12% (n=2, SEM=37.56) of the cells expressed CDlO; 32.47% ± 31.78% (n=2, SEM=22.47) of the cells expressed CD44; 5.56% (n=l) of the cells expressed CD200; 83.33% (n=l) of the cells expressed CD90; 83.52% (n=l) of the cells expressed CD38; 7.25% (n=l) of the cells expressed CD105; and 81.16% (n=l) of the cells expressed CDl 3.
6.2.2.7 Umbilical Cord-Derived Cells
[0277] Umbilical cord-derived cells were consistently positive for HLA-G, CD33, CD90, CD38, CD 105, and CDl 3, while the expression of CDl 17, CDlO, CD44, and CD200 varied (FIG. 8). The average expression of each marker for umbilical cord-derived cells was the following: 62.50% ± 53.03% (n=2, SEM=37.50) of the cells expressed HLA-G; 25.67% ± 11.28% (n=5, SEM=5.04) of the cells expressed CD33; 44.45% ± 62.85% (n=2, SEM=44.45) of the cells expressed CDl 17; 8.33% ± 11.79% (n=2, SEM=8.33) of the cells expressed CDlO; 21.43% ± 30.30% (n=2, SEM=21.43) of the cells expressed CD44; 0.0% (n=l) of the cells expressed CD200; 81.25% (n=l) of the cells expressed CD90; 64.29% (n=l) of the cells expressed CD38; 6.25% (n=l) of the cells expressed CD105; and 50.0% (n=l) of the cells expressed CDl 3. [0278] A summary of all marker expression averages is shown in FIG. 9.
6.2.2.8 BD FACS Aria Sort Report
[0279] The three distinct populations of placental cells that expressed the greatest percentages of HLA ABC, CD45, CD34, and CD 133 (cells derived from perfusate, amnion and chorion) were stained with 7AAD and the antibodies for these markers. The three populations were positively sorted for live cells expressing the phenotype of interest. The results of the BD FACS Aria sort are listed in table 2.
Table 2:
BD FACS Aria Sort Report
Events Sorted
Cell Source Events Processed (Phenotype of % Of Total
Interest)
Figure imgf000080_0001
[0280] The three distinct populations of positively sorted cells ("sorted") and their corresponding non-sorted cells were plated and the results of the culture were assessed on day 12 (Table 3). Sorted perfusate-derived cells, plated at a cell density of 40,600/cm2, resulted in small, round, non-adherent cells. Two out of the three sets of non-sorted perfusate-derived cells, each plated at a cell density of 40,600/cm2, resulted in mostly small, round, nonadherent cells with several adherent cells located around the periphery of well. Non-sorted perfusate-derived cells, plated at a cell density of 93,800/cm2, resulted in mostly small, round, non-adherent cells with several adherent cells located around the well peripheries. [0281] Sorted amnion-derived cells, plated at a cell density of 6,300/cm2, resulted in small, round, non-adherent cells. Non-sorted amnion-derived cells, plated at a cell density of 6,300/cm2, resulted in small, round, non-adherent cells. Non-sorted amnion-derived cells plated at a cell density of 62,500/cm2 resulted in small, round, non-adherent cells. [0282] Sorted chorion-derived cells, plated at a cell density of 6,300/cm2, resulted in small, round, non-adherent cells. Non-sorted chorion-derived cells, plated at a cell density of 6,300/cm2, resulted in small, round, non-adherent cells. Non-sorted chorion-derived cells plated at a cell density of 62,500/cm2, resulted in small, round, non-adherent cells. [0283] Subsequent to the performance of the experiments related above, and further culture of the placental stem cells, it was determined that the labeling of the antibodies for CDl 17 and CDl 33, in which a streptavidin-conjugated antibody was labeled with biotin-conjugated phycoerythrin (PE), produced background significant enough to resemble a positive reading. This background had initially resulted in the placental stem cells being deemed to be positive for both markers. When a different label, APC or PerCP was used, the background was reduced, and the placental stem cells were correctly determined to be negative for both CD117 and CD133.
6.3 EXAMPLE 3 : CHARACTERIZATION OF PLACENTAL STEM CELLS AND UMBILICAL CORD STEM CELLS
[0284] This Example demonstrates an exemplary cell surface marker profile of placental stem cells. [0285] Placental stem cells or umbilical cord stem cells, obtained by enzymatic digestion, in culture medium were washed once by adding 2 mL 2% FBS-PBS and centrifuging at 40Og for 5 minutes. The supernatant was decanted, and the pellet was resuspended in 100-200 μL 2% FBS-PBS. 4 tubes were prepared with BD™ CompBeads (Cat# 552843) by adding 100 μl of 2% FBS-PBS to each tube, adding 1 full drop (approximately 60 μl) of the BD™ CompBeads Negative Control and 1 drop of the BD™ CompBeads Anti-Mouse beads to each tube, and vortexing. To the 4 tubes of BD™ CompBeads, the following antibodies were added:
Figure imgf000081_0001
[0286] Control tubes were prepared as follows:
Figure imgf000081_0002
[0287] The following antibodies were added to the sample tubes:
Figure imgf000081_0003
[0288] The control and sample tubes were incubated in the dark at room temperature for 30 minutes. After incubation, the tubes were washed by adding 2mL 2% FBS-PBS and centrifuging at 40Og for 5 minutes. The supernatant was decanted, and the pellet was resuspended in 100-200 μL 2% FBS-PBS and acquire on flow cytometer. All other antibodies were used following this procedure. [0289J Matched placental stem cells from amniotic membrane and umbilical cord stem cells were analyzed using fluorescently-labeled antibodies and flow cytometry to identify cell surface markers that were present or absent. Markers analyzed included CD 105 (proliferation related endothelial specific marker); CD200 (marker associated with regulatory function); CD34 (expressed on endothelial cells and on hematopoietic stem cells); CDlO (stem cell/precursor cell marker); cytokeratin K (epithelial marker); CD44 (cell migration, lymphocyte homing, hematopoeisis); CD45 (lineage marker); CD 133 (marker for hematopoietic progenitor cells); CDl 17 (stem cell factor (c-Kit)); CD90 (expressed on primitive hematopoietic stem cells in normal bone marrow, cord blood and fetal liver cells); HLA ABC (pan MHC I, antigen presentation, immunogenicity); β-2 -microglobulin (associates with MHC I, antigen presentation, immunogenicity); HLA DR5DQ5DP (pan MHC II, antigen presentation, immunogenicity); and CD80/86 (co-stimulatory molecules for antigen presentation).
[0290] Flow cytometry results showed that for the placental stem cells that were tested, 93.83% of cells were CD105+, 90.76% of cells were CD200+, and 86.93% of cells were both CD 105+ and CD200+. 99.97% of cells were CD 10+, 99.15% of cells were CD34~, and 99.13% of cells were both CDlO+ and CD34". 98.71% of cells were cytokeratin positive, 99.95% of cells were CD44+, and 98.71% of cells were positive for both cytokeratin and CD44. 99.51% of cells were CD45~, 99.78% of cells were negative for CD133, and 99.39% of cells were negative for both CD45 and CDl 33. 99.31% of cells were positive for CD90, 99.7% were negative for CDl 17, and 99.01% were positive for CD90 and negative for CDl 17. 95.7% of cells were negative for both CD80 and CD86.
[0291 J Flow cytometry results for umbilical cord stem cells showed that 95.95% of cells were CD200+, 94.71% were CD105+, and 92.69% were CD105+ and CD200+. 99.93% of the cells were CDlO+, 99.99% of the cells were CD34~, and 99.6% of the cells were both CDlO+ and CD34~. 99.45% of the cells were cytokeratin positive, 99.78% of the cells were CD44+, and 99.3% of the cells were positive for both cytokeratin and CD44. 99.33% of the cells were CD45", 99.74% were CD133", and 99.15% of the cells were both CD45"~ and CD133". 99.84% of the cells were CDl 17", 98.78% of the cells were CD90+, and 98.64% of the cells were both CD90+ and CD 117".
[0292 J One phenotype (CD200+, CDlOS+, CDlO+, CD341 appears to be consistent over numerous such analyses. This phenotype is additionally positive for CD90, CD44, HLA ABC (weak), β-2-microglobulin (weak), and cytokeratin K, and negative for HLA DR5DQ5DP, CDl 17, CD 133, and CD45. 6.4 EXAMPLE 4: DETERMINATION OF ALDEHYDE DEHYDROGENASE ACTIVITY IN PLACENTAL STEM CELLS
[0293] The level of aldehyde dehydrogenase (ALDH) activity, a potential marker of stem cell engraftment capability, was determined using and ALDEFLUOR® Assay Kit from Stem Cell
Technologies, Inc. Typically, more primitive, undifferentiated stem cells demonstrate less
ALDH activity than more differentiated stem cells.
[0294] The assay uses ALDEFLUOR®, a fluorescent ALDH substrate (Aldagen, Inc.,
Durham, North Carolina). The manufacturer's protocol was followed. The dry
ALDEFLUOR® reagent is provided in a stable, inactive form. The ALDEFLUOR® was activated by dissolving the dry compound in dimethylsulfoxide (DMSO) and adding 2N HCl, and was added immediately to the cells. A control tube was also established by combing the cells with ALDEFLUOR® plus DEAB, a specific inhibitor of ALDH.
[0295] Cells analyzed included four umbilical cord stem cell lines and three placental stem cell lines from amnion-chorion plate, a bone marrow-derived mesenchymal stem cell line
(BM-MSC)7 an adipose-derived stem cell line (ADSC), a human villous trophoblast cell line
(HVT), and CD34+ stem cells purified from cord blood..
[0296] The assay proceeded as follows. Sample concentration was adjusted to IXlO6 cells
/ml with Assay buffer provided with the ALDEFLUOR® Assay Kit. 1 mL of adjusted cell suspension into experimental and control tube for each of the cell lines tested, and 5μl of
DEAB was additionally added to the control tube labeled as control.
[0297] ALDEFLUOR® substrate was activated by adding 25 μl of DMSO to the dry
ALDEFLUOR® Reagent, and let stand for 1 minute at RT. 25 μl of 2N HCL was added and mixed well. This mixture was incubated for 15 min at RT. 360 μl of ALDEFLUOR® Assay
Buffer was added to the vial and mixed. The resulting mixture was stored at 2-8°C during use.
[0298] 5μl of the activated ALDEFLUOR® reagent was added per 1 milliliter of sample to the experimental tubes, and 0.5 ml of this mixture was immediately transferred into the control tubes. The experimental and control tubes for each cell line were incubated for 30 minutes at 37 0C. After incubation, the tubes were centrifuged at 400 x g, and the supernatant was discarded. The cells in the resulting pellet were resuspended in 0.5 ml Assay Buffer and analyze by flow cytometry. Data was analyzed using FLOWJO™ software (Tree Star,
Ashland, Oregon). SSC vs FSC and SSC vs FLl plots were created in the FLOWJO™ workspace. Control and experimental data files were opened for each sample, and the appropriate gates were determined based on control samples. Positive cells were calculated as a percent ALDEFLUOR® positive out of the total number of events counted. [0299] Placental stem cell lines demonstrated ALDH activity of from about 3% to about 25% (3.53%, 8.76% and 25.26%). Umbilical cord stem cell lines demonstrated ALDH activity of from about 16% to about 20% (16.59%, 17.01%, 18.44% and 19.83%). In contrast, BM- MSC and HVT were negative and 1.5% respectively for ALDH, but the adipose derived MSC is close to 30% ALDH+. The positive control CD34+ cells purified from umbilical cord blood were, as expected, highly positive (75%) for ALDH.
6.5 EXAMPLE 5: COLLECTION OF PLACENTAL STEM CELLS BY CLOSED-CIRCUIT PERFUSION
[0300] This Example demonstrates one method of collecting placental stem cells by perfusion.
[0301] A post-partum placenta is obtained within 24 hours after birth. The umbilical cord is clamped with an umbilical cord clamp approximately 3 to 4 inches about the placental disk, and the cord is cut above the clamp. The umbilical cord is either discarded, or processed to recover, e.g., umbilical cord stem cells, and/or to process the umbilical cord membrane for the production of a biomaterial. Excess amniotic membrane and chorion is cut from the placenta, leaving approximately 1A inch around the edge of the placenta. The trimmed material is discarded.
[0302] Starting from the edge of the placental membrane, the amniotic membrane is separated from the chorion using blunt dissection with the fingers. When the amniotic membrane is entirely separated from the chorion, the amniotic membrane is cut around the base of the umbilical cord with scissors, and detached from the placental disk. The amniotic membrane can be discarded, or processed, e.g., to obtain stem cells by enzymatic digestion, or to produce, e.g., an amniotic membrane biomaterial.
[0303] The fetal side of the remaining placental material is cleaned of all visible blood clots and residual blood using sterile gauze, and is then sterilized by wiping with an iodine swab than with an alcohol swab. The umbilical cord is then clamped crosswise with a sterile hemostat beneath the umbilical cord clamp, and the hemostat is rotated away, pulling the cord over the clamp to create a fold. The cord is then partially cut below the hemostat to expose a cross-section of the cord supported by the clamp. Alternatively, the cord is clamped with a sterile hemostat. The cord is then placed on sterile gauze and held with the hemostat to provide tension. The cord is then cut straight across directly below the hemostat, and the edge of the cord near the vessel is re-clamped.
[0304] The vessels exposed as described above, usually a vein and two arteries, are identified, and opened as follows. A closed alligator clamp is advanced through the cut end of each vessel, taking care not to puncture the clamp through the vessel wall. Insertion is halted when the tip of the clamp is slightly above the base of the umbilical cord. The clamp is then slightly opened, and slowly withdrawn from the vessel to dilate the vessel. [0305] Plastic tubing, connected to a perfusion device or peristaltic pump, is inserted into each of the placental arteries. Plastic tubing, connected to a 250 mL collection bag, is inserted into the placental vein. The tubing is taped into place.
[0306] A small volume of sterile injection grade 0.9% NaCl solution to check for leaks. If no leaks are present, the pump speed is increased, and about 750 mL of the injection grade 0.9% NaCl solution is pumped through the placental vasculature. Perfusion can be aided by gently massaging the placental disk from the outer edges to the cord. When a collection bag is full, the bag is removed from the coupler connecting the tubing to the bag, and a new bag is connected to the tube.
[0307] When collection is finished, the collection bags are weighed and balanced for centrifugation. After centrifugation, each bag is placed inside a plasma extractor without disturbing the pellet of cells. The supernatant within the bags is then removed and discarded. The bag is then gently massaged to resuspend the cells in the remaining supernatant. Using a sterile 1 mL syringe, about 300-500 μL of cells is withdrawn from the collection bag, via a sampling site coupler, and transferred to a 1.5 mL centrifuge tube. The weight and volume of the remaining perfusate are determined, and 1/3 volume of hetastarch is added to the perfusate and mixed thoroughly. The number of cells per mL is determined. Red blood ceils are removed from the perfusate using a plasma extractor.
[0308] Placental cells are then immediately cultured to isolate placental stem cells, or are cryopreserved for later use.
6.6 EXAMPLE 6: DIFFERENTIATION OF PLACENTAL STEM CELLS
6.6.1 Induction Of Differentiation Into Neurons [0309] Neuronal differentiation of placental stem cells can also be accomplished as follows:
1. Placental stem cells are grown for 24 hr in preinduction medium consisting of DMEM/20% FBS and 1 mM beta-mercaptoethanol.
2. The preinduction medium is removed and cells are washed with PBS. 3. Neuronal induction medium consisting of DMEM and 1-10 mM betamercaptoethanol is added to the cells. Alternatively, induction media consisting of DMEM/2% DMSO/200 μM butylated hydroxyanisole may be used.
4. In certain embodiments, morphologic and molecular changes may occur as early as 60 minutes after exposure to serum-free media and betamercaptoethanol. RT/PCR may be used to assess the expression of e.g., nerve growth factor receptor and neurofilament heavy chain genes.
6.6.2 Induction Of Differentiation Into Adipocytes
[0310] Several cultures of placental stem cells derived from enzymatic digestion of amnion, at 50-70% confluency, were induced in medium comprising (1) DMEM/MCDB-201 with 2% FCS, 0.5% hydrocortisone, 0.5 mM isobutylmethylxanthine (IBMX), 60 μM indomethacin; or (2) DMEM/MCDB-201 with 2% FCS and 0.5% linoleic acid. Cells were examined for morphological changes; after 3-7 days, oil droplets appeared. Differentiation was also assessed by quantitative real-time PCR to examine the expression of specific genes associated with adipogenesis, i.e., PPAR-γ2, aP-2, lipoprotein lipase, and osteopontin. Two cultures of placental stem cells showed an increase of 6.5-fold and 24.3-fold in the expression of adipocyte-specifϊc genes, respectively. Four other cultures showed a moderate increase (1.5- 2.0-fold) in the expression of PPAR-γ2 after induction of adipogenesis. [0311] In another experiment, placental stem cells obtained from perfusate were cultured in DMEM/MCDB-201 (Chick fibroblast basal medium) with 2% FCS. The cells were trypsinized and centrifuged. The cells were resuspended in adipo-induction medium (AIM) 1 or 2. AIMl comprised MesenCult Basal Medium for human Mesenchymal Stem Cells (StemCell Technologies) supplemented with Mesenchymal Stem Cell Adipogenic Supplements (StemCell Technologies). AIM2 comprised DMEM/MCDB-201 with 2% FCS and LA-BSA (1%). About 1.25 x 105 placental stem cells were grown in 5 mL AIMl or AIM2 in T-25 flasks. The cells were cultured in incubators for 7-21 days. The cells developed oil droplet vacuoles in the cytoplasm, as confirmed by oil-red staining, suggesting the differentiation of the stem cells into adipocytes.
[0312] Adipogenic differentiation of placental stem cells can also be accomplished as follows:
1. Placental stem cells are grown in MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum. 2. Three cycles of induction/maintenance are used. Each cycle consists of feeding the placental stem cells with Adipogenesis Induction Medium (Cambrex) and culturing the cells for 3 days (at 37°C, 5% CO2), followed by 1-3 days of culture in Adipogenesis Maintenance Medium (Cambrex). An alternate induction medium that can be used contains 1 μM dexamethasone, 0.2 mM indomethacin, 0.01 mg/ml insulin, 0.5 mM IBMX, DMEM-high glucose, FBS5 and antibiotics.
3. After 3 complete cycles of induction/maintenance, the cells are cultured for an additional 7 days in adipogenesis maintenance medium, replacing the medium every 2-3 days.
4. A hallmark of adipogenesis is the development of multiple intracytoplasmic lipid vesicles that can be easily observed using the lipophilic stain oil red O. Expression of lipase and/or fatty acid binding protein genes is confirmed by RT/PCR in placental stem cells that have begun to differentiate into adipocytes.
6.6.3 Induction Of Differentiation Into Osteocytes
[0313] Osteogenic medium was prepared from 185 mL Cambrex Differentiation Basal Medium - Osteogenic and SingleQuots (one each of dexamethasone, 1-glutamine, ascorbate, pen/strep, MCGS5 and β-glycerophosphate). Placental stem cells from perfusate were plated, at about 3 x 103 cells per cm2 of tissue culture surface area in 0.2-0.3 mL MSCGM per cm2 tissue culture area. Typically, all cells adhered to the culture surface for 4-24 hours in MSCGM at 37°C in 5% CO2. Osteogenic differentiation was induced by replacing the medium with Osteogenic Differentiation medium. Cell morphology began to change from the typical spindle-shaped appearance of the adherent placental stem cells, to a cuboidal appearance, accompanied by mineralization. Some cells delaminated from the tissue culture surface during differentiation. [0314] Osteogenic differentiation can also be accomplished as follows:
1. Adherent cultures of placental stem cells are cultured in MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum.
2. Cultures are cultured for 24 hours in tissue culture flasks.
3. Osteogenic differentiation is induced by replacing MSCGM with Osteogenic Induction Medium (Cambrex) containing 0.1 μM dexamethasone, 0.05 mM ascorbic acid-2-phosphate, 10 mM beta glycerophosphate.
4. Cells are fed every 3-4 days for 2-3 weeks with Osteogenic Induction Medium. 5. Differentiation is assayed using a calcium-specific stain and RTVPCR for alkaline phosphatase and osteopontin gene expression.
6.6.4 Induction Of Differentiation Into Pancreatic Cells [0315] Pancreatic differentiation is accomplished as follows:
1. Placental stem cells are cultured in DMEM/20% CBS, supplemented with basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml. KnockOut Serum Replacement may be used in lieu of CBS.
2. Conditioned media from nestin-positive neuronal cell cultures is added to media at a 50/50 concentration.
3. Cells are cultured for 14-28 days, refeeding every 3-4 days.
4. Differentiation is characterized by assaying for insulin protein or insulin gene expression by RTYPCR.
6.6.5 Induction Qf Differentiation Into Cardiac Cells [0316] Myogenic (cardiogenic) differentiation is accomplished as follows:
1. Placental stem cells are cultured in DMEM/20% CBS, supplemented with retinoic acid, 1 μM; basic fibroblast growth factor, 10 ng/ml; and transforming growth factor beta-1, 2 ng/ml; and epidermal growth factor, 100 ng/ml. KnockOut Serum Replacement (Invitrogen, Carlsbad, California) may be used in lieu of CBS.
2. Alternatively, placental stem cells are cultured in DMEM/20% CBS supplemented with 50 ng/ml Cardiotropin-1 for 24 hours.
3. Alternatively, placental stem cells are maintained in protein-free media for 5-7 days, then stimulated with human myocardium extract (escalating dose analysis). Myocardium extract is produced by homogenizing I gm human myocardium in
1 % HEPES buffer supplemented with 1 % cord blood serum. The suspension is incubated for 60 minutes, then centrifuged and the supernatant collected.
4. Cells are cultured for 10-14 days, refeeding every 3-4 days.
5. Differentiation is confirmed by demonstration of cardiac actin gene expression by RT/PCR.
6.6.6 Induction Of Differentiation Into Chondrocytes 6.6.6.1 General Method
[0317] Chondrogenic differentiation of placental stem cells is generally accomplished as follows: W
1. Placental stem cells are maintained in MSCGM (Cambrex) or DMEM supplemented with 15% cord blood serum.
2. Placental stem cells are aliquoted into a sterile polypropylene tube. The cells are centrifuged (150 x g for 5 minutes), and washed twice in Incomplete Chondrogenesis Medium (Cambrex).
3. After the last wash, the cells are resuspended in Complete Chondrogenesis Medium (Cambrex) containing 0.01 μg/ml TGF-beta-3 at a concentration of 5 x 10(5) cells/ml.
4. 0.5 ml of cells is aliquoted into a 15 ml polypropylene culture tube. The cells are pelleted at 150 x g for 5 minutes. The pellet is left intact in the medium.
5. Loosely capped tubes are incubated at 37°C, 5% CO2 for 24 hours.
6. The cell pellets are fed every 2-3 days with freshly prepared complete chondrogenesis medium.
7. Pellets are maintained suspended in medium by daily agitation using a low speed vortex.
8. Chondrogenic cell pellets are harvested after 14-28 days in culture.
9. Chondrogenesis is characterized by e.g., observation of production of esoinophilic ground substance, assessing cell morphology, an/or RT/PCR confirmation of collagen 2 and/or collagen 9 gene expression and/or the production of cartilage matrix acid mucopolysaccharides, as confirmed by Alcian blue cytochemical staining.
6.6.6.2 Differentiation of Placental and Umbilical Cord Stem Cells
Into Chondrogenic Cells
[0318] The Example demonstrates the differentiation of placental stem cells into chondrogenic cells and the development of cartilage-like tissue from such cells. [0319] Cartilage is an avascular, alymphatic tissue that lacks a nerve supply. Cartilage has a low chondrocyte density (<5%), however these cells are surprisingly efficient at maintaining the extracellular matrix around them. Three main types of cartilage exist in the body: (1) articular cartilage, which facilitates joint lubrication in joints; (2) fibrocartilage, which provides shock absorption in, e.g., meniscus and intervertebral disc; and (3) elastic cartilage, which provides anatomical structure in, e.g., nose and ears. AU three types of cartilage are similar in biochemical structure.
[0320] Joint pain is a major cause of disability and provides an unmet need of relief in the area of orthopedics. Primary osteoarthritis (which can cause joint degeneration), and trauma are two common causes of pain. Approximately 9% of the U.S. population has osteoarthritis of hip or knee, and more than 2 million knee surgeries are performed yearly. Unfortunately, current treatments are more geared towards treatment of symptoms rather than repairing the cartilage. Natural repair occurs when fϊbroblast-like cells invade the area and fill it with fibrous tissue which is neither as resilient or elastic as the normal tissue, hence causing more damage. Treatment options historically included tissue grafts, subchondral drilling, or total joint replacement. More recent treatments however include CARTICEL®, an autologous chondrocyte injection; SYNVISC® and ORTHOVISC®, which are hyaluronic acid injections for temporary pain relief; and CHONDROGEN™, an injection of adult mesenchymal stem cells for meniscus repair. In general, the trend seems to be lying more towards cellular therapies and/or tissue engineered products involving chondrocytes or stem cells.
Materials and Methods.
[0321] Two placental stem cell lines, designated AC61665, P3 (passage 3) and AC63919, P5, and two umbilical cord stem cell lines, designated UC67249, P2 and UC67477, P3 were used in the studies outlined below. Human mesenchymal stem cells (MSC) were used as positive controls, and an osteosarcoma cell line, MC3T3, and human dermal fibroblasts (HDF) were used as negative controls.
[0322] Placental and umbilical cord stem cells were isolated and purified from full term human placenta by enzymatic digestion. Human MSC cells and HDF cells were purchased from Cambrex, and MC3T3 cells were purchased from American Type Culture Collection. All cell lines used were centrifuged into pellets in polypropylene centrifuge tubes at 800 RPM for 5 minutes and grown in both chondrogenic induction media (Cambrex) and non- inducing basal MSC media (Cambrex). Pellets were harvested and histologically analyzed at 7, 14, 21 and 28 days by staining for glycosaminoglycans (GAGs) with Alcian Blue, and/or for collagens with Sirius Red. Collagen type was further assessed with immunostaining. RNA analysis for cartilage-specific genes was performed at 7 and 14 days. [0323] Results
[0324] Experiment 1 : Chondrogenesis studies were designed to achieve three main objectives: (1) to demonstrate that placental and umbilical cord stem cells can differentiate and form cartilage tissue; (2) to demonstrate that placental and umbilical cord stem cells can differentiate functionally into chondrocytes; and (3) to validate results obtained with the stem cells by evaluating control cell lines. [0325] For objective 1, in a preliminary study, one placental stem cell line was cultured in chondrogenic induction medium in the form of cell pellets, either with or without bone morphogenic protein (BMP) at a final concentration of 500 ng/mL. Pellets were assessed for evidence of chondrogenic induction every week for 4 weeks. Results indicated that the pellets do increase in size over time. However, no visual differences were noted between the BMP+ and BMP" samples. Pellets were also histologically analyzed for GAG's, an indicator of cartilage tissue, by staining with Alcian Blue. BMP+ cells generally appeared more metabolically active with pale vacuoles whereas BMP" cells were smaller with dense-stained nuclei and less cytoplasm (reflects low metabolic activity). At 7 days, BMP+ cells had stained heavily blue, while BMP" had stained only faintly. By 28 days of induction, both BMP+ and BMP" cells were roughly equivalently stained with Alcian Blue. Overall, cell density decreased over time, and matrix overtook the pellet. In contrast, the MC3T3 negative cell line did not demonstrate any presence of GAG when stained with Alcian Blue. [0326J Experiment 2: Based on the results of Experiment 1, a more detailed study was designed to assess the chondrogenic differentiation potential of two placental stem cell and two umbilical cord stem cell lines. In addition to the Alcian Blue histology, cells were also stained with Sinus Red, which is specific for type II collagen. Multiple pellets were made for each cell line, with and without induction media.
[0327] The pelleted, cultured cell lines were first assessed by gross observation for macroscopic generation of cartilage. Overall, the stem cell lines were observed to make pellets as early as day 1. These pellets grew over time and formed a tough matrix, appearing white, shining and cartilage-like, and became mechanically tough. By visual inspection, pellets from placental stem cells or umbilical cord stem cells were much larger than the MSC controls. Control pellets in non-induction media started to fall apart by Day 11, and were much smaller at 28 days than pellets developed by cells cultured in chondrogenic induction medium. Visually, there were no differences between pellets formed by placental stem cells or umbilical cord. However, the UC67249 stem cell line, which was initiated in dexamethasone-free media, formed larger pellets. Negative control MC3T3 cells did not form pellets; however, HDFs did form pellets.
[0328] Representative pellets from all test groups were then subjected to histological analysis for GAG's and collagen. Generally, pellets formed by the stem cells under inducing conditions were much larger and stayed intact better than pellets formed under non-inducing conditions. Pellets formed under inducing conditions showed production of GAGs and increasing collagen content over time, and as early as seven days, while pellets formed under non-inducing conditions showed little to no collagen production, as evidenced by weak Alcian Blue staining. In general, the placental stem cells and umbilical cord stem cells appeared, by visual inspection, to produce tougher, larger pellets, and appeared to be producing more collagen over time, than the hMSCs. Moreover, over the course of the study, the collagen appeared to thicken, and the collagen type appeared to change, as evidenced by changes in the fiber colors under polarized light (colors correlate to fiber thickness which may be indicative of collagen type). Non-induced placental stem cells produced much less type II collagen, if any, compared to the induced stem cells. Over the 28-day period, cell density decreased as matrix production increased, a characteristic of cartilage tissue. [0329] These studies confirm that placental and umbilical cord stem cells can be differentiated along a chondrogenic pathway, and can easily be induced to form cartilage tissue. Initial observations indicate that such stem cells are preferable to MSCs for the formation of cartilage tissue.
6.7 EXAMPLE 7: HANGING DROP CULTURE OF PLACENTAL STEM CELLS
[0330] Placental adherent stem cells in culture are trypsinized at 37°C for about 5 minutes, and loosened from the culture dish by tapping. 10% FBS is added to the culture to stop trypsinization. The cells are diluted to about 1 x 104 cells per mL in about 5 mL of medium. Drops (either a single drop or drops from a multi-channel micropipette are placed on the inside of the lid of a 100 mL Petri dish. The lid is carefully inverted and placed on top of the bottom of the dish, which contains about 25 ml of sterile PBS to maintain the moisture content in the dish atmosphere. Cells are grown for 6-7 days.
6.8 EXAMPLE 8: PLACENTAL TISSUE DIGESTION TO OBTAIN PLACENTAL STEM CELLS
[0331] This Example demonstrates a scaled up isolation of placental stem cells by enzymatic digestion.
[0332] Approximately 10 grams of placental tissue (amnion and chorion) is obtained, macerated, and digested using equal volumes of collagenase A (1 mg/ml) (Sigma) and Trypsin-EDTA (0.25%) (Gibco-BRL) in a total volume of about 30 ml for about 30 minutes at 37°C. Cells liberated by the digestion are washed 3X with culture medium, distributed into four T-225 flasks and cultured as described in Example 1. Placental stem cell yield is between about 4 x 108 and 5 x 10s cells per 1Og starting material. Cells, characterized at passage 3, are predominantly CDlO+, CD90+, CD105+, CD200+, CD34" and CD45~. 6.9 EXAMPLE 9: PRODUCTION OF CRYOPRESERVED STEM CELL PRODUCT AND STEM CELL BANK
[0333J This Example demonstrates the isolation of placental stem cell and the production of a frozen stem cell-based product.
[0334] Summary: Placental tissue is dissected and digested, followed by primary and expansion cultures to achieve an expanded cell product that produces many cell doses. Cells are stored in a two-tiered cell bank and are distributed as a frozen cell product. All cell doses derived from a single donor placenta are defined as a lot, and one placenta lot is processed at a time using sterile technique in a dedicated room and Class 100 laminar flow hood. The cell product is defined as being CD105+, CD200+ 5 CDlO+, and CD34~, having a normal karyotype and no or substantially no maternal cell content.
6.9.1 Obtaining Stem Cells
[0335] Tissue Dissection and Digestion: A placenta is obtained less than 24 hours after expulsion. Placental tissue is obtained from amnion, a combination of amnion and chorion, or chorion. The tissue is minced into small pieces, about 1 mm in size. Minced tissue is digested in lmg/ml Collagenase IA for 1 hour at 37°C followed by Trypsin-EDTA for 30 minutes at 37°C. After three washes in 5% FBS in PBS, the tissue is resuspended in culture medium.
[0336] Primary Culture: The purpose of primary culture is to establish cells from digested placental tissue. The digested tissue is suspended in culture medium and placed into Corning T-flasks, which are incubated in a humidified chamber maintained at 37°C with 5% CO2. Half of the medium is replenished after 5 days of culture. High-density colonies of cells form by 2 weeks of culture. Colonies are harvested with Trypsin-EDTA, which is then quenched with 2% FBS in PBS. Cells are centrifuged and resuspended in culture medium for seeding expansion cultures. These cells are defined as Passage 0 cells having doubled 0 times. [0337] Expansion Culture: Cells harvested from primary culture, harvested from expansion culture, or thawed from the cell bank are used to seed expansion cultures. Cell Factories (NUNC™) are treated with 5% CO2 in air at 50 ml/min/tray for 10 min through a sterile filter and warmed in a humidified incubator maintained at 37°C with 5% COz. Cell seeds are counted on a hemacytometer with trypan blue, and cell number, viability, passage number, and the cumulative number of doublings are recorded. Cells are suspended in culture medium to about 2.3 X lO4 cells/ml and 110 ml/tray are seeded in the Cell Factories. After 3- 4 days and again at 5-6 days of culture, culture medium is removed and replaced with fresh medium, followed by another treatment with 5% CO2 in air. When cells reach approximately 105 cells/cm2, cells are harvested with Trypsin-EDTA, followed by quenching with 2% FBS in PBS. Cell are then centrifuged and resuspended in culture medium. [0338] Cryopreservation: Cells to be frozen down are harvested from culture with Trypsin- EDTA, quenched with 2% FBS in PBS, and counted on a hemacytometer. After centrifugation. cells are resuspended with 10% DMSO in FBS to a concentration of about 1 million cells/ml for cells to be used for assembly of a cell bank, and 10 million cells/ml for individual frozen cell doses. The cell solution is transferred to a freezing container, which is placed in an isopropyl alcohol bath in a — 800C freezer. The following day, cells are transferred to liquid nitrogen.
6.9.2 Design OfA Stem Cell Bank
[0339J A "lot" is defined as all cell doses derived from a single donor placenta. Cells maintained normal growth, karyotype, and cell surface maker phenotype for over 8 passages and 30 doublings during expansion culture. Given this limitation, doses comprise cells from 5 passages and about 20 doublings. To generate a supply of equivalent cells, a single lot is expanded in culture and is stored in a two-tiered cell bank and frozen doses. In particular, cells harvested from the primary culture, which are defined as Passage 0 cells having undergone 0 doublings, are used to initiate an expansion culture. After the first passage, approximately 4 doublings occur, and cells are frozen in a Master Cell Bank (MCB). Vials from the MCB are used to seed additional expansion cultures. After two additional passages of cells thawed from the MCB, cells are frozen down in a Working Cell Bank (WCB), approximately 12 cumulative doublings. Vials from the WCB are used to seed an expansion culture for another 2 passages, resulting in Passage 5 cells at approximately 20 doublings that are frozen down into individual doses.
6.9.3 Thawing Cells For Culture
[0340] Frozen containers of cells are placed into a sealed plastic bag and immersed in a 37°C water bath. Containers are gently swirled until all of the contents are melted except for a small piece of ice. Containers are removed from the sealed plastic bag and a 1OX volume of culture medium is slowly added to the cells with gentle mixing. A sample is counted on the hemacytometer and seeded into expansion cultures. 6.9.4 Thawing Cells for Injection
[0341] Frozen containers of cells are transferred to the administration site in a dry nitrogen shipper. Prior to administration, containers are placed into a sealed plastic bag and immersed in a 370C water bath. Containers are gently swirled until all of the contents are melted except for a small piece of ice. Containers are removed from the sealed plastic bag and an equal volume of 2.5% HS A/5% Dextran is added. Cells are injected with no further washing.
6.9.5 Testing and Specifications
[0342] A maternal blood sample accompanies all donor placentas. The sample is screened for Hepatitis B core antibody and surface antigen, Hepatitis C Virus antibody and nucleic acid, and HIV I and II antibody and nucleic acid. Placental processing and primary culture begins prior to the receipt of test results, but continues only for placentas associated with maternal blood samples testing negative for all viruses. A lot is rejected if the donor tests positive for any pathogen. In addition, the tests described in Table 3 are performed on the MCB, the WCB, and a sample of the cell dose material derived from a vial of the WCB. A lot is released only when all specifications are met. Table 3: Cell testing and specifications
Figure imgf000095_0001
*For the product designed to be 40 ml of frozen cells/dose and a maximum of 5 EU/ml, the cell product is below the upper limit of 5EU/kg/dose for recipients over 40kg in body weight.
6.9.6 Surface Marker Phenotype Analysis
[0343] Cells are placed in 1% paraformaldehyde (PFA) in PBS for 20 minutes and stored in a refrigerator until stained (up to a week). Cells are washed with 2% FBS, 0.05% sodium azide in PBS (Staining Buffer) and then resuspended in staining buffer. Cells are stained with the following antibody conjugates: CD105-FITC, CD200-PE, CD34-PECy7, CDlO-APC. Cells are also stained with isotype controls. After 30 minute incubation, the cells are washed and resuspended with Staining Buffer, followed by analysis on a flow cytometer. Cells having an increased fluorescence compared to isotype controls are counted as positive for a marker.
6.10 EXAMPLE 10: IDENTIFICATION OF PLACENTAL STEM CELL- SPECIFIC GENES
[0344] Gene expression patterns from placental stem cells from amnion-chorion (AC) and umbilical cord (UC) were compared to gene expression patterns of multipotent bone marrow- derived mesenchymal stem cells (BM) and dermal fibroblasts (DF), the latter of which is considered to be terminally differentiated. Cells were grown for a single passage, an intermediate number of passages, and large number of passages (including until senescence). Results indicate that the number of population doublings has a major impact on gene expression. A set of genes was identified that are up-regulated in AC and UC, and either down-regulated or absent in BM and DF, and that are expressed independent of passage number. This set of placental stem cell- or umbilical cord stem cell-specific genes encodes a number of cytoskeleton and cell-to-cell adhesion proteins associated with epithelial cells and an immunoglobulin-like surface protein, CD200, implicated in maternal-fetal immune tolerance. Placental stem cells and umbilical cord stem cells will be referred to collectively hereinafter in this Example as AC/UC stem cells.
6.10.1 Methods and Materials
6.10.1.1 Cells and Cell Culture
[0345] BM (Cat* PT-2501) and DF (Cat# CC-2511) were purchased from Cambrex. AC and UC originated from passage 0 tissue culture flasks. AC and UC in the flasks were obtained by digestion from a donor placenta designated 2063919. T-75 culture flasks were seeded at 6000 cells/cm2 and cells were passaged when they became confluent. Population doublings W
were estimated from trypan blue cell counts. Cultures were assayed for gene expression after 3, 11-14, and 24-3 S population doublings.
6.10.1.2 RNA1 Microarrays, and Analysis
[0346] Cells were lysed directly in their tissue culture flasks, with the exception of one culture that was trypsinized prior to lysis. Total RNA was isolated with the RNeasy kit from QIAGEN. RNA integrity and concentrations were determined with an Agilent 2100 Bioanalyzer. Ten micrograms of total RNA from each culture were hybridized on an Affymetrix GENECHIP® platform. Total RNA was converted to labeled cRNAs and hybridized to oligonucleotide Human Genome Ul 33 A 2.0 arrays according to the manufacture's methods. Image files were processed with the Affymetrix MAS 5.0 software, and normalized and analyzed with Agilent GeneSpring 7.3 software.
6.10.2 Results
6.10.2.1 Selection of BM-MSC, AC/UCStem Cell, and DF
Culture Time-Points for Microarray Analyses
[0347J To establish a gene expression pattern unique to AC/UC stem cells, two stem cell lines, AC(6) and UC(6), were cultured in parallel with BM-MSC and DF. To maximize identifying a gene expression profile attributable to cellular origin and minimize exogenous influences all cells were grown in the same medium, seeded, and sub-cultured using the same criteria. Cells were harvested after 3 population doublings, 11-14 doublings, or 35 doublings or senescence, whichever came first. Genes whose expression in AC/UC stem cells are unchanged by time-in-culture and are up-regulated relative to BM and DF are candidates for AC/UC stem cell-specific genes.
[0348] FIG. 10 shows growth profiles for the four cell lines in the study; circles indicate which cultures were harvested for RNA isolation. In total twelve samples were collected. BM, AC(6), and UC(6) were harvested after three population doublings; these samples were regarded as being in culture for a "short" period of time. A short-term DF sample was not collected. Intermediate length cultures, 11 to 14 doublings, were collected for all cell types. Long-term cultures were collected from all cell lines at about 35 population doublings or just prior to senescence, whichever came first. Senescence occurred before 15 doublings for BM and at 25 doublings for DF. The purchased BM and DF cells were expanded many times prior to gene analysis, and cannot be considered early-stage. However, operationally, BM grown for three doublings (BM-03) are deemed a short-term culture. Likewise, BM-11 is operationally referred to as an intermediate length culture, but because senescence occurred at 14 doublings, BM-11 is most likely a long-term culture biologically.
6.10.2.2 Hierarchical Clustering Shows Relatedness between BM, ACAJC Stem Cells, and DF
[0349] Microarray analysis identifies patterns of gene expression, and hierarchical clustering
(HC) attempts to find similarities in the context of two dimensions - genes in the first dimension and different conditions (different RNA samples) in the second. The GeneChips used in this experiment contained over 22,000 probe sets (referred to as the "all genes list"), but many of these sets interrogate genes that are not expressed in any condition. To reduce the all genes list, genes not expressed or expressed at low levels (raw values below 250) in all samples were eliminated to yield a list of 8,215 genes.
6.10.2.3 Gene Expression Analysis Using the Line Graph View
[0350] Gene expression patterns of the 8215 genes were displayed using the line graph view in GeneSpring (FIG. 11). The x-axis shows the twelve experimental conditions and the y- axis shows the normalized probe set expression values on a log scale. The y-axis covers a 10,000-fold range, and genes that are not expressed or expressed at very low levels are set to a value of 0.01. By default the normalized value is set to 1. Each line represents a single gene (actually a probe set, some genes have multiple probe sets) and runs across all twelve conditions as a single color. Colors depict relative expression levels, as described for the heatmaps, but the coloring pattern is determined by selecting one condition. AC-03 is the selected condition in FIG. 11. Genes up-regulated relative to the normalized value are displayed by the software as red, and those that are down-regulated, are displayed as blue. The obvious upward and downward pointing spikes in AC-03 through UC-11 indicate that many genes are differentially expressed across these conditions. The striking similarity in the color patterns between AC-03 and UC-03 show that many of the same genes are up or down- regulated in these two samples. Horizontal line segments indicate that a gene's expression level is unchanged across a number of conditions. This is most notable by comparing UC-36, UC-38, and UC-38-T. There are no obvious spikes, but there is a subtle trend in that a number of red lines between UC-36 and UC-38-T are below the normalized value of 1. This indicates that these genes, which are up-regulated in AC-03 and UC-03, are down-regulated in the later cultures. The fact that the expression patterns between UC-38 and UC-38-T are so similar indicates that trypsinizing cells just prior to RNA isolation has little effect on gene expression. [0351J In addition to the computationally intensive HC method, by visual inspection the two BM samples are more similar to each other than to the other conditions. The same is true for the two DF cultures. And despite the large number of differentially expressed genes present in the BM and DF samples, the general appearance suggests that two BMs and the two DFs are more similar to each other than to AC/UC stem cells. This is confirmed by the HC results described above.
[0352] When the above process is applied using AC-1 1 as the selected condition, it is clear that AC-11 and UC-11 share many of the same differentially expressed genes, but the total number of genes in common between these two conditions appears less than the number of differentially expressed genes shared by AC-03 and UC-03. FIG. 12 shows genes differentially over-expressed, by six-fold or more relative to the baseline, in AC-03. The majority of genes up-regulated in AC-03 are also up-regulated in UC-03, and more divergent in BM and DF.
6.10.2.4 Filtering Methods Used to Identify AC/UC Stem Cell-
Specific Genes
[0353] Genes that remain constant across all AC/UC samples, and are down-regulated in BM and DF, are considered ACAJC stem cell-specific. Two filtering methods were combined to create a list of 58 AC/UC stem cell-specific genes (Table 4). Table 4: 58 Placental stem cell or Umbilical cord stem cell-specific genes
Symbol Gene Biological Process,
Description, and Additional Annotation
ACTG2 actin, gamma 2, smooth muscle development, cytoskeleton, muscle, enteric expressed in umbilical cord artery and prostate epithelia
ADARBl adenosine deaminase, RNA- RNA processing, central nervous system speciflc, Bl (REDl homolog development rat)
AMIGO2 amphoterin induced gene 2 homophilic and heterophils cell adhesion, adhesion molecule with Ig like domain 2
ARTS-I type 1 tumor necrosis factor proteolysis, antigen processing, receptor shedding angiogenesis, expressed in placenta aminopeptidase regulator
B4GALT6 UDP-Gal:betaGlcNAc beta 1,4- carbohydrate metabolism, integral to galactosyltransferase, membrane, may function in intercellular polypeptide 6 recognition and/or adhesion
BCHE butyrylcholinesterase cholinesterase activity, serine esterase activity, hydrolase activity
Cl lorfP chromosome 11 open reading hypothetical protein, p53-like transcription factor, expressed in retinal pigment
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
[0354] First, 58 genes were identified by selecting those genes over-expressed ≥ three- fold in at least seven of eight AC/UC stem cell conditions relative to all BM and DF samples (FIG. 13). Filtering on eight of the eight AC/UC stem cell conditions yielded a similar list. The second filtering method used "absent" and "present" calls provided by the Affymetrix MAS 5.0 software. A list was created by identifying genes absent in all BM and DF conditions and present in AC-03, AC-11, UC-03, and UC-11. Gene calls in the later AC/UC stem cell conditions were not stipulated.
[0355] The two lists overlapped significantly and were combined. The combined list was trimmed further by eliminating (1) several genes expressed at very low levels in most or all AC/UC stem cell conditions, and (2) genes carried on the Y chromosome. AC and UC cells used in this study were confirmed to be male by FISH analysis, and the BM and DF were derived from a female donor. The resulting list of 46 AC/UC stem cell-specific genes is shown in Table 5.
Table 5. AC/UC-Specifϊc Genes Listed by Ontology
Figure imgf000102_0002
Figure imgf000103_0001
[0356] This list of 46 genes encodes a collection of proteins presenting a number of ontology groups. The most highly represented group, cell adhesion, contains eight genes. No genes encode proteins involved in DNA replication or cell division. Sixteen genes with specific references to epithelia are also listed.
6.10.3 Discussion
[0357] An expression pattern specific to placental stem cells, and distinguishable from bone marrow-derived mesenchymal cells, was identified. Operationally, this pattern includes 46 genes that are over expressed in all placental stem cell samples relative to all BM and DF samples.
[0358] The experimental design compared cells cultured for short, medium, and long periods of time in culture. For AC and UC cells, each culture period has a characteristic set of differentially expressed genes. During the short-term or early phase (AC-03 and UC-03) two hundred up-regulated genes regress to the mean after eight population doublings. Without being bound by theory, it is likely that this early stage gene expression pattern resembles the expression profile of AC and UC while in the natural placental environment. In the placenta these cells are not actively dividing, they are metabolizing nutrients, signaling between themselves, and securing their location by remodeling the extracellular surroundings. [0359] Gene expression by the intermediate length cultures is defined by rapid cell division and genes differentially expressed at this time are quite different from those differentially expressed during the early phase. Many of the genes up-regulated in AC-1 1 and UC-1 1, along with BM-03 and DF- 14, are involved in chromosome replication and cell division. Based on gene expression, BM-03 appears biologically to be a mid-term culture. In this middle stage cell type-specific gene expression is overshadowed by cellular proliferation. In addition, almost every gene over expressed in the short-term AC or UC cultures is down- regulated in the middle and later stage conditions. 143 genes were up-regulated > five-fold during this highly proliferative phase, constituting approximately 1.7% of the expressed genes.
[0360] The long-term cultures represent the final or senescent phase. In this phase, cells have exhausted their ability to divide, and, especially for AC and UC, the absolute number of differentially expressed genes is noticeably reduced. This may be the result of cells being fully adapted to their culture environment and a consequently reduced burden to biosynthesize. Surprisingly, late BM and DF cultures do not display this same behavior; a large number of genes are differentially expressed in BM-11 and DF-24 relative to AC and UC and the normalized value of 1. AC and UC are distinguishable from BM and DF most notably in the long-term cultures.
[0361] The placental stem cell-specific gene list described here is diverse. COL4A1 and COL4A2 are coordinately regulated, and KRT 18 and KRT8 also appear to be co-expressed. Eight of the genes encode proteins involved in cell to cell contact, three of which (DSC3, DSG2, and PKP2) are localized to desmosomes, intercellular contact points anchored to intermediate filament cytoskeleton proteins such as keratin 18 and keratin 8. Tight cell-to- cell contact is characteristic of epithelial and endothelial cells and not typically associated with fibroblasts. Table 3 lists 16 genes, of the 46 total, characteristic to epithelial cells. Placental stem cells are generally described as fibroblast-like small spindle-shaped cells. This morphology is typically distinct from BM and DF, especially at lower cell densities. Also of note is the expression pattern of CD200, which is present in AC/UC stem cell and absent in all BM and DF samples. Moreover, CD200 has been shown to be associated with immune tolerance in the placenta during fetal development (see, e.g., Clark et ah, Am. J. Reprod. Immunol. 50(3):l 87-195 (2003)).
[0362] This subset of genes of 46 genes constitutes a set of molecular biomarkers that distinguishes AC/UC stem cells from bone marrow-derived mesenchymal stem cells or fibroblasts.

Claims

WHAT IS CLAIMED:
1. A method of producing a cell population comprising identifying placental cells that adhere to a substrate and: express CD200 and HLA-G; express CD73, CD 105, and CD200; express CD200 and OCT-4; express CD73, CD 105 and HLA-G; express CD73 and CD 105, and facilitate the formation of one or more embryoid-like bodies in a population of placental cells when said population is cultured under conditions that allow for the formation of embryoid-like bodies; or express OCT-4, and (c) facilitate the formation of one or more embryoid-like bodies in a population of placental cells when said population is cultured under conditions that allow for the formation of embryoid-like bodies; and isolating said cells from other cells to form a cell population.
2. The method of claim 1, wherein said substrate comprises fibronectin.
3. The method of claim 1, wherein said identifying is accomplished using an antibody.
4. The method of claim 1, wherein said identifying is accomplished using flow cytometry.
5. The method of claim 1, wherein said isolating is accomplished using magnetic beads.
6. The method of claim 1. wherein said isolating is accomplished by fluorescence-activated cell sorting.
7. The method of claim I5 wherein said cell population is expanded after said isolating.
8. A method of producing a stem cell line, comprising transforming a stem cell with a DNA sequence that encodes a growth-promoting protein; and exposing said stem cell to conditions that promote production of said growth-promoting protein.
9. The method of claim S, wherein said growth-promoting protein is v-myc, N- myc, c-myc, p53, SV40 large T antigen, polyoma large T antigen, EIa adenovirus or human papillomavirus E7 protein.
10. The method of claim 8, wherein said DNA sequence is regulatable.
11. The method of claim 8, wherein said DNA sequence is regulatable by tetracycline.
12. The method of claim 8, wherein said growth-promoting protein has a regulatable activity.
13. The method of claim 8, wherein said growth-promoting protein is a temperature-sensitive mutant.
14. A method of producing a stem cell population, comprising identifying adherent placental stem cells that express one or more genes, wherein said genes are ACTG2, ADARBl, AMIGO2, ATRS-I, B4GALT6, BCHE, Cl Iorf9, CD200, COL4A1, COL4A2, CP A4, DMD, DSC3, DSG2, ELOVL2, F2RL1, FLJ10781, GATA6, GPR126, GPRC5B, ICAMl, IER3, IGFBP7, ILIA, IL6, IL18, KRT18, KRT8, LIPG, LRAP, MATN2, MEST, NFE2L3, NUAKl, PCDH7, PDLIM3, PJP2, RTNl5 SERPINB9, ST3GAL6, ST6GALNAC5, SLC12A8, TCF21, TGFB2, VTN, and ZC3H12A, at a detectably higher level than a bone marrow-derived mesenchymal stem cell that has undergone the same number of passages in culture as said placental stem cell, and isolating said placental cells.
15. The method of claim 14, wherein said adherent placental cell expresses five or more of said genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell that has undergone the same number of passages in culture as said placental stem cell.
16. The method of claim 14, wherein said adherent placental cell expresses ten or more of said genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell that has undergone the same number of passages in culture as said placental stem cell.
17. The method of claim 14, wherein said adherent placental cell expresses twenty or more of said genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell that has undergone the same number of passages in culture as said placental stem cell.
18. The method of claim 14, wherein said adherent placental cell expresses each of said genes at a detectably higher level than a bone marrow-derived mesenchymal stem cell that has undergone the same number of passages in culture as said placental stem cell.
19. A method of making a placental stem cell bank, comprising: expanding primary culture placental stem cells from a human post-partum placenta for a first plurality of population doublings; cryopreserving said placental stem cells to form a Master Cell Bank; expanding a plurality of placental stem cells from the Master Cell Bank for a second plurality of population doublings; cryopreserving said placental stem cells to form a Working Cell Bank; expanding a plurality of placental stem cells from the Working Cell Bank for a third plurality of population doublings; and cryopreserving said placental stem cells in individual doses, wherein said individual doses collectively compose a placental stem cell bank.
20. The method of claim 19, wherein the total number of population doublings is about 20.
21. The method of claim 19, wherein said first plurality of population doublings is about four population doublings; said second plurality of population doublings is about eight population doublings; and said third plurality of population doublings is about eight population doublings.
22. The method of claim 19, wherein said primary culture placental stem cells comprise placental stem cells from placental perfusate.
23. The method of claim 19, wherein said primary culture placental stem cells comprise placental stem cells from digested placental tissue.
24. The method of claim 19, wherein said primary culture placental stem cells comprise placental stem cells from placental perfusate and from digested placental tissue.
25. The method of claim 19, wherein all of said placental stem cells in said placental stem cell primary culture are from the same placenta.
26. The method of claim 19, further comprising the step of selecting CD200+ or HLA-G+ placental stem cells from said plurality of said placental stem cells from said Working Cell Bank to form individual doses.
27. The method of claim 19, wherein said individual doses comprise from about
104 to about 105 placental stem cells.
28. The method of claim 19, wherein said individual doses comprise from about
105 to about 106 placental stem ceils.
29. The method of claim 19, wherein said individual doses comprise from about
106 to about 107 placental stem cells.
30. The method of claim 19, wherein said individual doses comprise from about
107 to about 108 placental stem cells.
31. A composition comprising conditioned medium, wherein said medium is conditioned by placental stem cells that are:
CD200+ and HLA-G+; CD73+, CD105+, and CD200+; CD200+ and OCT-4+;
CD73+, CDl 05+ and HLA-G+;
CD73+ and CD105+ and facilitate the formation of one or more embryo id-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow the formation of an embryoid-like body; or
OCT-4+ and facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies.
32. The composition of claim 31 , wherein said placental stem cells have been grown in said medium for at least one day.
33. The composition of claim 31, wherein said placental stem cells have been grown in said medium for at least three days.
34. A composition comprising a population of placental stem cells that are: CD200+ and HLA-G+;
CD73+, CD105\ and CD200+;
CD200+ and OCT-4+;
CD73+, CDl 05+ and HLA-G+;
CD73+ and CD105+ and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said population of placental stem cell when said population of placental cells is cultured under conditions that allow the formation of an embryoid-like body; or
OCT-4+ and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said population of placental stem cell when said population of placental cells is cultured under conditions that allow the formation of an embryoid-like body; or a combination of the foregoing; and a stem cell that is not obtained from a placenta, wherein at least 70% of said placental stem cells are non-maternal in origin.
35. The composition of claim 34, wherein said stem cell not obtained from a placenta is an embryonic stem cell.
36. The composition of claim 34, wherein said stem cell not obtained from a placenta is a mesenchymal stem cell.
37. The composition of claim 34, wherein said stem cell not obtained from a placenta is a bone marrow-derived stem cell.
38. The composition of claim 34, wherein said stem cell not obtained from a placenta is a hematopoietic progenitor cell.
39. The composition of claim 34, wherein said stem cell not obtained from a placenta is a somatic stem cell.
40. The composition of claim 39, wherein said somatic stem cell is a neural stem cell, a hepatic stem cell, a pancreatic stem cell, an endothelial stem cell, a cardiac stem cell, or a muscle stem cell.
41. A method of isolating a population of placental stem cells, comprising culturing a portion of a placenta for a time sufficient for a plurality of placental stem cells to proliferate out of said tissue, and isolating said placental stem cells from said tissue.
42. The method of claim 41 , wherein said tissue is amniotic membrane, chorion, a combination of amnion and chorion, or a portion of any thereof, or a combination of any of the foregoing.
43. An isolated adherent placental stem cell, wherein said stem cell is: CD200+ and HLA-G+;
CD73+, CD105+, and CD200+;
CD200+ and OCT-4+;
CD73+, CD105+ and HLA-G+;
CD73+ and CD105+ and facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow the formation of an embryoid-like body; or
OCT-4 and facilitates the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies, or a combination thereof; and wherein said placental stem cell is non-maternal in origin.
44. A population of isolated adherent placental stem cells, wherein said stem cells are:
CD200+ and HLA-G+; CD73+, CD105+, and CD200+; CD200+ and OCT-4+; CD73+, CD105+ and HLA-G+; CD73+ and CDlOS+ and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising said stem cell when said population is cultured under conditions that allow the formation of an embryoid-like body; or
OCT-4+ and facilitate the formation of one or more embryoid-like bodies in a population of placental cells comprising the stem cell when said population is cultured under conditions that allow formation of embryoid-like bodies; and wherein at least 70% of said placental stem cells are non-maternal in origin.
45. The population of claim 44, wherein at least 90% of said placental stem cells are non-maternal in origin.
46. The population of claim 44, wherein at least 99% of said placental stem cells are non-maternal in origin.
47. A composition comprising the isolated placental stem cell of claim 43.
48. A composition comprising the isolated placental stem cell population of claim 44.
49. The composition of claim 47 that is in a form suitable for intravenous administration.
50. The composition of claim 48 that is in a form suitable for intravenous administration.
51. The composition of claim 47 wherein said composition is in a container suitable for intravenous administration of said composition.
52. The composition of claim 48 wherein said composition is in a container suitable for intravenous administration of said composition.
53. A method of collecting placental stem cells, comprising: perfusing a mammalian placenta that has been drained of cord blood and perfused to remove residual blood; perfusing said placenta with a perfusion solution through placental vasculature; collecting said perfusion solution only from said vasculature, wherein said perfusion solution after perfusion comprises a population of cells that comprises placental stem cells, wherein at least 95% of said placental stem cells are fetal in origin; and isolating a plurality of said placental stem cells from said population of cells.
54. The method of claim 53, wherein the perfusion solution is passed through the umbilical vein and collected from the umbilical arteries.
55. The method of claim 53, wherein the perfusion solution is passed through the umbilical arteries and collected from the umbilical vein.
56. An isolated population of adherent placental stem cells collected by the method of claim 53, wherein at least 95% of said isolated population of placental stem cells is fetal in origin.
57. The isolated population of claim 51 , wherein greater than 99% of said placental stem cells are fetal in origin.
58. The isolated population of claim 51, wherein the placental stem cells are at least 50% of said population of placental cells.
59. The isolated population of claim 51 , wherein the placental stem cells are at least 95% of said population of placental cells.
PCT/US2006/049491 2005-12-29 2006-12-28 Placental stem cell populations WO2007079183A2 (en)

Priority Applications (36)

Application Number Priority Date Filing Date Title
ES06848281.9T ES2549111T3 (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020187026653A KR20180105266A (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020197025129A KR20190104428A (en) 2005-12-29 2006-12-28 Placental stem cell populations
EP12150934.3A EP2471907B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
EP12150924.4A EP2471905B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
EP12150919.4A EP2471904B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
PL12150934T PL2471907T3 (en) 2005-12-29 2006-12-28 Placental stem cell populations
PL12150928T PL2471906T3 (en) 2005-12-29 2006-12-28 Placental stem cell populations
EP18196002.2A EP3486314B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020227015349A KR20220063304A (en) 2005-12-29 2006-12-28 Placental stem cell populations
EP06848281.9A EP1976977B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020237019606A KR20230091193A (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020217031368A KR20210122908A (en) 2005-12-29 2006-12-28 Placental stem cell populations
PL12150919T PL2471904T3 (en) 2005-12-29 2006-12-28 Placental stem cell populations
NZ595786A NZ595786A (en) 2005-12-29 2006-12-28 Placental stem cell populations
PL06848281T PL1976977T3 (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020207030297A KR20200123283A (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020177008335A KR101900809B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
EP12150928.5A EP2471906B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
AU2006332678A AU2006332678B2 (en) 2005-12-29 2006-12-28 Placental stem cell populations
KR1020157020367A KR101722577B1 (en) 2005-12-29 2006-12-28 Placental stem cell populations
PL12150924T PL2471905T3 (en) 2005-12-29 2006-12-28 Placental stem cell populations
CA2635253A CA2635253C (en) 2005-12-29 2006-12-28 Placental stem cell populations
JP2008548745A JP5550235B2 (en) 2005-12-29 2006-12-28 Placental stem cell population
NZ569562A NZ569562A (en) 2005-12-29 2006-12-28 Placental stem cell populations, CD200
CN200680053575.3A CN101395266B (en) 2005-12-29 2006-12-28 placental stem cell populations
IL192457A IL192457A0 (en) 2005-12-29 2008-06-26 Placental stem cell populations
IL243876A IL243876B (en) 2005-12-29 2012-11-20 Adherent cd-90+ placental stem cell populations
IL223156A IL223156A0 (en) 2005-12-29 2012-11-20 Placental stem cell populations
IL223194A IL223194A (en) 2005-12-29 2012-11-22 Adherent placental stem cell populations
AU2014201181A AU2014201181B2 (en) 2005-12-29 2014-03-04 Placental stem cell populations
IL243876A IL243876A0 (en) 2005-12-29 2016-01-31 Placental stem cell populations
AU2017248523A AU2017248523A1 (en) 2005-12-29 2017-10-19 Placental stem cell populations
IL266958A IL266958A (en) 2005-12-29 2019-05-28 Placental stem cell populations
AU2019268061A AU2019268061A1 (en) 2005-12-29 2019-11-19 Placental stem cell populations
AU2022201955A AU2022201955A1 (en) 2005-12-29 2022-03-22 Placental stem cell populations

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US75496805P 2005-12-29 2005-12-29
US60/754,968 2005-12-29
US84664106P 2006-09-22 2006-09-22
US60/846,641 2006-09-22

Publications (2)

Publication Number Publication Date
WO2007079183A2 true WO2007079183A2 (en) 2007-07-12
WO2007079183A3 WO2007079183A3 (en) 2007-11-15

Family

ID=38069162

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/049491 WO2007079183A2 (en) 2005-12-29 2006-12-28 Placental stem cell populations

Country Status (18)

Country Link
US (7) US20080032401A1 (en)
EP (7) EP1976977B1 (en)
JP (2) JP5550235B2 (en)
KR (9) KR101900809B1 (en)
CN (5) CN103060263B (en)
AU (1) AU2006332678B2 (en)
CA (2) CA2635253C (en)
DK (5) DK2471907T3 (en)
ES (6) ES2703502T3 (en)
HK (1) HK1184817A1 (en)
IL (6) IL192457A0 (en)
MX (4) MX348414B (en)
NZ (4) NZ569562A (en)
PE (1) PE20080146A1 (en)
PL (6) PL2471907T3 (en)
PT (5) PT2471905T (en)
WO (1) WO2007079183A2 (en)
ZA (2) ZA201004182B (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008019148A2 (en) * 2006-08-04 2008-02-14 Anthrogenesis Corporation Tumor suppression using placental stem cells
WO2008051568A2 (en) * 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
WO2008100497A1 (en) * 2007-02-12 2008-08-21 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
WO2008100498A2 (en) * 2007-02-12 2008-08-21 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
EP2014294A1 (en) * 2007-07-13 2009-01-14 Institut National De La Sante Et De La Recherche Medicale (Inserm) Use of CD200 as a mesenchymal stem cells marker
EP2023717A2 (en) * 2006-05-11 2009-02-18 Naoko Takebe Methods for collecting and using placenta cord blood stem cells
WO2009061024A1 (en) 2007-11-09 2009-05-14 Rnl Bio Co., Ltd Method for isolating and culturing adult stem cells derived from human amniotic epithelium
WO2009061447A2 (en) * 2007-11-07 2009-05-14 Anthrogenesis Corporation Treatment of premature birth complications
WO2010040262A1 (en) * 2008-10-10 2010-04-15 深圳市嘉天源生物科技有限公司 Methods for isolating animal embryonic mesenchymal stem cells and extracting secretion substance thereof
US20100272694A1 (en) * 2008-10-17 2010-10-28 Affiliated Hospital Of Ningxia Medical University Clinic compliant method for banking human placental mesenchymal cells
WO2010141654A1 (en) * 2009-06-05 2010-12-09 Anthrogenesis Corporation Improved method of collecting placental cells
WO2011010966A1 (en) * 2009-07-23 2011-01-27 Agency For Science, Technology And Research (A*Star) Pre-natal mesenchymal stem cells
CN102186338A (en) * 2008-08-20 2011-09-14 人类起源公司 Improved cell composition and methods of making the same
JP2012500792A (en) * 2008-08-22 2012-01-12 アンスロジェネシス コーポレーション Methods and compositions for the treatment of bone defects with placental cell populations
EP2471904A2 (en) 2005-12-29 2012-07-04 Anthrogenesis Corporation Placental stem cell populations
WO2012092458A3 (en) * 2010-12-30 2012-08-23 Anthrogenesis Corporation Compositions comprising placental stem cells and platelet rich plasma, and methods of use thereof
CN102676451A (en) * 2011-09-05 2012-09-19 博雅干细胞科技有限公司 Method for separating mesenchymal stem cells from placenta
EP2651451A1 (en) * 2010-12-17 2013-10-23 Anthrogenesis Corporation Treatment of spinal cord injury and traumatic brain injury using placental stem cells
US20140219970A1 (en) * 2010-04-08 2014-08-07 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
EP2727998A3 (en) * 2008-04-21 2014-08-13 Viacyte, Inc. Methods for purifying pancreatic endoderm cells derived from human embryonic stem cells
US20140242038A1 (en) * 2011-10-11 2014-08-28 The Trustees Of Columbia University In The City Of New York Method for generating beta cells
WO2015073800A2 (en) 2013-11-15 2015-05-21 Anthrogenesis Corporation Compositions comprising human placental perfusate cells, subpopulations thereof, and their uses
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
EP2773746A4 (en) * 2011-11-01 2015-08-05 Neostem Inc Adult mesenchymal stem cell (msc) compositions and methods for preparing the same
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
JP2017002071A (en) * 2007-09-26 2017-01-05 アンスロジェネシス コーポレーション Angiogenic cells from human placental perfusate
EP2968418A4 (en) * 2013-03-15 2017-02-22 Nutech Medical, Inc. Preparations derived from placental materials of making and using same
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
EP3299452A1 (en) * 2000-12-06 2018-03-28 Anthrogenesis Corporation Method of collecting placental stem cells
US9956248B2 (en) 2010-02-18 2018-05-01 Osiris Therapeutics, Inc. Methods of manufacture of therapeutic products comprising vitalized placental dispersions
EP3214171A4 (en) * 2014-10-29 2018-07-11 Sungkwang Medical Foundation Placenta-derived cells excreting c3 or c1r complement and composition containing same
EP3103463B1 (en) 2007-09-19 2020-01-01 Pluristem Ltd. Adherent cells from adipose or placenta tissues and use thereof in therapy
WO2020194307A1 (en) * 2019-03-27 2020-10-01 Pluristem Ltd. Methods and compositions for aesthetic and cosmetic treatment and stimulating hair growth

Families Citing this family (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7311905B2 (en) * 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
EP2316463B1 (en) 2001-02-14 2017-04-12 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making them
US7700090B2 (en) * 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US7498171B2 (en) * 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
KR20100125479A (en) * 2002-11-26 2010-11-30 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US7875272B2 (en) * 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8703121B2 (en) 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
AU2004296765B2 (en) * 2003-12-02 2011-03-24 Celgene Corporation Methods and compositions for the treatment and management of hemoglobinopathy and anemia
US20050176139A1 (en) * 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
WO2005097190A2 (en) * 2004-03-26 2005-10-20 Celgene Corporation Systems and methods for providing a stem cell bank
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
JP5340599B2 (en) * 2004-12-23 2013-11-13 エシコン・インコーポレイテッド Umbilical tissue-derived postpartum cells and methods for producing and using the same
WO2006102643A2 (en) * 2005-03-24 2006-09-28 Caritas St. Elizabeth Medical Center Boston, Inc. Stably transformed bone marrow-derived cells and uses thereof
MX343814B (en) * 2005-10-13 2016-11-24 Anthrogenesis Corp Immunomodulation using placental stem cells.
KR20080056302A (en) * 2005-10-13 2008-06-20 안트로제네시스 코포레이션 Production of oligodendrocytes from placenta-derived stem cells
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
ES2391034T3 (en) 2005-12-19 2012-11-20 Ethicon, Inc. In vitro expansion of postpartum derived cells in rotary bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
CN101374941A (en) * 2005-12-29 2009-02-25 人类起源公司 Improved composition for collecting and preserving placental stem cells and methods of using the composition
US9944900B2 (en) * 2006-01-18 2018-04-17 Hemacell Perfusion Pulsatile perfusion extraction method for non-embryonic pluripotent stem cells
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
ES2623141T3 (en) 2007-01-17 2017-07-10 Noveome Biotherapeutics, Inc. New methods to modulate inflammatory and / or immune responses
WO2008109816A1 (en) * 2007-03-08 2008-09-12 Hemacell Perfusion, Inc. Method for isolation of afterbirth derived cells
US20100172830A1 (en) * 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
CA2688504A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US20100129328A1 (en) * 2007-07-24 2010-05-27 Sing George L Methods for promoting hair growth
EP3689421A1 (en) 2007-09-07 2020-08-05 MiMedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
US10925903B2 (en) 2007-09-13 2021-02-23 Reprobiogen Inc. Use of cells derived from first trimester umbilical cord tissue
US20090074731A1 (en) * 2007-09-13 2009-03-19 Librach Clifford L Method of isolation and use of cells derived from first trimester umbilical cord tissue
ES2719931T3 (en) 2007-09-28 2019-07-16 Celularity Inc Tumor suppression using human placental perfusate and intermediate natural killer cells that come from human placenta
CN102036688B (en) 2007-10-05 2014-07-02 伊西康公司 Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
CA2752838A1 (en) * 2008-02-18 2009-08-27 Genetic Technologies Limited Cell processing and/or enrichment methods
US20120028352A1 (en) * 2008-03-17 2012-02-02 Agency For Science, Technology And Research Microcarriers for Stem Cell Culture
EP2479260B1 (en) 2008-03-17 2016-01-06 Agency For Science, Technology And Research Microcarriers for stem cell culture
US9458431B2 (en) 2008-03-17 2016-10-04 Agency For Science, Technology And Research Microcarriers for stem cell culture
US8497122B2 (en) 2008-04-11 2013-07-30 Washington University Biomarkers for Niemann-pick C disease and related disorders
EP2303293A1 (en) * 2008-05-27 2011-04-06 Pluristem Ltd. Methods of treating inflammatory colon diseases
CA2734237C (en) * 2008-08-20 2019-07-02 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
WO2010026573A1 (en) * 2008-09-02 2010-03-11 Pluristem Ltd. Methods of selection of cells for transplantation
EP3115451A1 (en) * 2008-09-02 2017-01-11 Pluristem Ltd. Adherent cells from placenta tissue and use thereof in therapy
US9096827B2 (en) * 2008-09-02 2015-08-04 Pluristem Ltd. Adherent cells from placenta tissue and use thereof in therapy
RU2015130665A (en) 2008-11-19 2018-12-24 Антродженезис Корпорейшн AMNIOTIC ADHESIVE CELLS
NZ592839A (en) * 2008-11-21 2012-10-26 Anthrogenesis Corp Treatment of diseases, disorders or conditions of the lung using placental cells
AU2009327384B2 (en) 2008-12-19 2014-07-10 DePuy Synthes Products, LLC Treatment of lung and pulmonary diseases and disorders
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
US8771677B2 (en) 2008-12-29 2014-07-08 Vladimir B Serikov Colony-forming unit cell of human chorion and method to obtain and use thereof
PT2411506T (en) 2009-03-25 2019-03-19 Celularity Inc Tumor suppression using human placenta-derived intermediate natural killer cells and immunomodulatory compounds
EP2411504B1 (en) 2009-03-26 2017-05-10 DePuy Synthes Products, Inc. Human umbilical cord tissue cells as therapy for alzheimer's disease
CA2795268A1 (en) 2009-04-21 2010-10-28 Genetic Technologies Limited Methods for obtaining fetal genetic material
US8796315B2 (en) 2009-06-25 2014-08-05 Darlene E. McCord Methods for improved wound closure employing olivamine and human umbilical vein endothelial cells
MX2012000110A (en) * 2009-07-02 2012-04-02 Anthrogenesis Corp Method of producing erythrocytes without feeder cells.
CN101942413B (en) * 2009-07-07 2015-02-11 翁炳焕 Birth defect cell bank and construction method thereof
ES2845691T3 (en) * 2009-10-23 2021-07-27 Caladrius Biosciences Inc Compositions and uses for the treatment of progressive myocardial injuries due to vascular insufficiency
KR20120115602A (en) * 2010-01-26 2012-10-18 안트로제네시스 코포레이션 Treatment of bone-related cancers using placental stem cells
AU2013203479B2 (en) * 2010-04-07 2016-05-19 Celularity Inc. Angiogenesis using placental stem cells
TWI756797B (en) 2010-04-07 2022-03-01 美商瑟魯勒瑞堤股份有限公司 Angiogenesis using placental stem cells
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
CN103097520B (en) 2010-07-13 2017-12-05 人类起源公司 The method for producing NK
KR101211913B1 (en) * 2010-07-16 2012-12-13 주식회사 알앤엘바이오 Medium for Culturing Mesenchymal Stem Cells Derived from Amnion and Method for Culturing Mesenchymal Stem Cells Derived from Amnion Using thereof
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
US20120171180A1 (en) 2010-12-30 2012-07-05 Sascha Abramson Compositions comprising amnion derived adherent cells and platelet-rich plasma
WO2012092485A1 (en) 2010-12-31 2012-07-05 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
US20140017209A1 (en) * 2011-03-22 2014-01-16 Pluristem Ltd. Methods for treating radiation or chemical injury
KR101835917B1 (en) 2011-03-22 2018-03-07 플루리스템 리미티드 Methods for treating radiation or chemical injury
US20120276215A1 (en) * 2011-04-26 2012-11-01 Riordan Neil H Therapeutic Conditioned Media
EP2731440A4 (en) * 2011-07-15 2015-04-29 Anthrogenesis Corp Treatment of radiation injury using amnion derived adherent cells
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US9611513B2 (en) 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
US11311367B2 (en) 2012-01-31 2022-04-26 Wake Forest University Health Sciences Tissue-engineered gut-sphincter complexes and methods of making the same
EP3338548B1 (en) 2012-02-23 2020-04-08 Celularity, Inc. Identification of antitumor compounds using placenta
WO2013131192A1 (en) * 2012-03-06 2013-09-12 Aris Aziz Placental stem cells, methods for isolating same and use thereof
KR101422559B1 (en) * 2012-03-09 2014-07-24 창원대학교 산학협력단 Culture medium of adipose-derived stem cell, method for preparing the same, and composition for promoting hair growth comprising the same
KR101523040B1 (en) * 2012-06-26 2015-05-26 라정찬 Method for Preparing High Concentration of Stem Cells
US9657290B2 (en) 2012-07-03 2017-05-23 The Board Of Trustees Of The Leland Stanford Junior University Scalable bio-element analysis
JP2015526088A (en) 2012-08-13 2015-09-10 アントフロゲネシス コーポレーション Natural killer cells and uses thereof
KR101532556B1 (en) * 2012-09-03 2015-06-30 메디포스트(주) Method for culturing mesenchymal stem cells
MX2015002710A (en) 2012-09-04 2015-07-23 Anthrogenesis Corp Methods of tissue generation.
DK2892996T3 (en) * 2012-09-07 2019-10-07 Biolamina Ab STEM CELL BANK
DK2909314T3 (en) 2012-10-19 2021-03-08 Celularity Inc Treatment of pain with amnion derived adherent cells
CA2892375A1 (en) 2012-11-30 2014-06-05 Darlene E. MCCORD Hydroxytyrosol and oleuropein compositions for induction of dna damage, cell death and lsd1 inhibition
WO2014089440A1 (en) 2012-12-07 2014-06-12 Anthrogenesis Corporation Treating oral lesions using placental extracellular matrix
AU2013359076A1 (en) 2012-12-14 2015-07-02 Anthrogenesis Corporation Anoikis resistant placental stem cells and uses thereof
AU2013204922B2 (en) 2012-12-20 2015-05-14 Celgene Corporation Chimeric antigen receptors
EP2959909B1 (en) * 2013-02-20 2019-12-18 Samsung Life Public Welfare Foundation Composition for treating inflammatory brain diseases which includes stem cell as active ingredient
WO2014159826A1 (en) * 2013-03-14 2014-10-02 Antrhogenesis Corporation Use of placental stem cells in treatment of acute kidney injury
JP2016512433A (en) 2013-03-14 2016-04-28 アントフロゲネシス コーポレーション Enhanced placental stem cells and uses thereof
US9896656B2 (en) * 2013-03-15 2018-02-20 Wake Forest University Health Sciences Neural progenitor cell differentiation
NO2975611T3 (en) 2013-03-15 2018-06-09
CA2907397C (en) 2013-03-15 2022-11-22 Anthrogenesis Corporation Modified t lymphocytes
US9795639B1 (en) * 2013-03-16 2017-10-24 BioDlogics, LLC Methods for the treatment of erectile dysfunction by human birth tissue material compostion
CN103173405B (en) * 2013-03-21 2017-11-07 北京京蒙高科干细胞技术有限公司 A kind of simple separation of Human plactnta myofibroblast mother cell and authentication method
KR101583569B1 (en) * 2013-05-15 2016-01-08 라정찬 Composition for Intravenous Administration Comprising Stem Cells
US20160304832A1 (en) 2013-06-24 2016-10-20 Anthrogenesis Corporation Extracellular matrix composition beads for cell culture
JP6545690B2 (en) * 2014-01-08 2019-07-17 サムスン ライフ パブリック ウェルフェア ファウンデーション Stem cells derived from trophoblast basal layer and cell therapeutic agent containing the same
CN106459908B (en) * 2014-01-08 2019-12-13 社会福祉法人三星生命公益财团 Stem cells derived from pure trophoblast and cell therapeutic agent comprising the same
CN103756965B (en) * 2014-01-27 2016-04-06 山东省齐鲁干细胞工程有限公司 A kind of method of lavation hemopoietic stem cell from placenta
WO2015164233A1 (en) * 2014-04-21 2015-10-29 Anthrogenesis Corporation Treatment of conditions and complications in infants
EP3750993B1 (en) 2014-07-11 2023-12-27 Celgene Corporation Methods of improving vector transduction efficiency into t lymphocytes
SG11201701111SA (en) 2014-08-12 2017-03-30 Anthrogenesis Corp Car-t lymphocytes engineered to home to lymph node b cell zone, skin, or gastrointestinal tract
AU2015330855A1 (en) 2014-10-09 2017-04-27 Celularity Inc. Placenta-derived adherent cell exosomes and uses thereof
US10370653B2 (en) 2015-02-22 2019-08-06 The Board Of Trustees Of The Leland Stanford Junior University Micro-screening apparatus, process, and products
WO2016149492A1 (en) * 2015-03-18 2016-09-22 Anthrogenesis Corporation Methods of banking placental tissue and cells
CA2986702C (en) 2015-05-21 2023-04-04 David Wang Modified demineralized cortical bone fibers
NZ737474A (en) * 2015-05-28 2022-11-25 Celularity Inc Placental-derived stem cells to restore the regenerative engine, correct proteomic defects and extend lifespan
WO2017039251A1 (en) * 2015-08-28 2017-03-09 주식회사 차바이오텍 Enhanced postnatal adherent cell, and use for same
SG10201912572VA (en) 2015-09-15 2020-02-27 Celularity Inc Treatment of diabetic peripheral neuropathy using placental cells
MX2018004005A (en) * 2015-09-29 2018-06-12 Celularity Inc Cell potency assay.
US20190015190A1 (en) 2015-12-30 2019-01-17 Wake Forest University Health Sciences Tissue-engineered gut-sphincter complexes and methods of making the same
EP3429360A4 (en) * 2016-03-16 2019-08-28 Cell Medicine, Inc. Mesenchymal stem cells with enhanced efficacy
EP3516392A4 (en) * 2016-09-19 2020-05-20 The Board of Trustees of the Leland Stanford Junior University Micro-screening and sorting apparatus, process, and products
EP3538277A4 (en) 2016-11-14 2020-05-27 Orca Biosystems, Inc. Methods and apparatuses for sorting target particles
WO2018106742A1 (en) 2016-12-05 2018-06-14 Celularity, Inc. Treatment of lymphedema and related conditions using placental adherent cells
US10433112B2 (en) * 2017-02-22 2019-10-01 Middle Chart, LLC Methods and apparatus for orienteering
CN110621321A (en) 2017-03-15 2019-12-27 浩康生物系统公司 Compositions and methods for hematopoietic stem cell transplantation
WO2018186421A1 (en) * 2017-04-03 2018-10-11 株式会社カネカ Cell population including mesenchymal stem cells, production method therefor, and pharmaceutical composition
CN107586757A (en) * 2017-11-01 2018-01-16 天津昂赛细胞基因工程有限公司 One boar Mesenchymal stem cell nutrient solution and preparation method thereof
US10142452B1 (en) * 2018-05-29 2018-11-27 Guangzhou Zhijing Technology Co., Ltd Mobile terminal unlock method
KR20210111762A (en) 2018-11-30 2021-09-13 셀룰래리티 인코포레이티드 Proliferation of Natural Killer Cells and ILC3 Cells Using Novel Aromatic Compounds
CN109908351B (en) * 2019-04-08 2021-04-06 中南大学 Application of circ _ ADARB1 in preparation of nasopharyngeal carcinoma treatment preparation and treatment preparation
US20220265712A1 (en) 2019-06-14 2022-08-25 Celularity Inc. Populations of natural killer cells for treating cancers
WO2021003254A1 (en) * 2019-07-01 2021-01-07 Auxocell Laboratories, Inc. Cryopreservation medium comprising a tissue extract
CN110468099B (en) * 2019-07-19 2023-08-15 海门生原干细胞科技有限公司 Bone marrow mesenchymal stem cell culture medium and application thereof
US20220273716A1 (en) 2019-07-25 2022-09-01 Celularity Inc. Populations of natural killer cells comprising a cd38 chimeric antigen receptor
WO2021022229A1 (en) 2019-07-31 2021-02-04 Celularity Inc. Populations of natural killer cells comprising a cleavage resistant cd16
CN110592005A (en) * 2019-08-14 2019-12-20 广州乾晖生物科技有限公司 Method for separating mesenchymal stem cells
WO2021113849A1 (en) 2019-12-05 2021-06-10 Celularity Inc. Her2+ cancer treatment with populations of natural killer cells comprising a cleavage resistant cd16
US20220000919A1 (en) 2020-01-29 2022-01-06 Celularity Inc. Placental derived natural killer cells for treatment of coronavirus infections
US20230210909A1 (en) 2020-05-08 2023-07-06 Celularity Inc. Placenta-derived adherent (pda) stem cell for the treatment of adults with sars-cov-2 related acute respiratory failure and ards (covid-19)
WO2021257073A1 (en) * 2020-06-17 2021-12-23 Joy Kong Method for producing mesenchymal stem cells from neonatal stem cells
CN111956787A (en) * 2020-07-06 2020-11-20 沃森克里克(北京)生物科技有限公司 Hair growth promoter and hair growth promoter and application thereof
WO2022120382A1 (en) * 2020-12-03 2022-06-09 42Bio, Inc. Regenerative tissue extract from non-human mammalian placenta
CN112592885B (en) * 2020-12-23 2022-06-17 山东华领奥源生物科技有限公司 Separation culture method of placenta sub-totipotent stem cells
WO2023278628A1 (en) 2021-06-29 2023-01-05 Celularity Inc. Human placental hematopoietic stem cell derived natural killer cells in acute myeloid leukemia (aml) remission with minimal residual disease (mrd) or relapsed/refractory aml
WO2023010123A1 (en) 2021-07-29 2023-02-02 Celularity Inc. Placenta-dervied nk cells as a senolytic for therapeutic and other uses
WO2023137344A1 (en) 2022-01-11 2023-07-20 Celularity Inc. Cleavage resistant cd16 constructs and uses thereof
CN116144583B (en) * 2023-04-19 2023-08-25 北京雪颜博润生物科技有限公司 Method for obtaining human placenta embryo-derived stem cells and application

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006111706A1 (en) * 2005-04-16 2006-10-26 Axordia Limited Cytotrophoblast stem cell
WO2007047468A2 (en) * 2005-10-13 2007-04-26 Anthrogenesis Corporation Immunomodulation using placental stem cells

Family Cites Families (334)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3862002A (en) 1962-05-08 1975-01-21 Sanfar Lab Inc Production of physiologically active placental substances
US3372581A (en) 1965-04-27 1968-03-12 Tell Sven Axel Dynamometer
US4829000A (en) 1985-08-30 1989-05-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Reconstituted basement membrane complex with biological activity
US4798824A (en) 1985-10-03 1989-01-17 Wisconsin Alumni Research Foundation Perfusate for the preservation of organs
US5266480A (en) 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US5863531A (en) * 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US5744347A (en) 1987-01-16 1998-04-28 Ohio University Edison Biotechnology Institute Yolk sac stem cells and their uses
NZ226750A (en) 1987-10-29 1990-09-26 Amrad Corp Ltd Immortalisation of neural precursor cells by introducing a retrovirus vector containing a myc-oncogene
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
GB8803697D0 (en) 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
FR2646438B1 (en) 1989-03-20 2007-11-02 Pasteur Institut A METHOD FOR SPECIFIC REPLACEMENT OF A COPY OF A GENE PRESENT IN THE RECEIVER GENOME BY INTEGRATION OF A GENE DIFFERENT FROM THAT OR INTEGRATION
US5763266A (en) 1989-06-15 1998-06-09 The Regents Of The University Of Michigan Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells
US5437994A (en) 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5399493A (en) 1989-06-15 1995-03-21 The Regents Of The University Of Michigan Methods and compositions for the optimization of human hematopoietic progenitor cell cultures
US5605822A (en) * 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5635386A (en) 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
AU633958B2 (en) 1989-07-25 1993-02-11 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
US5464764A (en) * 1989-08-22 1995-11-07 University Of Utah Research Foundation Positive-negative selection methods and vectors
KR100233781B1 (en) 1989-11-06 1999-12-01 스테펜 에이. 쉐어윈 Production of proteins using homologous recombination
US5272071A (en) 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
US5061620A (en) * 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5635387A (en) 1990-04-23 1997-06-03 Cellpro, Inc. Methods and device for culturing human hematopoietic cells and their precursors
US6326198B1 (en) 1990-06-14 2001-12-04 Regents Of The University Of Michigan Methods and compositions for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5226914A (en) 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5837539A (en) 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
US6010696A (en) 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US5197985A (en) 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5733542A (en) 1990-11-16 1998-03-31 Haynesworth; Stephen E. Enhancing bone marrow engraftment using MSCS
US5190556A (en) 1991-03-19 1993-03-02 O.B. Tech, Inc. Cord cutter sampler
US5744361A (en) * 1991-04-09 1998-04-28 Indiana University Expansion of human hematopoietic progenitor cells in a liquid medium
US5750376A (en) 1991-07-08 1998-05-12 Neurospheres Holdings Ltd. In vitro growth and proliferation of genetically modified multipotent neural stem cells and their progeny
WO1993003139A1 (en) 1991-08-08 1993-02-18 Kao Corporation Cell culture support, production thereof, and production of cell cluster using same
EP0529751A1 (en) 1991-08-09 1993-03-03 W.R. Grace & Co.-Conn. Cell culture substrate, test material for cell culture and preparations thereof
WO1993004169A1 (en) 1991-08-20 1993-03-04 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
US5356373A (en) 1991-11-15 1994-10-18 Miles Inc. Method and apparatus for autologous transfusions in premature infants
CA2125985C (en) 1991-12-23 2001-04-17 Stewart Craig Stem cell inhibiting proteins
EP0590156A4 (en) 1992-03-31 1994-07-13 Toray Industries Novel, physiologically active protein and hemopoietic stem cell growth promoter
US5460964A (en) 1992-04-03 1995-10-24 Regents Of The University Of Minnesota Method for culturing hematopoietic cells
US5552267A (en) 1992-04-03 1996-09-03 The Trustees Of Columbia University In The City Of New York Solution for prolonged organ preservation
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young Scar inhibitory factor and use thereof
US5672346A (en) 1992-07-27 1997-09-30 Indiana University Foundation Human stem cell compositions and methods
US5693482A (en) 1992-07-27 1997-12-02 California Institute Of Technology Neural chest stem cell assay
US5849553A (en) 1992-07-27 1998-12-15 California Institute Of Technology Mammalian multipotent neural stem cells
EP0669974A1 (en) 1992-11-16 1995-09-06 Rhone-Poulenc Rorer Pharmaceuticals Inc. Pluripotential quiescent stem cell population
US5772992A (en) 1992-11-24 1998-06-30 G.D. Searle & Co. Compositions for co-administration of interleukin-3 mutants and other cytokines and hematopoietic factors
US5654186A (en) 1993-02-26 1997-08-05 The Picower Institute For Medical Research Blood-borne mesenchymal cells
UA40613C2 (en) * 1993-03-31 2001-08-15 Про-Нейрон, Інк. Polypeptide having activity of stem cells proliferation (variants), pharmaceutical composition (variants), pharmaceutical composition for inhibiting division stem cells in mammals, pharmaceutical composition for the treatment of diseases, including hyperproliferation states, pharmaceutical composition for differential protection of normal stem cells being different of leucosis, method for purification of polypeptide
GB9308271D0 (en) 1993-04-21 1993-06-02 Univ Edinburgh Method of isolating and/or enriching and/or selectively propagating pluripotential animal cells and animals for use in said method
US5709854A (en) * 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
US5372581A (en) 1993-07-21 1994-12-13 Minneapolis Children's Services Corporation Method and apparatus for placental blood collection
IL107483A0 (en) 1993-11-03 1994-02-27 Yeda Res & Dev Bone marrow transplantation
US5591625A (en) * 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US6288030B1 (en) * 1993-12-22 2001-09-11 Amgen Inc. Stem cell factor formulations and methods
US6001654A (en) 1994-01-28 1999-12-14 California Institute Of Technology Methods for differentiating neural stem cells to neurons or smooth muscle cells using TGT-β super family growth factors
US5942496A (en) 1994-02-18 1999-08-24 The Regent Of The University Of Michigan Methods and compositions for multiple gene transfer into bone cells
US5580724A (en) 1994-03-25 1996-12-03 Board Of Regents, The University Of Texas System Differential expansion of fetal stem cells in maternal circulation for use in prenatal genetic analysis
US6174333B1 (en) * 1994-06-06 2001-01-16 Osiris Therapeutics, Inc. Biomatrix for soft tissue regeneration using mesenchymal stem cells
JPH10505250A (en) * 1994-06-06 1998-05-26 ケース ウエスターン リザーブ ユニバーシティ Biomatrix for tissue regeneration
DE4422667A1 (en) 1994-06-30 1996-01-04 Boehringer Ingelheim Int Process for the production and cultivation of hematopoietic progenitor cells
US6103522A (en) * 1994-07-20 2000-08-15 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoiesis
US5516532A (en) * 1994-08-05 1996-05-14 Children's Medical Center Corporation Injectable non-immunogenic cartilage and bone preparation
US5840502A (en) 1994-08-31 1998-11-24 Activated Cell Therapy, Inc. Methods for enriching specific cell-types by density gradient centrifugation
US5827742A (en) 1994-09-01 1998-10-27 Beth Israel Deaconess Medical Center, Inc. Method of selecting pluripotent hematopioetic progenitor cells
US5665557A (en) 1994-11-14 1997-09-09 Systemix, Inc. Method of purifying a population of cells enriched for hematopoietic stem cells populations of cells obtained thereby and methods of use thereof
US5874301A (en) 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5914268A (en) 1994-11-21 1999-06-22 National Jewish Center For Immunology & Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5789147A (en) 1994-12-05 1998-08-04 New York Blood Center, Inc. Method for concentrating white cells from whole blood by adding a red cell sedimentation reagent to whole anticoagulated blood
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US7410773B2 (en) 1995-02-02 2008-08-12 Ghazi Jaswinder Dhoot Method of preparing an undifferentiated cell
US5695998A (en) 1995-02-10 1997-12-09 Purdue Research Foundation Submucosa as a growth substrate for islet cells
US6011000A (en) 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US5906934A (en) * 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US6974571B2 (en) 1995-03-28 2005-12-13 Thomas Jefferson University Isolated stromal cells and methods of using the same
US5716616A (en) 1995-03-28 1998-02-10 Thomas Jefferson University Isolated stromal cells for treating diseases, disorders or conditions characterized by bone defects
US5677139A (en) 1995-04-21 1997-10-14 President And Fellows Of Harvard College In vitro differentiation of CD34+ progenitor cells into T lymphocytes
US5733541A (en) * 1995-04-21 1998-03-31 The Regent Of The University Of Michigan Hematopoietic cells: compositions and methods
ES2128850T3 (en) 1995-04-27 1999-05-16 Procter & Gamble SUPPORT TREATMENT TREATED WITH A REVERSE EMULSION WITH A HIGH INTERNAL AQUEOUS PHASE MADE WITH AN EMULSIONANT ORGANOPOLISILOXANE - POLYOXYALKYLENE.
US5925567A (en) 1995-05-19 1999-07-20 T. Breeders, Inc. Selective expansion of target cell populations
US5908782A (en) 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US5830708A (en) 1995-06-06 1998-11-03 Advanced Tissue Sciences, Inc. Methods for production of a naturally secreted extracellular matrix
AU6223296A (en) 1995-06-07 1996-12-30 Novartis Ag Methods for obtaining compositions enriched for hematopoieti c stem cells and antibodies for use therein
US5877299A (en) 1995-06-16 1999-03-02 Stemcell Technologies Inc. Methods for preparing enriched human hematopoietic cell preparations
US5654381A (en) 1995-06-16 1997-08-05 Massachusetts Institute Of Technology Functionalized polyester graft copolymers
US6306575B1 (en) 1995-06-16 2001-10-23 Stemcell Technologies, Inc. Methods for preparing enriched human hematopoietic cell preparations
US5935576A (en) 1995-09-13 1999-08-10 Fordham University Compositions and methods for the treatment and prevention of neoplastic diseases using heat shock proteins complexed with exogenous antigens
US5800539A (en) 1995-11-08 1998-09-01 Emory University Method of allogeneic hematopoietic stem cell transplantation without graft failure or graft vs. host disease
US5858782A (en) * 1995-11-13 1999-01-12 Regents Of The University Of Michigan Functional human hematopoietic cells
WO1997018298A1 (en) 1995-11-14 1997-05-22 Regents Of The University Of Minnesota Ex vivo culture of stem cells
AU713280B2 (en) 1995-11-16 1999-11-25 Case Western Reserve University In vitro chondrogenic induction of human mesenchymal stem cells
EP0861894B1 (en) * 1995-11-17 2006-03-08 Asahi Kasei Kabushiki Kaisha Differentiation-suppressive polypeptide
US5716794A (en) * 1996-03-29 1998-02-10 Xybernaut Corporation Celiac antigen
WO1997040137A1 (en) * 1996-04-19 1997-10-30 Osiris Therapeutics, Inc. Regeneration and augmentation of bone using mesenchymal stem cells
WO1997041208A1 (en) 1996-04-26 1997-11-06 Case Western Reserve University Skin regeneration using mesenchymal stem cells
US6455678B1 (en) 1996-04-26 2002-09-24 Amcell Corporation Human hematopoietic stem and progenitor cell antigen
US5919176A (en) 1996-05-14 1999-07-06 Children's Hospital Medical Center Of Northern California Apparatus and method for collecting blood from an umbilical cord
US5827740A (en) 1996-07-30 1998-10-27 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
US6358737B1 (en) 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US5851984A (en) 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US5916202A (en) 1996-08-30 1999-06-29 Haswell; John N. Umbilical cord blood collection
US6227202B1 (en) 1996-09-03 2001-05-08 Maulana Azad Medical College Method of organogenesis and tissue regeneration/repair using surgical techniques
US5945337A (en) 1996-10-18 1999-08-31 Quality Biological, Inc. Method for culturing CD34+ cells in a serum-free medium
US5919702A (en) 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
US5969105A (en) 1996-10-25 1999-10-19 Feng; Yiqing Stem cell factor receptor agonists
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6152142A (en) 1997-02-28 2000-11-28 Tseng; Scheffer C. G. Grafts made from amniotic membrane; methods of separating, preserving, and using such grafts in surgeries
US6231880B1 (en) 1997-05-30 2001-05-15 Susan P. Perrine Compositions and administration of compositions for the treatment of blood disorders
EP1028737B1 (en) 1997-07-03 2007-04-04 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US7514074B2 (en) 1997-07-14 2009-04-07 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
ES2251773T5 (en) 1997-07-14 2009-05-12 Osiris Therapeutics, Inc. REGENERATION OF THE CARDIAC MUSCLE USING MESENQUIMATOSE PRECURSOR CELLS.
US6077708A (en) 1997-07-18 2000-06-20 Collins; Paul C. Method of determining progenitor cell content of a hematopoietic cell culture
US5879318A (en) 1997-08-18 1999-03-09 Npbi International B.V. Method of and closed system for collecting and processing umbilical cord blood
AU9127098A (en) 1997-09-04 1999-03-22 Osiris Therapeutics, Inc. Ligands that modulate differentiation of mesenchymal stem cells
US5968829A (en) 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
US6093531A (en) 1997-09-29 2000-07-25 Neurospheres Holdings Ltd. Generation of hematopoietic cells from multipotent neural stem cells
AU729377B2 (en) 1997-10-23 2001-02-01 Asterias Biotherapeutics, Inc. Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
US6248587B1 (en) 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6059968A (en) 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
US6291240B1 (en) 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
JP4441115B2 (en) 1998-03-13 2010-03-31 オシリス セラピューティクス,インコーポレイテッド Methods and uses of human non-self mesenchymal stem cells
US6368636B1 (en) * 1998-03-18 2002-04-09 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
PT1066052E (en) 1998-03-18 2006-06-30 Osiris Therapeutics Inc MESENCHIMATE STEM CELLS FOR PREVENTION AND TREATMENT
AU2999199A (en) 1998-04-03 1999-10-25 Osiris Therapeutics, Inc. Mesenchymal stem cells as immunosuppressants
AU770319C (en) 1998-05-04 2004-11-25 Point Therapeutics, Inc. Hematopoietic stimulation
JP2002513545A (en) 1998-05-07 2002-05-14 ザ ユニヴァーシティー オブ サウス フロリダ Bone marrow cells as a source of neurons for brain and spinal cord repair
US6835377B2 (en) 1998-05-13 2004-12-28 Osiris Therapeutics, Inc. Osteoarthritis cartilage regeneration
WO1999061588A1 (en) 1998-05-22 1999-12-02 Osiris Therapeutics, Inc. Production of megakaryocytes by co-culturing human mesenchymal stem cells with cd34+ cells
DE69936720T2 (en) 1998-05-29 2008-04-30 Osiris Therapeutics, Inc. HUMAN CD45 + AND / OR FIBROBLASTES + MESENCHYMAL STEM CELLS
CA2330190C (en) 1998-06-08 2009-12-22 Osiris Therapeutics, Inc. Regulation of hematopoietic stem cell differentiation by the use of human mesenchymal stem cells
CA2329519A1 (en) 1998-06-08 1999-12-16 Osiris Therapeutics, Inc. In vitro maintenance of hematopoietic stem cells
US6713245B2 (en) 1998-07-06 2004-03-30 Diacrin, Inc. Methods for storing neural cells such that they are suitable for transplantation
EP1100488B1 (en) * 1998-07-28 2003-04-23 SYNTHES AG Chur Use of creatine compounds for treatment of bone or cartilage cells and tissues
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
JP3517359B2 (en) 1998-09-14 2004-04-12 テルモ株式会社 Cell separation / collection apparatus and cell separation / collection method
WO2000017325A1 (en) 1998-09-23 2000-03-30 Mount Sinai Hospital Trophoblast cell preparations
WO2000027995A1 (en) 1998-11-09 2000-05-18 Monash University Embryonic stem cells
AU1720400A (en) 1998-11-12 2000-05-29 Cell Science Therapeutics, Inc. Lymphoid tissue-specific cell production from hematopoietic progenitor cells in three-dimensional devices
US6548299B1 (en) 1999-11-12 2003-04-15 Mark J. Pykett Lymphoid tissue-specific cell production from hematopoietic progenitor cells in three-dimensional devices
US6184035B1 (en) 1998-11-18 2001-02-06 California Institute Of Technology Methods for isolation and activation of, and control of differentiation from, skeletal muscle stem or progenitor cells
US6102871A (en) 1998-11-23 2000-08-15 Coe; Rosemarie O. Blood collection funnel
US6328765B1 (en) 1998-12-03 2001-12-11 Gore Enterprise Holdings, Inc. Methods and articles for regenerating living tissue
WO2000038762A1 (en) 1998-12-24 2000-07-06 Biosafe S.A. Blood separation system particularly for concentrating hematopoietic stem cells
US6911201B1 (en) 1999-02-04 2005-06-28 Technion Research & Development Foundation Ltd. Method of producing undifferentiated hemopoietic stem cells using a stationary phase plug-flow bioreactor
US20030007954A1 (en) 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
CA2370330C (en) 1999-04-16 2009-01-20 Wm. Marsh Rice University Biodegradable poly(propylene fumarate) networks cross linked with poly(propylene fumarate)-diacrylate macromers
IN191359B (en) 1999-04-20 2003-11-29 Nat Inst Immunology
AU4860900A (en) 1999-06-02 2000-12-18 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6333029B1 (en) 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US6355699B1 (en) 1999-06-30 2002-03-12 Ethicon, Inc. Process for manufacturing biomedical foams
US8147824B2 (en) 1999-08-05 2012-04-03 Athersys, Inc. Immunomodulatory properties of multipotent adult progenitor cells and uses thereof
WO2002064748A2 (en) 2001-02-14 2002-08-22 Furcht Leo T Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
EP2348104A1 (en) 1999-08-05 2011-07-27 Mcl Llc Multipotent adult stem cells and methods for isolation
US8075881B2 (en) * 1999-08-05 2011-12-13 Regents Of The University Of Minnesota Use of multipotent adult stem cells in treatment of myocardial infarction and congestive heart failure
US7015037B1 (en) 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US6239157B1 (en) 1999-09-10 2001-05-29 Osiris Therapeutics, Inc. Inhibition of osteoclastogenesis
WO2001021767A2 (en) 1999-09-24 2001-03-29 Morphogen Pharmaceuticals, Inc. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US6685936B2 (en) 1999-10-12 2004-02-03 Osiris Therapeutics, Inc. Suppressor cells induced by culture with mesenchymal stem cells for treatment of immune responses in transplantation
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
DE19954421A1 (en) 1999-11-12 2001-05-31 Lohmann Therapie Syst Lts Film-like preparation for the biphase release of pharmacologically active or other substances
JP2003530826A (en) 1999-11-17 2003-10-21 ユニバーシティー オブ ロチェスター Human ex vivo immune system
US7455983B2 (en) 2000-01-11 2008-11-25 Geron Corporation Medium for growing human embryonic stem cells
IL151650A0 (en) 2000-03-09 2003-04-10 Saneron Ccel Therapeutics Inc Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US7405078B2 (en) 2000-03-16 2008-07-29 Cellseed Inc. Bed material for cell culture, method for co-culture of cell and co-cultured cell sheet obtainable therefrom
WO2001075094A1 (en) 2000-04-04 2001-10-11 Thomas Jefferson University Application of myeloid-origin cells to the nervous system
JP2003535148A (en) 2000-06-06 2003-11-25 グラクソ グループ リミテッド How to treat cancer
US20050009876A1 (en) 2000-07-31 2005-01-13 Bhagwat Shripad S. Indazole compounds, compositions thereof and methods of treatment therewith
US7560280B2 (en) 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
EP1757681A1 (en) 2000-11-22 2007-02-28 Geron Corporation Tolerizing allografts of pluripotent stem cells
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US20080152629A1 (en) 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US7091353B2 (en) 2000-12-27 2006-08-15 Celgene Corporation Isoindole-imide compounds, compositions, and uses thereof
US20030045552A1 (en) 2000-12-27 2003-03-06 Robarge Michael J. Isoindole-imide compounds, compositions, and uses thereof
EP2314673B1 (en) * 2001-02-14 2013-07-24 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2316463B1 (en) 2001-02-14 2017-04-12 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making them
US20020132343A1 (en) 2001-03-19 2002-09-19 Clark Lum System and method for delivering umbilical cord-derived tissue-matched stem cells for transplantation
US20030044977A1 (en) * 2001-08-10 2003-03-06 Norio Sakuragawa Human stem cells originated from human amniotic mesenchymal cell layer
DE10139783C1 (en) 2001-08-14 2003-04-17 Transtissue Technologies Gmbh Cell compositions for the treatment of osteoarthritis, and methods for their production
US20030044976A1 (en) 2001-08-27 2003-03-06 Advanced Cell Technology De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
EP1288293A1 (en) 2001-08-30 2003-03-05 Norio Sakuragawa Human neural stem cells originated from human amniotic mesenchymal cell layer
CN1195055C (en) * 2001-09-06 2005-03-30 周胜利 Method for establishing hematopoietic stem cells bank by extracting hematopoietic cells from placenta tissues
US9969980B2 (en) 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
JP2005508393A (en) 2001-11-09 2005-03-31 アーテセル・サイエンシズ・インコーポレーテツド Methods and compositions for the use of stromal cells to support embryonic and adult stem cells
JP4330995B2 (en) 2001-11-15 2009-09-16 チルドレンズ メディカル センター コーポレーション Methods for isolating, proliferating, and differentiating fetal stem cells from chorionic villi, amniotic fluid, and placenta, and methods for their therapeutic use
JP3728750B2 (en) 2001-11-22 2005-12-21 ニプロ株式会社 Cultured skin and method for producing the same
US7799324B2 (en) 2001-12-07 2010-09-21 Geron Corporation Using undifferentiated embryonic stem cells to control the immune system
JP3934539B2 (en) 2001-12-12 2007-06-20 独立行政法人科学技術振興機構 Adult or postnatal tissue progenitor cells derived from placenta
AU2002350352A1 (en) 2001-12-21 2003-07-15 Mount Sinai Hospital Cellular compositions and methods of making and using them
US20040018178A1 (en) 2002-01-22 2004-01-29 Advanced Cell Technology Stem cell-derived endothelial cells modified to disrupt tumor angiogenesis
WO2003068937A2 (en) 2002-02-13 2003-08-21 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
KR100489248B1 (en) * 2002-02-19 2005-05-11 메디포스트(주) Isolation and culture-expansion methods of mesenchymal stem/progenitor cells from umbilical cord blood, and differentiation method of umbilical cord blood-derived mesenchymal stem/progenitor cells into various mesenchymal tissues
US20030161818A1 (en) 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7736892B2 (en) 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20080299090A1 (en) 2002-02-25 2008-12-04 Kansas State University Research Foundation Use Of Umbilical Cord Matrix Cells
CN100579577C (en) 2002-03-15 2010-01-13 退伍军人事务研发服务部 Methods and compositions using cellular asialodeterminants and glycoconjugates for targeting cells to tissues and organs
US20030187515A1 (en) 2002-03-26 2003-10-02 Hariri Robert J. Collagen biofabric and methods of preparing and using the collagen biofabric
AU2003221173A1 (en) * 2002-03-27 2003-10-08 Asahi Kasei Kabushiki Kaisha Placenta-origin mesenchymal cells and medicinal use thereof
MXPA04009996A (en) 2002-04-12 2005-07-01 Celgene Corp Methods for identification of modulators of angiogenesis, compounds discovered thereby, and methods of treatment using the compounds.
US20050118715A1 (en) 2002-04-12 2005-06-02 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US7498171B2 (en) * 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20040161419A1 (en) * 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
AU2003239159A1 (en) 2002-04-19 2003-11-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education Placental derived stem cells and uses thereof
US20050058641A1 (en) 2002-05-22 2005-03-17 Siemionow Maria Z. Tolerance induction and maintenance in hematopoietic stem cell allografts
KR20050008757A (en) 2002-05-30 2005-01-21 셀진 코포레이션 Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
AU2003273574A1 (en) 2002-05-31 2003-12-19 Osiris Therapeutics, Inc. Intraperitoneal delivery of genetically engineered mesenchymal stem cells
US7422736B2 (en) * 2002-07-26 2008-09-09 Food Industry Research And Development Institute Somatic pluripotent cells
EP1527161B1 (en) 2002-07-31 2015-10-28 Yves Saint-Laurent Parfums Stem cells derived from adipous tissue and differentiated cells derived from said cells
JPWO2004018658A1 (en) 2002-08-23 2005-12-08 宣男 櫻川 Human bone stem cells
KR100476790B1 (en) 2002-09-13 2005-03-16 주식회사 제넨메드 A cell preservation solution and the freezing and preservation procedures of animal cells using the said solution
US20040062753A1 (en) 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
KR20100125479A (en) 2002-11-26 2010-11-30 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
JP2004210713A (en) * 2002-12-27 2004-07-29 Asahi Kasei Corp Medical cell preparation derived from placenta
BRPI0407221A (en) 2003-02-11 2006-01-31 John E Davies Wharton Jelly Extract, Method for Obtaining a Human Parent Cell, Methods for Producing a Cell Population, Cell Populations, and Method for Producing Bone Tissue
US20040219136A1 (en) 2003-02-13 2004-11-04 Hariri Robert J Use of umbilical cord blood to treat individuals having a disease, disorder or condition
WO2004087896A2 (en) 2003-03-31 2004-10-14 Pfizer Products Inc. Hepatocyte differentiation of stem cells
CN1548529A (en) 2003-05-09 2004-11-24 中国人民解放军军事医学科学院基础医 Separation method of buffering stem cell in human placenta
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8703121B2 (en) 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US7569385B2 (en) 2003-08-14 2009-08-04 The Regents Of The University Of California Multipotent amniotic fetal stem cells
WO2005038014A1 (en) 2003-10-17 2005-04-28 Innovative Dairy Products Pty Ltd As Trustee For The Participants Of The Cooperative Research Centre For Innovative Dairy Products Isolation of stem cell-like cells and use thereof
US20050089513A1 (en) * 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
WO2005047491A2 (en) 2003-11-10 2005-05-26 Amgen Inc. Methods of using g-csf mobilized c-kit+cells in the production of embryoid body-like cell clusters for tissue repair and in the treatment of cardiac myopathy
KR100560340B1 (en) 2003-11-11 2006-03-14 한훈 Method of isolating and culturing mesenchymal stem cell derived from umbilical cord blood
JP2005151907A (en) 2003-11-27 2005-06-16 Shigeo Saito Human stem cell derived from placenta or amnion and method for establishing the same and method for differentiation-induction to organ
TWI338714B (en) 2003-12-02 2011-03-11 Cathay General Hospital Method of isolation and enrichment of mesenchymal stem cells from amniotic fluid
AU2004296765B2 (en) 2003-12-02 2011-03-24 Celgene Corporation Methods and compositions for the treatment and management of hemoglobinopathy and anemia
WO2007059007A2 (en) * 2005-11-14 2007-05-24 Cythera, Inc. Markers of definitive endoderm
US20050176139A1 (en) 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
US20050266391A1 (en) 2004-01-15 2005-12-01 Bennett Brydon L Methods for preserving tissue
AU2005227295A1 (en) 2004-03-22 2005-10-06 Osiris Therapeutics, Inc. Mesenchymal stem cells and uses therefor
US20080095749A1 (en) 2004-03-22 2008-04-24 Sudeepta Aggarwal Mesenchymal stem cells and uses therefor
WO2005097190A2 (en) 2004-03-26 2005-10-20 Celgene Corporation Systems and methods for providing a stem cell bank
WO2005105992A1 (en) 2004-04-21 2005-11-10 New York Eye & Ear Infirmary Chondrocyte culture formulations
JP2006006249A (en) 2004-06-28 2006-01-12 Hiroshima Univ Method for culturing amnion-derived cell and utilization of the same
CN100564518C (en) 2004-07-20 2009-12-02 成都军区昆明总医院 Placenta amnion cell extract and induce application in the differentiation at mescenchymal stem cell
US7244759B2 (en) 2004-07-28 2007-07-17 Celgene Corporation Isoindoline compounds and methods of making and using the same
US20080292597A1 (en) 2004-07-29 2008-11-27 David A Steenblock Umbilical Cord Stem Cell Composition & Method of Treating Neurological Diseases
DK1789534T3 (en) * 2004-08-16 2014-11-17 Cellres Corp Pte Ltd ISOLATION OF stem / progenitor cells FOSTER HINDE FROM an umbilical cord
US20090104158A1 (en) 2004-09-03 2009-04-23 Moraga Biotechnology Corporation Non-Embryonic Totipotent Blastomere-Like Stem Cells And Methods Therefor
EP1799812A4 (en) 2004-09-16 2009-09-09 Gamida Cell Ltd Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
US7147626B2 (en) * 2004-09-23 2006-12-12 Celgene Corporation Cord blood and placenta collection kit
US7909806B2 (en) 2004-09-23 2011-03-22 Anthrogenesis Corporation Cord blood and placenta collection kit
CN101506355B (en) 2004-09-24 2012-06-27 成血管细胞系统公司 Multipotential expanded mesenchymal precursor cell progeny (MEMP) and uses thereof
US20060069009A1 (en) 2004-09-28 2006-03-30 Messina Darin J Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US20060069008A1 (en) 2004-09-28 2006-03-30 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US8039258B2 (en) 2004-09-28 2011-10-18 Ethicon, Inc. Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20060171930A1 (en) 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
WO2006083394A2 (en) 2004-12-21 2006-08-10 Ethicon, Inc. Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
JP5340599B2 (en) 2004-12-23 2013-11-13 エシコン・インコーポレイテッド Umbilical tissue-derived postpartum cells and methods for producing and using the same
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
US20070041943A1 (en) 2004-12-29 2007-02-22 Children's Hospital Research Expression of virus entry inhibitors and recombinant AAV thereof
AU2006203990B2 (en) 2005-01-07 2011-08-11 Wake Forest University Health Sciences Regeneration of pancreatic islets by amniotic fluid stem cell therapy
WO2006091766A2 (en) 2005-02-24 2006-08-31 Jau-Nan Lee Human trophoblast stem cells and use thereof
US20060222634A1 (en) 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US20100028306A1 (en) 2005-03-31 2010-02-04 Stemnion, Inc. Amnion-Derived Cell Compositions, Methods of Making and Uses Thereof
CN101248171B (en) 2005-04-12 2015-11-25 迈索布拉斯特股份有限公司 By tissue non-specific alkaline phosphatase isolation of adult multipotential cells
CA2606983A1 (en) 2005-04-28 2006-11-09 Stemcell Institute Inc. Method of preparing organ for transplantation
EP1881062B1 (en) 2005-05-09 2012-01-18 Olympus Corporation Methode for culturing mesenchymal stem cell and method for producing biological tissue prosthesis
AU2006202209B2 (en) 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
EP1893249A2 (en) 2005-06-10 2008-03-05 Celgene Corporation Human placental collagen compositions, processes for their preparation, methods of their use and kits comprising the compositions
KR20080026198A (en) * 2005-06-30 2008-03-24 안트로제네시스 코포레이션 Repair of tympanic membrane using placenta derived collagen biofabric
US7928280B2 (en) 2005-07-13 2011-04-19 Anthrogenesis Corporation Treatment of leg ulcers using placenta derived collagen biofabric
EP1919365A2 (en) * 2005-07-13 2008-05-14 Anthrogenesis Corporation Ocular plug formed from placenta derived collagen biofabric
WO2007011693A2 (en) * 2005-07-14 2007-01-25 Medistem Laboratories, Inc. Compositions of placentally-derived stem cells for the treatment of cancer
US7923007B2 (en) 2005-08-08 2011-04-12 Academia Sinica Brain tissue damage therapies
EP1915440A4 (en) 2005-08-19 2009-11-04 Bio Regenerate Inc Compositions of cells enriched for combinations of various stem and progenitor cell populations, methods of use thereof and methods of private banking thereof
KR20080056302A (en) 2005-10-13 2008-06-20 안트로제네시스 코포레이션 Production of oligodendrocytes from placenta-derived stem cells
CN101326280A (en) * 2005-10-13 2008-12-17 人类起源公司 Immunomodulation using placental stem cells
CN100344757C (en) * 2005-10-18 2007-10-24 天津昂赛细胞基因工程有限公司 Human placenta, umbilical cord mesenchyma stemcell stock and its construction method
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
ES2391034T3 (en) 2005-12-19 2012-11-20 Ethicon, Inc. In vitro expansion of postpartum derived cells in rotary bottles
CA2634820C (en) 2005-12-22 2021-05-25 Jane Ennis Viable cells from frozen umbilical cord tissue
US20070160588A1 (en) 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
PT2471905T (en) 2005-12-29 2019-01-11 Celularity Inc Placental stem cell populations
CN101374941A (en) 2005-12-29 2009-02-25 人类起源公司 Improved composition for collecting and preserving placental stem cells and methods of using the composition
US20070253931A1 (en) 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
US20080286249A1 (en) 2006-01-12 2008-11-20 Varney Timothy R Use of mesenchymal stem cells for treating genetic diseases and disorders
MX348735B (en) 2006-01-13 2017-06-27 Mesoblast Int Sarl MESENCHYMAL STEM CELLS EXPRESSING TNF-a RECEPTOR.
CN100545260C (en) 2006-01-13 2009-09-30 深圳市北科生物科技有限公司 The separation of human amnion mesenchymal stem cell and cultural method and medical composition
US9944900B2 (en) 2006-01-18 2018-04-17 Hemacell Perfusion Pulsatile perfusion extraction method for non-embryonic pluripotent stem cells
NZ570614A (en) 2006-01-23 2012-07-27 Athersys Inc Multipotent adult progenitor cell treatment of brain injuries and diseases
CA3028279C (en) 2006-01-23 2021-08-03 Athersys, Inc. Mapc therapeutics without adjunctive immunosuppressive treatment
EP1845154A1 (en) 2006-04-12 2007-10-17 RNL Bio Co., Ltd. Multipotent stem cells derived from placenta tissue and cellular therapeutic agents comprising the same
BRPI0711599B8 (en) 2006-05-11 2021-07-27 Hli Cellular Therapeutics Llc methods for collecting and using stem cells from placental umbilical cord blood
WO2007136673A2 (en) 2006-05-19 2007-11-29 Medistem Laboratories, Inc. Treatment of disc degenerative disease and compositions for same
US20080050814A1 (en) 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
US20080064098A1 (en) 2006-06-05 2008-03-13 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
KR20090031895A (en) * 2006-06-09 2009-03-30 안트로제네시스 코포레이션 Placental niche and use thereof to culture stem cells
US20070287176A1 (en) 2006-06-13 2007-12-13 Alireza Rezania Chorionic villus derived cells
US8475788B2 (en) 2006-06-14 2013-07-02 Stemnion, Inc. Methods of treating spinal cord injury and minimizing scarring
WO2007149548A2 (en) 2006-06-22 2007-12-27 Medistem Laboratories, Inc. Treatment of erectile dysfunction by stem cell therapy
AU2007297575A1 (en) 2006-06-26 2008-03-27 Lifescan, Inc. Pluripotent stem cell culture
WO2008003042A2 (en) 2006-06-28 2008-01-03 The University Of Medicine And Dentistry Of New Jersey Amnion-derived stem cells and uses thereof
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US20090081171A1 (en) 2006-08-11 2009-03-26 Yu-Show Fu Cell system for alleviating syndromes of Parkinson's disease in a mammal
US8105634B2 (en) * 2006-08-15 2012-01-31 Anthrogenesis Corporation Umbilical cord biomaterial for medical use
US20080050347A1 (en) 2006-08-23 2008-02-28 Ichim Thomas E Stem cell therapy for cardiac valvular dysfunction
WO2008036374A2 (en) 2006-09-21 2008-03-27 Medistem Laboratories, Inc. Allogeneic stem cell transplants in non-conditioned recipients
US20080131522A1 (en) 2006-10-03 2008-06-05 Qing Liu Use of placental biomaterial for ocular surgery
US8071135B2 (en) 2006-10-04 2011-12-06 Anthrogenesis Corporation Placental tissue compositions
NZ597223A (en) 2006-10-06 2013-09-27 Anthrogenesis Corp Native (telopeptide) placental collagen compositions
WO2008051568A2 (en) 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
ES2524443T3 (en) 2006-11-13 2014-12-09 DePuy Synthes Products, LLC In vitro expansion of postpartum cells using microcarriers
US8241621B2 (en) 2006-12-18 2012-08-14 Medistem Laboratories Stem cell mediated treg activation/expansion for therapeutic immune modulation
CN103356711A (en) 2007-02-12 2013-10-23 人类起源公司 Immunomodulation using placental stem cells
EP2129775A1 (en) 2007-02-12 2009-12-09 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
US20100172830A1 (en) 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
US20080254005A1 (en) 2007-04-06 2008-10-16 Medistem Labortories Stem Cell Therapy for the Treatment of Autism and Other Disorders
CN101978045A (en) 2007-09-26 2011-02-16 细胞基因细胞疗法公司 Angiogenic cells from human placental perfusate
ES2719931T3 (en) 2007-09-28 2019-07-16 Celularity Inc Tumor suppression using human placental perfusate and intermediate natural killer cells that come from human placenta
KR20170116221A (en) 2007-11-07 2017-10-18 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
US8216997B2 (en) 2008-08-14 2012-07-10 Acceleron Pharma, Inc. Methods for increasing red blood cell levels and treating anemia using a combination of GDF traps and erythropoietin receptor activators
WO2010021714A2 (en) * 2008-08-20 2010-02-25 Anthrogenesis Corporation Improved cell composition and methods of making the same
CA2734237C (en) * 2008-08-20 2019-07-02 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
MX2011001992A (en) 2008-08-22 2011-03-29 Anthrogenesis Corp Methods and compositions for treatment of bone defects with placental cell populations.
RU2015130665A (en) 2008-11-19 2018-12-24 Антродженезис Корпорейшн AMNIOTIC ADHESIVE CELLS
NZ592839A (en) 2008-11-21 2012-10-26 Anthrogenesis Corp Treatment of diseases, disorders or conditions of the lung using placental cells
US20100323446A1 (en) 2009-06-05 2010-12-23 Jill Renee Barnett Method of collecting placental cells
MX2012000110A (en) * 2009-07-02 2012-04-02 Anthrogenesis Corp Method of producing erythrocytes without feeder cells.
KR20120115602A (en) 2010-01-26 2012-10-18 안트로제네시스 코포레이션 Treatment of bone-related cancers using placental stem cells
US20110280849A1 (en) 2010-03-26 2011-11-17 Anthrogenesis Corporation Tumor suppression using human placenta-derived intermediate natural killer cells and immunomodulatory compounds
TWI756797B (en) 2010-04-07 2022-03-01 美商瑟魯勒瑞堤股份有限公司 Angiogenesis using placental stem cells
CN102933221A (en) 2010-04-08 2013-02-13 人类起源公司 Treatment of sarcoidosis using placental stem cells
CN103097520B (en) 2010-07-13 2017-12-05 人类起源公司 The method for producing NK
US20120171161A1 (en) 2010-12-30 2012-07-05 Sascha Abramson Compositions comprising placental stem cells and platelet rich plasma, and methods of use thereof
US20120171180A1 (en) 2010-12-30 2012-07-05 Sascha Abramson Compositions comprising amnion derived adherent cells and platelet-rich plasma
AU2011352154A1 (en) 2010-12-30 2013-07-18 Anthrogenesis Corporation Methods for cryopreserving and encapsulating cells
WO2012092485A1 (en) 2010-12-31 2012-07-05 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
MX357749B (en) 2011-06-01 2018-07-23 Anthrogenesis Corp Treatment of pain using placental stem cells.

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006111706A1 (en) * 2005-04-16 2006-10-26 Axordia Limited Cytotrophoblast stem cell
WO2007047468A2 (en) * 2005-10-13 2007-04-26 Anthrogenesis Corporation Immunomodulation using placental stem cells

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
ANKER IN'T P ET AL: "Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta" STEM CELLS, ALPHAMED PRESS, DAYTON, OH, US, vol. 22, no. 7, 2004, pages 1338-1345, XP002371702 ISSN: 1066-5099 *
CLARK DAVID A ET AL: "Placental trophoblast from successful human pregnancies expresses the tolerance signaling molecule, CD200 (OX-2)" AMERICAN JOURNAL OF REPRODUCTIVE IMMUNOLOGY, MUNKSGAARD INTERNATIONAL PUBLISHERS, COPENHAGEN, DK, vol. 50, no. 3, September 2003 (2003-09), pages 187-195, XP002430047 ISSN: 1046-7408 *
DATABASE WPI Week 200357 Derwent Publications Ltd., London, GB; AN 2003-599015 XP002443363 & CN 1 407 088 A (ZHOU S) 2 April 2003 (2003-04-02) *
DATABASE WPI Week 200676 Derwent Publications Ltd., London, GB; AN 2006-730789 XP002443364 & CN 1 786 154 A (TIANJIN AGSAI CELL GENE ENG CO LTD) 14 June 2006 (2006-06-14) *
FRANK H G ET AL: "Cell culture models of human trophoblast: primary culture of trophoblast--a workshop report." PLACENTA APR 2001, vol. 22 Suppl A, April 2001 (2001-04), pages S107-S109, XP002443188 ISSN: 0143-4004 *
HARBACHEUSKI RYHOR ET AL: "Placenta derived adherent cells (PDACs) suppress tumor cells of diverse origin." BLOOD, vol. 108, no. 11, Part 2, November 2006 (2006-11), page 288B, XP009084838 & SYMPOSIUM OF THE INTERNATIONAL-SOCIETY-OF-MOLECULAR-EVOLUTI ON; GUANANACASTE, COSTA RICA; JANUARY 08 -12, 2001 ISSN: 0006-4971 *
HOEK R M ET AL: "Down-regulation of the macrophage lineage through interaction with OX2 (CD200)" SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE,, US, vol. 290, no. 5497, 1 December 2000 (2000-12-01), pages 1768-1771, XP002263649 ISSN: 0036-8075 *
LI CHANG DONG ET AL: "MESENCHYMAL STEM CELLS DERIVED FROM HUMAN PLACENTA SUPPRESS ALLOGENEIC UMBILICAL CORD BLOOD LYMPHOCYTE PROLIFERATION" CELL RESEARCH - XIBAO YANJIU, BEIJING, CN, vol. 15, no. 7, July 2005 (2005-07), pages 539-547, XP009080356 ISSN: 1001-0602 *
LIN YI ET AL: "Murine CD200(+)CK7(+) trophoblasts in a poly (I : C)-induced embryo resorption model" REPRODUCTION (CAMBRIDGE), vol. 130, no. 4, October 2005 (2005-10), pages 529-537, XP002443406 ISSN: 1470-1626 *
PALUDAN CASPER ET AL: "Immune suppression by placenta derived adherent cells (PDAC) correlate with monocyte chemoattractant protein-1 and IL-2 secretion." BLOOD, vol. 108, no. 11, Part 2, November 2006 (2006-11), page 48B, XP009084830 & SYMPOSIUM OF THE INTERNATIONAL-SOCIETY-OF-MOLECULAR-EVOLUTI ON; GUANANACASTE, COSTA RICA; JANUARY 08 -12, 2001 ISSN: 0006-4971 *
YEN B LINJU ET AL: "Isolation of multipotent cells from human term placenta." STEM CELLS (DAYTON, OHIO) 2005, vol. 23, no. 1, January 2005 (2005-01), pages 3-9, XP002443187 ISSN: 1066-5099 *
ZHANG YI ET AL: "Human placenta-derived mesenchymal progenitor cells support culture expansion of long-term culture-initiating cells from cord blood CD34+ cells" EXPERIMENTAL HEMATOLOGY, NEW YORK, NY, US, vol. 32, no. 7, July 2004 (2004-07), pages 657-664, XP002389863 ISSN: 0301-472X *

Cited By (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2305795B1 (en) * 2000-12-06 2019-07-03 Celularity, Inc. Method of collecting placental stem cells
EP3299452A1 (en) * 2000-12-06 2018-03-28 Anthrogenesis Corporation Method of collecting placental stem cells
US10383897B2 (en) 2005-12-29 2019-08-20 Celularity, Inc. Placental stem cell populations
EP2471904A2 (en) 2005-12-29 2012-07-04 Anthrogenesis Corporation Placental stem cell populations
US9078898B2 (en) 2005-12-29 2015-07-14 Anthrogenesis Corporation Placental stem cell populations
US9364585B2 (en) 2006-05-11 2016-06-14 Anthrogenesis Corporation Methods for collecting and using placenta cord blood stem cells
EP2023717A2 (en) * 2006-05-11 2009-02-18 Naoko Takebe Methods for collecting and using placenta cord blood stem cells
EP2023717A4 (en) * 2006-05-11 2009-12-16 Naoko Takebe Methods for collecting and using placenta cord blood stem cells
EP2705848A1 (en) * 2006-08-04 2014-03-12 Anthrogenesis Corporation Tumor suppression using placental stem cells
AU2007281920B2 (en) * 2006-08-04 2014-01-23 Celularity Inc. Tumor suppression using placental stem cells
WO2008019148A3 (en) * 2006-08-04 2008-08-07 Anthrogenesis Corp Tumor suppression using placental stem cells
WO2008019148A2 (en) * 2006-08-04 2008-02-14 Anthrogenesis Corporation Tumor suppression using placental stem cells
EP2420567A3 (en) * 2006-10-23 2015-09-30 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
WO2008051568A3 (en) * 2006-10-23 2008-07-24 Anthrogenesis Corp Methods and compositions for treatment of bone defects with placental cell populations
US10105399B2 (en) 2006-10-23 2018-10-23 Celularity, Inc. Methods and compositions for treatment of bone defects with placental cell populations
EP3483263A1 (en) * 2006-10-23 2019-05-15 Celularity, Inc. Methods and compositions for treatment of bone defects with placental cell populations
WO2008051568A2 (en) * 2006-10-23 2008-05-02 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
EP2630959A1 (en) * 2007-02-12 2013-08-28 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
EP2687220A3 (en) * 2007-02-12 2014-05-14 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
EP2687220A2 (en) * 2007-02-12 2014-01-22 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
WO2008100497A1 (en) * 2007-02-12 2008-08-21 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
WO2008100498A3 (en) * 2007-02-12 2008-10-30 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
WO2008100498A2 (en) * 2007-02-12 2008-08-21 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US10494607B2 (en) 2007-02-12 2019-12-03 Celularity, Inc. CD34+,CD45−placental stem cell-enriched cell populations
EP2915537A3 (en) * 2007-02-12 2015-10-28 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
EP3763376A1 (en) * 2007-02-12 2021-01-13 Celularity, Inc. Treatment of inflammatory diseases using placental stem cells
EP3103462A1 (en) * 2007-02-12 2016-12-14 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
WO2009010885A3 (en) * 2007-07-13 2009-03-12 Inst Nat Sante Rech Med Use of cd200 as a mesenchymal stem cells marker
WO2009010885A2 (en) * 2007-07-13 2009-01-22 Institut National De La Sante Et De La Recherche M Use of cd200 as a mesenchymal stem cells marker
EP2014294A1 (en) * 2007-07-13 2009-01-14 Institut National De La Sante Et De La Recherche Medicale (Inserm) Use of CD200 as a mesenchymal stem cells marker
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
EP3103463B1 (en) 2007-09-19 2020-01-01 Pluristem Ltd. Adherent cells from adipose or placenta tissues and use thereof in therapy
JP2017002071A (en) * 2007-09-26 2017-01-05 アンスロジェネシス コーポレーション Angiogenic cells from human placental perfusate
JP2018172425A (en) * 2007-09-26 2018-11-08 アンスロジェネシス コーポレーション Angiogenic cells derived from human placental perfusate
WO2009061447A2 (en) * 2007-11-07 2009-05-14 Anthrogenesis Corporation Treatment of premature birth complications
JP2015120713A (en) * 2007-11-07 2015-07-02 アンスロジェネシス コーポレーション Methods for treating complications of premature birth
CN106214702A (en) * 2007-11-07 2016-12-14 人类起源公司 Cord blood purposes in treatment premature labor complication
EP3345609A1 (en) * 2007-11-07 2018-07-11 Anthrogenesis Corporation Use of umbilical cord blood in the treatment of premature birth complications
JP2011503064A (en) * 2007-11-07 2011-01-27 アンスロジェネシス コーポレーション Treatment of preterm complications
WO2009061447A3 (en) * 2007-11-07 2010-01-21 Anthrogenesis Corporation Use of umbilical cord blood in the treatment of premature birth complications
EP2205724A4 (en) * 2007-11-09 2011-01-05 Rnl Bio Co Ltd Method for isolating and culturing adult stem cells derived from human amniotic epithelium
JP2010537663A (en) * 2007-11-09 2010-12-09 アールエヌエル バイオ カンパニー リミテッド Method for isolating and culturing adult stem cells derived from human amnion epithelium
WO2009061024A1 (en) 2007-11-09 2009-05-14 Rnl Bio Co., Ltd Method for isolating and culturing adult stem cells derived from human amniotic epithelium
US8455251B2 (en) 2007-11-09 2013-06-04 Rnl Bio Co., Ltd. Method for isolating and culturing adult stem cells derived from human amniotic epithelium
EP2205724A1 (en) * 2007-11-09 2010-07-14 RNL Bio Co., Ltd. Method for isolating and culturing adult stem cells derived from human amniotic epithelium
EP2727998A3 (en) * 2008-04-21 2014-08-13 Viacyte, Inc. Methods for purifying pancreatic endoderm cells derived from human embryonic stem cells
US20190261623A1 (en) * 2008-08-20 2019-08-29 Celularity, Inc. Cell composition and methods of making the same
EP3172963A1 (en) * 2008-08-20 2017-05-31 Anthrogenesis Corporation Improved cell composition and methods of making the same
JP2015232000A (en) * 2008-08-20 2015-12-24 アンスロジェネシス コーポレーション Improved cell compositions, and methods of making the same
CN102186338B (en) * 2008-08-20 2016-03-16 人类起源公司 The cell composition improved and prepare the method for described composition
JP2012500021A (en) * 2008-08-20 2012-01-05 アンスロジェネシス コーポレーション Improved cell composition and method for producing the same
EP3539380A3 (en) * 2008-08-20 2019-12-18 Celularity, Inc. Improved cell composition and methods of making the same
JP2018138565A (en) * 2008-08-20 2018-09-06 アンスロジェネシス コーポレーション Improved cell composition and methods of making the same
CN102186338A (en) * 2008-08-20 2011-09-14 人类起源公司 Improved cell composition and methods of making the same
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US20210289769A1 (en) * 2008-08-20 2021-09-23 Celularity Inc. Cell composition and methods of making the same
JP2012500792A (en) * 2008-08-22 2012-01-12 アンスロジェネシス コーポレーション Methods and compositions for the treatment of bone defects with placental cell populations
WO2010040262A1 (en) * 2008-10-10 2010-04-15 深圳市嘉天源生物科技有限公司 Methods for isolating animal embryonic mesenchymal stem cells and extracting secretion substance thereof
US20100272694A1 (en) * 2008-10-17 2010-10-28 Affiliated Hospital Of Ningxia Medical University Clinic compliant method for banking human placental mesenchymal cells
WO2010141654A1 (en) * 2009-06-05 2010-12-09 Anthrogenesis Corporation Improved method of collecting placental cells
WO2011010966A1 (en) * 2009-07-23 2011-01-27 Agency For Science, Technology And Research (A*Star) Pre-natal mesenchymal stem cells
US10258650B2 (en) 2010-02-18 2019-04-16 Osiris Therapeutics, Inc. Methods of manufacture of immunocompatible chorionic membrane products
US9956248B2 (en) 2010-02-18 2018-05-01 Osiris Therapeutics, Inc. Methods of manufacture of therapeutic products comprising vitalized placental dispersions
US10646519B2 (en) 2010-02-18 2020-05-12 Osiris Therapeutics, Inc. Methods of manufacture of therapeutic products comprising vitalized placental dispersions
US11638725B2 (en) 2010-02-18 2023-05-02 Osiris Therapeutics, Inc. Methods of manufacture of immunocompatible chorionic membrane products
US11590172B2 (en) 2010-02-18 2023-02-28 Osiris Therapeutics, Inc. Immunocompatible chorionic membrane products
US11207353B2 (en) 2010-02-18 2021-12-28 Osiris Therapeutics, Inc. Immunocompatible amniotic membrane products
US11510947B2 (en) 2010-02-18 2022-11-29 Osiris Therapeutics, Inc. Methods of manufacture of immunocompatible amniotic membrane products
US10576104B2 (en) 2010-02-18 2020-03-03 Osiris Therapeutics, Inc. Methods of manufacture of immunocompatible amniotic membrane products
US10272116B2 (en) 2010-02-18 2019-04-30 Osiris Therapeutics, Inc. Immunocompatible amniotic membrane products
US20140219970A1 (en) * 2010-04-08 2014-08-07 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
EP2651451A1 (en) * 2010-12-17 2013-10-23 Anthrogenesis Corporation Treatment of spinal cord injury and traumatic brain injury using placental stem cells
EP2651451A4 (en) * 2010-12-17 2014-05-21 Anthrogenesis Corp Treatment of spinal cord injury and traumatic brain injury using placental stem cells
WO2012092458A3 (en) * 2010-12-30 2012-08-23 Anthrogenesis Corporation Compositions comprising placental stem cells and platelet rich plasma, and methods of use thereof
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
JP2017137324A (en) * 2011-06-01 2017-08-10 アントフロゲネシス コーポレーション Treatment of pain using placental stem cells
US11090339B2 (en) 2011-06-01 2021-08-17 Celularity Inc. Treatment of pain using placental stem cells
CN102676451A (en) * 2011-09-05 2012-09-19 博雅干细胞科技有限公司 Method for separating mesenchymal stem cells from placenta
US20180237751A1 (en) * 2011-10-11 2018-08-23 The Trustees Of Columbia University In The City Of New York Method for generating beta cells
US20140242038A1 (en) * 2011-10-11 2014-08-28 The Trustees Of Columbia University In The City Of New York Method for generating beta cells
EP2773746A4 (en) * 2011-11-01 2015-08-05 Neostem Inc Adult mesenchymal stem cell (msc) compositions and methods for preparing the same
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
EP2968418A4 (en) * 2013-03-15 2017-02-22 Nutech Medical, Inc. Preparations derived from placental materials of making and using same
EP3068432A4 (en) * 2013-11-15 2017-04-19 Anthrogenesis Corporation Compositions comprising human placental perfusate cells, subpopulations thereof, and their uses
WO2015073800A2 (en) 2013-11-15 2015-05-21 Anthrogenesis Corporation Compositions comprising human placental perfusate cells, subpopulations thereof, and their uses
EP3214171A4 (en) * 2014-10-29 2018-07-11 Sungkwang Medical Foundation Placenta-derived cells excreting c3 or c1r complement and composition containing same
WO2020194307A1 (en) * 2019-03-27 2020-10-01 Pluristem Ltd. Methods and compositions for aesthetic and cosmetic treatment and stimulating hair growth

Also Published As

Publication number Publication date
US9078898B2 (en) 2015-07-14
DK2471903T3 (en) 2018-05-28
CA2635253C (en) 2017-03-14
KR20080081083A (en) 2008-09-05
AU2006332678B2 (en) 2013-12-05
EP1976977B1 (en) 2015-07-08
PL2471906T3 (en) 2019-09-30
PT2471906T (en) 2019-02-06
EP2471903A2 (en) 2012-07-04
DK2471906T3 (en) 2019-02-04
CN103525758A (en) 2014-01-22
ES2711278T3 (en) 2019-04-30
KR20230091193A (en) 2023-06-22
PT2471903T (en) 2018-05-18
JP5550235B2 (en) 2014-07-16
JP2009521929A (en) 2009-06-11
IL223194A (en) 2015-05-31
AU2006332678A1 (en) 2007-07-12
US20070275362A1 (en) 2007-11-29
CA2957336C (en) 2023-09-26
EP2471904A2 (en) 2012-07-04
PL2471903T3 (en) 2018-09-28
EP2471907A3 (en) 2012-10-31
ZA201004182B (en) 2013-02-27
NZ595786A (en) 2013-05-31
MX2018012512A (en) 2020-11-12
KR20190104428A (en) 2019-09-09
EP2471907A2 (en) 2012-07-04
MX338534B (en) 2016-04-21
PT2471905T (en) 2019-01-11
NZ622377A (en) 2015-10-30
CN101395266B (en) 2018-06-15
WO2007079183A3 (en) 2007-11-15
US20160129048A1 (en) 2016-05-12
US8057788B2 (en) 2011-11-15
HK1184817A1 (en) 2014-01-30
JP2013099343A (en) 2013-05-23
IL243876A0 (en) 2016-04-21
ES2671355T3 (en) 2018-06-06
PL2471904T3 (en) 2019-08-30
CN103146640A (en) 2013-06-12
EP2471907B1 (en) 2018-09-26
EP2471905B1 (en) 2018-09-05
IL223156A0 (en) 2012-12-31
MX2020001944A (en) 2020-07-13
KR20180105266A (en) 2018-09-27
MX348414B (en) 2017-06-12
EP3486314B1 (en) 2023-03-22
EP3486314A3 (en) 2019-08-07
KR101900809B1 (en) 2018-09-20
EP2471905A2 (en) 2012-07-04
PL2471907T3 (en) 2019-07-31
ES2549111T3 (en) 2015-10-23
US20130028871A1 (en) 2013-01-31
IL266958A (en) 2019-07-31
ZA201703181B (en) 2021-08-25
US20100297689A1 (en) 2010-11-25
EP2471903A3 (en) 2013-02-27
EP2471903B1 (en) 2018-02-14
US8691217B2 (en) 2014-04-08
EP2471904B1 (en) 2018-10-24
CN101395266A (en) 2009-03-25
KR20210122908A (en) 2021-10-12
NZ712789A (en) 2017-06-30
ES2703502T3 (en) 2019-03-11
IL223194A0 (en) 2012-12-31
IL243876B (en) 2019-06-30
PT2471907T (en) 2019-01-28
CN103146640B (en) 2015-09-09
ES2708933T3 (en) 2019-04-12
PL1976977T3 (en) 2015-12-31
JP5739918B2 (en) 2015-06-24
PT2471904T (en) 2019-02-25
US20130022581A1 (en) 2013-01-24
US8202703B2 (en) 2012-06-19
CN103060263A (en) 2013-04-24
KR101722577B1 (en) 2017-04-03
EP2471905A3 (en) 2012-10-31
EP2471906B1 (en) 2018-10-10
KR20220063304A (en) 2022-05-17
PL2471905T3 (en) 2019-08-30
DK2471904T3 (en) 2019-02-18
EP2471906A2 (en) 2012-07-04
US20080032401A1 (en) 2008-02-07
KR20150092353A (en) 2015-08-12
PE20080146A1 (en) 2008-02-11
KR101585241B1 (en) 2016-01-13
EP3486314A2 (en) 2019-05-22
EP2471904A3 (en) 2012-10-31
NZ569562A (en) 2012-04-27
CN103060263B (en) 2016-03-16
ES2706726T3 (en) 2019-04-01
EP2471906A3 (en) 2012-10-31
US8591883B2 (en) 2013-11-26
DK2471907T3 (en) 2019-01-21
CA2635253A1 (en) 2007-07-12
US20140377230A1 (en) 2014-12-25
CN103525758B (en) 2018-02-13
KR20170037689A (en) 2017-04-04
CN108559725A (en) 2018-09-21
KR20200123283A (en) 2020-10-28
CA2957336A1 (en) 2007-07-12
IL192457A0 (en) 2009-02-11
DK2471905T3 (en) 2019-01-02
EP1976977A2 (en) 2008-10-08
US10383897B2 (en) 2019-08-20

Similar Documents

Publication Publication Date Title
US10383897B2 (en) Placental stem cell populations
US20150037316A1 (en) Placental Stem Cell Populations
AU2022201955A1 (en) Placental stem cell populations

Legal Events

Date Code Title Description
DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2635253

Country of ref document: CA

Ref document number: MX/a/2008/008365

Country of ref document: MX

Ref document number: 2006332678

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2008548745

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 569562

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2006332678

Country of ref document: AU

Date of ref document: 20061228

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020087018508

Country of ref document: KR

Ref document number: 2006848281

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200680053575.3

Country of ref document: CN

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 223157

Country of ref document: IL

Ref document number: 223156

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 223194

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 1020157020367

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 243876

Country of ref document: IL