CA2530422C - Postpartum-derived cells for use in treatment of disease of the heart and circulatory system - Google Patents

Postpartum-derived cells for use in treatment of disease of the heart and circulatory system Download PDF

Info

Publication number
CA2530422C
CA2530422C CA2530422A CA2530422A CA2530422C CA 2530422 C CA2530422 C CA 2530422C CA 2530422 A CA2530422 A CA 2530422A CA 2530422 A CA2530422 A CA 2530422A CA 2530422 C CA2530422 C CA 2530422C
Authority
CA
Canada
Prior art keywords
cells
cell
pharmaceutical composition
derived
umbilical
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2530422A
Other languages
French (fr)
Other versions
CA2530422A1 (en
Inventor
Ian Ross Harris
Alexander M. Harmon
Anthony J. Kihm
Darin J. Messina
Sanjay Mistry
Agnieszka Seyda
Chin-Feng Yi
Anna Gosiewska
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DePuy Synthes Products Inc
Original Assignee
DePuy Synthes Products Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by DePuy Synthes Products Inc filed Critical DePuy Synthes Products Inc
Publication of CA2530422A1 publication Critical patent/CA2530422A1/en
Application granted granted Critical
Publication of CA2530422C publication Critical patent/CA2530422C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical 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
    • A61K35/12Materials 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
    • 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
    • A61K38/18Growth factors; Growth regulators
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1808Epidermal growth factor [EGF] urogastrone
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1833Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • 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
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1891Angiogenesic factors; Angiogenin
    • 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
    • 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
    • A61K38/20Interleukins [IL]
    • A61K38/204IL-6
    • 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
    • A61K38/20Interleukins [IL]
    • A61K38/2053IL-8
    • 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
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • 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
    • 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
    • C12N2500/32Amino acids
    • 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
    • C12N2500/34Sugars
    • 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
    • C12N2500/44Thiols, e.g. mercaptoethanol
    • 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
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • C12N2500/95Protein-free medium and culture conditions
    • 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
    • C12N2501/12Hepatocyte growth factor [HGF]
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/21Chemokines, e.g. MIP-1, MIP-2, RANTES, MCP, PF-4
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/02Coculture with; Conditioned medium produced by embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/03Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from non-embryonic pluripotent stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins

Abstract

Cells derived from postpartum tissue and methods for their use in treatment of diseases of the heart or circulatory system are disclosed. Cells may be used in either differentiated or undifferentiated forms, in homogenous cultures, or as populations with other cells, and in conjunction with other bioactive factors.

Description

c'3.174 .)=-t I I' POSTPARTUM-DERIVED CELLS FOR USE IN TREATMENT OF DISEASE OF
THE HEART AND CIRCULATORY SYSTEM
FIELD OF THE INVENTION
The invention relates to postpartum-derived cells that have the potential to divide, and to differentiate along mesenchymal lineage, towards cardiomyogenic, angiogenic and vasculogenic phenotypes. The invention also relates to methods for the use of such postpartum-derived cells in therapeutic treatment of diseases of the heart and circulatory system.
BACKGROUND OF THE INVENTION

It is generally recognized that the cells which comprise the heart muscle in mammals are post-mitotic. This leads to difficulties in injured or diseased heart muscles, which are largely unable to repair damaged cells that become necrotic. After such damage, the efficiency and output of the heart muscle are decreased, placing additional stress on the heart, leading to further damage and necrosis, and ultimately to heart failure.
The downward spiral from healthy heart to failing heart can result from a number of conditions including physical injury, heart disease, including congenital heart disease, and infections of the heart or circulatory tissue. Diseases of the heart and circulatory system are often ultimately fatal, particularly conditions which result in heart failure, for example cardiomyopathies. At present there is no cure for most such conditions and many patients require, for example, ventricular assist devices and eventually heart transplants.
Presently, there is interest in using either stem cells, which can divide and differentiate, or muscles cells from other sources, including smooth and skeletal muscles cells, to assist the heart to repair or reverse tissue damage, restore function, or to at least halt the damage cycle leading to further loss of healthy heart tissue. In addition many circulatory diseases and injuries involve chronic or acute damage to, or necrosis of, circulatory tissues, and the cells of which such tissues are comprised. Cell-based and cell-derived therapies are of interest for these conditions.
There is thus a need in the art for cell-based, or cell-derived therapies which can aid in healing damaged heart or circulatory tissues, or which can result in the repair or replacement of such damage in a patient.
SUMMARY OF THE INVENTION
The invention, in one of its aspects is generally directed to isolated postpartum-derived cells which are derived from postpartum tissue which is substantially free of blood and which is capable of self-renewal and expansion in culture and having the potential to differentiate along mesenchymal lineage, towards cardiomyogenic, angiogenic and vasculogenic phenotypes, and further towards cells such as cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g. vascular smooth muscle cells), as well as cells of the excitatory and conductive systems, and progenitors or more primitive relatives of the foregoing. The invention, in other aspects, is directed to populations of cells comprising such cells, and therapeutic cell compositions, as well as methods of using the populations of cells for therapeutic treatment of cardiac or circulatory damage or disease
- 2 -= = In a first aspect, the invention provides isolated postpartum-derived cells comprising L-valine-requiring cells derived from postpartum tissue substantially free of blood. The cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes; for example cardiomyocytes, or their progenitors. The cells are capable of growth in atmospheres containing oxygen from about 5% to at least about 20%
and comprise at least one of the following characteristics:
the potential for at least about 40 doublings in culture;
attachment and expansion on a coated or uncoated tissue culture vessel, wherein a coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin, or fibronectin;
production of at least one of tissue factor, virnentin, and alpha-smooth muscle actin;
production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and BLA-DR,DP,DQ, as detected by flow cytometry;
expression of at least one of interleukin 8; reticulon 1; chemokine (C-X-C
motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3;
expression of at least one of C-type (calcium dependent, carbohydrate-recognition domain) lectin, superfamily member 2 (activation-induced); Wilms tumor 1;
aldehyde dehycirogenase 1 family, member A2; and renin; oxidized low density lipoprotein (lectin-like) receptor 1; Honzo sapiens, clone IMAGE:4179671, mRNA, partial cds; protein Icinase C, zeta;
hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; Homo sapiens mRNA;
cDNA DKFZp547K1113 (from clone DKFZp547K1113);
expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, is reduced for at least one of: short stature homeobox 2; heat shock 271cDa protein 2; chemokine (C-X-C motif) ligand 12 (stoma' cell-derived factor 1);
elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein;
similar to neuralin 1; tetTanectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-
- 3 -related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha;
procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila);
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7;
DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3;
distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus BiB 19kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit Vila polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
insulin-like growth factor binding protein 2, 36kDa;
secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MTPla, RANTES, and TIMPl; and lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP lb, 1309, MDC, and VEGF, as detected by ELISA.
In certain embodiments, the postpartum-derived cell is an umbilicus-derived cell. In other embodiments it is a placenta-derived cell. In specific embodiments, the cell has all identifying features of any one of: cell type PLA 071003 (P8) (ATCC Accession No. PTA-6074); cell type PLA 071003 (P11) (ATCC Accession No. PTA-6075); cell type PLA

(P16) (ATCC Accession No. PTA-6079); cell type UMB 022803 (P7) (ATCC Accession No.
PTA-6067); or cell type LIMB 022803 (P17) (ATCC Accession No. PTA-6068).
The postpartum-derived cells are preferably isolated in the presence of one or more enzyme activities such as metalloprotease activity, neutral protease activity and mucolytic enzyme activity. The cells preferably comprise a normal karyotype, which is maintained as the cells are passaged.
- 4 -Preferred cells produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C, and do not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
In another of its several aspects, the invention provides populations of cells comprising the cells described above. In certain embodiments the populations are incubated in the presence of one or more factors which stimulate stem cell differentiation along a cardiogenic pathway or lineage. In other embodiments, cells are incubatedin the presence of compounds which tend to stimulate differentiation along angiogenic, hemangiogenic, and vasculogenic pathways, or towards more committed cells such as cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g.
vascular smooth muscle cells), as well as cells of the excitatory and conductive systems, and progenitors of any of the foregoing, for example, myoblasts, cardiomyoblasts, hemangioblasts, angioblasts and the like or their progenitors. The populations can be provided therapeutically to a patient, for example with heart or circulatory disease, such as a cardiomyopathy, or with a cardiac injury, such as from myocardial infarction, or with any damage or disease of the heart or circulatory system. In presently preferred embodiments, the population comprises about 50% postpartum-derived cells, while in other preferred embodiments the population is a substantially homogeneous population of postpartum-derived cells.
The invention provides in another of its aspects therapeutic cell compositions comprising a pharmaceutically-acceptable carrier and postpartum-derived cells derived from human postpartum tissue substantially free of blood. The cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes; for example smooth or skeletal muscles phenotypes. In preferred embodiments, cells can differentiate along pathways leading to phenotypes of cardiomyocytes, cells of the endothelium, myocardium, epicardium, vascular endothelium, as well as smooth muscle cells (e.g. vascular smooth muscle cells), and cells of the excitatory and conductive systems (e.g.
Purkinje cells), and progenitors of the foregoing.. The cells are capable of growth in an atmosphere containing oxygen from about 5% to at least about 20%. The cells also require L-valine for growth; have the potential for at least about 40 doublings in culture; attach and expand on a coated or uncoated_ tissue culture vessel, wherein a coated tissue culture vessel is coated with gelatin, laminin, or fibronectin; produce tissue factor, vimentin, and alpha-smooth muscle actin;
produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C; and do not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
- 5 -C1471 -q = The therapeutic cell compositions provided can be provided therapeutically in a patient with a cardiomyopathy, or other heart disease, or a cardiac injury, or any disease of the circulatory system which, for example, involves cell injury or tissue/cell necrosis. In certain embodiments, the therapeutic cell compositions comprise cells induced to differentiate along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway or lineage.
The therapeutic cell compositions can comprise cells or cellular products that stimulate adult stem cells naturally present in the heart, blood, blood vessels or the like to divide, or differentiate, or both.
Preferably the therapeutic cell compositions can at least survive, grow, stimulate in growth, stimulate angiogenesis or vasculogenesis, but in certain cases even nonviable cells such as senescent cells will be therapeutic, and in some embodiments, even dead cells, or fractions thereof will provide therapeutic improvements for a patient.
The therapeutic cell compositions are provided, for example, by injection. In certain preferred embodiments, the therapeutic cell compositions are provided by intracardiac injection.
In other embodiments, they are placed adjacent ot an inner or outer aspect of the cardiac muscles directly, or on a matrix as a cell-matrix complex. The therapeutic cell compositions provided in the form of a matrix-cell complex comprise matrices including, for example, biocompatible scaffolds, lattices, self-assembling structures and the like, whether bioabsorbable or not, liquid or solid. Many such matrices are known to those of skill in the arts of tissue engineering, wound healing, and the like. Thereapeutic compositions can also comprise additional biological compounds or phararnaceuticals that may improve the therapeutic value to the patient, and the compositions can also comprise one or more additional cells or cell types.
The therapeutic cell compositions, in certain embodiments also comprise cells that:
express at least one of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand
6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3;
express at least one of C-type.(calcium dependent, carbohydrate-recognition domain) lectin, superfamily member 2 (activation-induced); Wilms tumor 1; aldehyde dehydrogenase 1 family, member A2; and renin; oxidized low density lipoprotein (lectin-like) receptor 1; Homo sapiens, clone IMAGE:4179671, mRNA, partial cds; protein lcinase C, zeta;
hypothetical protein bKFZp564F013; downregulated in ovarian cancer 1; Homo sapiens mRNA; and cDNA
DKFZp547K1113 (from clone DKFZp547K1113);
have reduced expression, relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, for at least one of: short stature homeobox 2; heat shock 27kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA;
cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein;
similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha;
procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila);
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha
7;
DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3;
distal-less , homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKE7p564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus BiB 19kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit Vila polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
insulin-like growth factor binding protein 2, 361cDa;
secrete at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIPla, RANTES, and TIMPl; and do not secrete at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
In yet another of its aspects, the invention provides methods for treating a patient with a disease or injury of the heart or circulatory system comprising administering a therapeutic postpartum-derived cell composition to a patient with a disease or injury of the heart or circulatory system; and evaluating the patient for improvements, for example in cardiac function.

In certain preferred embodiments the disease is a cardiomyopathy, either idiopathic or with a known cause, and either ischemic or nonischemic in nature.
Measurement of improvement include any means known in the art, but preferred improvements include improvements in hemo dynamic measurements including but not limited to chest cardiac output (CO), cardiac index (CI), pulmonary artery wedge pressures (PAWP), and cardiac index (CI), % fractional shortening (%FS), ejection fraction (EF), left ventricular ejection fraction (LVEF); left ventricular end diastolic diameter (LVEDD), left ventricular end systolic diameter (LVESD), contractility (e.g. dP/dt), pressure-volume loops, measurements of cardiac work, as well as an increase in atrial or ventricular functioning; an increase in pumping efficiency, a decrease in the rate of loss of pumping efficiency, a decrease in loss of hemodynamic functioning; and a decrease in complications associated with cardiomyopathy.
In some presently preferred embodiments, the method comprises inducing the therapeutic postpartum-derived cells to differentiate along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway or or a towards cells or progenitors of cells such as cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g. vascular smooth muscle cells), as well as cells of the excitatory and conductive systems. Cells which differentiate towards myoblasts,cardiomyoblasts, angioblasts, hemangioblasts, and the like are contemplated for use herein.
The administering is preferably in vivo by transplanting, grafting, implanting, injecting, fusing, delivering via catheter, or providing as a matrix-cell complex, or any other means known in the art for providing cell therapy. For many applications, the cells can simply be injected, for example intravenously, and the cells will locate or home in accordance with their commitment in the direction of a phenotype associated with a particular tissue. For example, cells differentiated in a cardiomyogenic lineage may home in on the cardiac muscle when injected intravenously anywhere in the body.
The invention also provides in another aspect, methods for treating a patient with a disease or injury of the heart or circulatory system comprising administering a therapeutic postpartum-derived cell composition to a patient with a disease or injury of the heart or circulatory system; and evaluating the patient for improvements in cardiac function, wherein the administering is with a population of another cell type. Administration of cocultures, mixed populations or other nonclonal populations are sometimes preferred. Other cell types which can be coadministered are stem cells in certain embodiments, while in others myoblasts, myocytes, cardiomyoblasts, cardiomyocytes, angioblasts, hemangioblasts or a progenitors of myoblasts, myocytes, cardiomyoblasts, angioblasts, hemangioblasts, or cardiomyocytes are used. Other
- 8 -angiogenic, hemangiogenic, and vasculogenic cells, or more commited cells from such lineages are also suitable for coadministration.
Also provided herein are methods for treating a patient with a disease or injury of the heart or circulatory system comprising administering a therapeutic postpartum-derived cell composition to a patient with a disease or injury of the heart or circulatory system; and evaluating the patient for improvements in the diseased or injured state, wherein the therapeutic cell compostion is administered as a matrix-cell complex. In certain embodiments, the matrix is a scaffold, preferably bioabsorbable, comprising in addition to the matrix proper, at least the postpartum-derived cells.
Co-cultures comprising the cells or cultures of the invention with other mammalian cells are also provided herein. Preferably these co-cultures comprise another mammalian cell line whose growth or therapeutic potential, for example, is improved by the presence of the umbilicus-derived cells. Human cell lines are particular preferred. Such co-cultures are useful for therapeutic application in vitro or in vivo.
Also provided herein are therapeutic compositions comprising a postpartum-derived cell and another therapeutic agent, factor, or bioactive agent, such as a pharmaceutical compound.
Such bioactive agents include, but are not limited to, IGF, LIF, PDGF, EGF, FGF, as well as antithrombogenic, antiapoptotic agents, anti-inflammatory agents, immunosuppressive or immunomodulatory agents, and antioxidants. Such therapeutic compositions can further comprise one or more additional cell types in addition to the PPDCs and the bioactive component.
In addition to the above, compositions derived from the cells are provided herein. Cell lysates, soluble cell fractions and membrane-enriched cell fractions are provided herein.
Extracellular matrices derived from the cells, for example, fractions comprising basement membranes are also useful and are provided herein.
Compositons of the invention also include conditioned culture media as provided herein.
Such media have first been used to grow the cells or cultures of the invention, which during growth secrete one or more useful products into the medium. Conditioned medium from these novel cells are useful for many purposes, including for example, supporting the growth of other mammalian cells in need of growth factors or trophic factors secreted into the media by the cells and cultures of the invention, and promoting, for example, angiogenesis.
Kits are also provided herein. Various preferred kits are those providing the required components for practicing the several aspects of the inventions, for example kits for growth and maintenance of the cells, kits for isolation of the cells, kits for coculturing the cells, kits for
- 9 -utilizing the cells and culture in vitro and kits for utilizing the cells and cultures in vivo are all provided herein. Kits are also provided for the preparation of cell fractions derived from the cells including for example, cell lysates, soluble and membrane-enriched fractions, and extracellular fractions.
Specific aspects of the invention provide:
a pharmaceutical composition for use in the treatment of a disease of the heart or circulatory system comprising a pharmaceutically acceptable carrier and isolated umbilical-derived cells, wherein the cells are capable of self-renewal and expansion in culture, have the potential to differentiate into a cell of cardiomyocyte phenotypes; and have each of the following characteristics: potential for at least about 40 doublings in culture;
production of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C; lack of production of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; and expression of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3;
use of isolated umbilical-derived cells as described herein, for the treatment of a disease of the heart or circulatory system; and use of isolated umbilical-derived cells as described herein, in the manufacture of a medicament for the treatment of a disease of the heart of circulatory system.
DOCSTOR: 2587929\1
- 10-In another aspect, there is provided use of an effective amount of umbilical-derived cells in the treatment of a disease of the heart or circulatory system in a patient, wherein the cells are derived from umbilical cord tissue substantially free of blood, wherein the cells are capable of self-renewal and expansion in culture, have the potential to differentiate into a cell of cardiomyocyte phenotypes; and have each of the following characteristics: potential for at least about 40 doublings in culture; production of 0D10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C; lack of production of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, WA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; and expression of interleukin 8;
reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2);
chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3.
These and other aspects of the invention are described in more detail below.
DOCSTOR: 2587846\1 -10a -DETAILED DESCRIPTION
Definitions Various terms used throughout the specification and claims are defined as set forth below.
Stern cells are undifferentiated cells defined by the ability of a single cell both to self-renew, and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute substantially to most, if not all, tissues following injection into blastocysts.
Stem cells are classified according to their developmental potential as: (1) totipotent; (2) pluripotent; (3) multipotent; (4) oligopotent; and (5) unipotent. Totipotent cells are able to give rise to all embryonic and extraembryonic cell types. P/uripoteizt cells are able to give rise to all - embryonic cell types. Multipotent cells include those able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem cells (HSC) can produce progeny that include HSC (self-renewal), blood cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood). Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells; and cells that are unipotent are able to give rise to a single cell lineage (e.g., spermatogenic stem cells).
Stem cells are also categorized on the basis of the source from which they may be obtained. An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types. An embryonic stem cell is a pluripotent cell from the inner cell mass of a blastocyst-stage embryo. A fetal stem cell is one that originates from fetal tissues or membranes. A postpartum stern cell is a multipotent or plurinotent cell that - 10b.

originates substantially from extraembryonic tissue available after birth, namely, the placenta and the umbilical cord. These cells have been found to possess features characteristic of pluripotent stem cells, including rapid proliferation and the potential for differentiation into many cell lineages. Postpartum stem cells may be blood-derived (e.g., as are those obtained from umbilical cord blood) or non-blood-derived (e.g., as obtained from the non-blood tissues of the umbilical cord and placenta).
Embryonic tissue is typically defined as tissue originating from the embryo (which in humans refers to the period from fertilization to about six weeks of development. Fetal tissue refers to tissue originating from the fetus, which in humans refers to the period from about six weeks of development to parturition. Extraembryonic tissue is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryoriic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta (which itself forms from the chorion and the maternal decidua basalis).
Differentiation is the process by which an unspecialized ("uncommitted") or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example. A differentiated cell is one that has taken on a more specialized ("committer) position within the lineage of a cell. The term committed, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type. De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell. As used herein, the lineage of a cell defines the heredity of the cell, i.e. which cells it came from and what cells it can give rise to. The lineage of a cell places the cell within a hereditary scheme of development and differentiation.
In a broad sense, a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors.
By that definition, stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells. When referring to the cells of the present invention, as described in greater detail below, this broad definition ofprogenitor cell may be u_sed. In a narrower sense, a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types. This type of progenitor cell is generally not able to self-
- 11 -renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a non-renewing progenitor cell or as an intermediate progenitor or precursor cell.
As used herein, the phrase differentiates into a mesodermal, ectodermal or endodermal lineage refers to a cell that becomes committed to a specific mesodeillial, ectodermal or endodermal lineage, respectively. Examples of cells that differentiate into a mesodermal lineage or give rise to specific mesodermal cells include, but are not limited to, cells that are adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoietic, hemangiogenic, myogenic, nephrogenic, urogenitogenic, osteogenic, pericardiogenic, or stromal. Examples of cells that differentiate into ectodermal lineage include, but are not limited to epidermal cells, neurogenic cells, and neurogliagenic cells. Examples of cells that differentiate into endodennal lineage include, but are not limited to, pleurigenic cells, hepatogenic cells, cells that give rise to the lining of the intestine, and cells that give rise to pancreogenic and splanchogenic cells.
The cells of the present invention are generally referred to as umbilicus-derived cells (or UDCs). They also may sometimes be referred to more generally herein as postpartum-derived cells or postpartum cells (PPDCs). In addition, the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense. The term derived is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a Growth Medium to expand the population and/or to produce a cell line). The in vitro manipulations of umbilical stem cells and the unique features of the umbilicus-derived cells of the present invention are described in detail below.
Various terms are used to describe cells in culture. Cell culture refers generally to cells taken from a living organism and grown under controlled condition ("in culture" or "cultured").
A primal); cell culture is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture. Cells are expanded in culture when they are placed in a Growth Medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells. When cells are expanded in culture, the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as doubling time.
A cell line is a population of cells formed by one or more sub cultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been passaged. A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture. The primary culture, i.e., the first culture following the isolation of cells from tissue, is
-12-designated PO. Following the first subculture, the cells are described as a secondary culture (P1 or passage 1). After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number. The expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and tim_e between passaging.
A conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide trophic support to other cells. Such trophic factors include, but are not limited to hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, and granules. The medium containing the cellular factors is the conditioned medium.
Generally, a trophic factor is defined as a substance that promotes survival, growth, proliferation and /or maturation of a cell, or stimulates increased activity of a cell.
When referring to cultured vertebrate cells, the term senescence (also replicative senescence or cellular senescence) refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's limit). Although cellular senescence was first described using fibroblast-like cells, most normal human cell types that can be grown successfully in culture undergo cellular senescence. The in vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide).
Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone. Thus, cells made quiescent by removing essential growth factors are able to resume growth and division when the growth factors are re-introduced, and thereafter carry out the same number of doublings as equivalent cells grown continuously. Similarly, when cells are frozen in liquid nitrogen after various numbers of population doublings and then thawed and cultured, they undergo substantially the same number of doublings as cells maintained unfrozen in culture. Senescent cells are not dead or dying cells;
they are actually resistant to programmed cell death (apoptosis), and have been maintained in their nondividing state for as long as three years. These cells are very much alive and metabolically active, but they do not divide. The nondividing state of senescent cells has not yet been found to be reversible by any biological, chemical, or viral agent.
- 13 -As used herein, the ten-n Growth Medium generally refers to a medium sufficient for the culturing of umbilicus-derived cells. In particular, one presently preferred medium for the culturing of the cells of the invention in comprises Dulbecco's Modified Essential Media (also abbreviated DMEM herein). Particularly preferred is DMEM-low glucose (also DMEM-LG
herein) (Invitrogen, Carlsbad, CA). The DMEM-low glucose is preferably supplemented with serum, most preferably fetal bovine serum or human serum. Typically, 15% (v/v) fetal bovine serum (e.g. defined fetal bovine serum, Hyclone, Logan UT) is added, along with antibiotics/antimycotics ((preferably 100 Unit/milliliter penicillin, 100 milligrams/milliliter streptomycin, and 0.25 microgram/milliliter amphotericin B; Invitrogen, Carlsbad, CA)), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis MO). In some cases different growth media are used, or different supplementations are provided, and these are normally indicated in the text as supplementations to Growth Medium. In certain chemically-defined media the cells may be grown without serum present at all. In such cases, the cells may require certain growth factors, which can be added to the medium to support and sustain the cells. Presently preferred factors to be added for growth on serum-free media include one or more of bFGF, EGF, IGF-I, and PDGF.
- In more preferred embodiments, two, three or all four of the factors are add to serum free or chemically defined media. In other embodiments, LIF is added to serum-free medium to support or improve growth of the cells.
Also relating to the present invention, the tern standard growth conditions, as used herein refers to culturing of cells at 37 C, in a standard atmosphere comprising 5%
CO2,. Relative humidity is maintained at about 100%. While the foregoing conditions are useful for culturing, it is to be understood that such conditions are capable of being varied by the skilled artisan who will appreciate the options available in the art for culturing cells.
The following abbreviations are used herein:
ANG2 (or Ang2) for angiopoietin 2;
APC for antigen-presenting cells;
BDNF for brain-derived neurotrophic factor;
bFGF for basic fibroblast growth factor;
bid (BID) for "bis in die" (twice per day);
BSP for bone sialoprotein;
TM
C:1) for collagenase and dispase treatment TM
C:D:H for collagenase, dispase, and hyaluronidase treatment CK18 for cytokeratin 18;
CXC ligand 3 for chemokine receptor ligand 3;
- 14 -DAPI for 4' - 6-Diamidino-2-phenylindole-2HC1 DMEM for Dulbecco's Minimal Essential Medium;
DMEM:lg (or DMEM:Lg, DMEM:LG) for DMEM with low glucose;
EDTA for ethylene diamine tetraacetic acid;
EGF for epidermal growth factor;
FAGS for fluorescent activated cell sorting;
FBS for fetal bovine serum;
GCP-2 for granulocyte chemotactic protein 2;
GCP-2 for granulocyte chemotactic protein-2;
GFAP for glial fibrillary acidic protein;
HB-EGF for heparin-binding epidermal growth factor;
HCAEC for Human coronary artery endothelial cells;
HGF for hepatocyte growth factor;
hMSC for Human mesenchymal stem cells;
HNF-1 alpha for hepatocyte-specific transcription factor;
HUVEC for Human umbilical vein endothelial cells;
1309 for a chemokine and the ligand for the CCR8 receptor, responsible for chemoattraction of TH2 type T-cells. 1309 binds to endothelial cells, stimulates chemotaxis and invasion of these cells, and enhances HUVEC differentiation into capillary-like structures in an in vitro Matrigel assay. Furthermore, 1309 is an inducer of angiogenesis in vivo in both the rabbit cornea and the chick chorioallantoic membrane assay (CAM).
IL-6 for interleukin-6;
IL-8 for interleukin-8;
K19 for keratin 19;
K8 for keratin 8;
KGF for keratinocyte growth factor;
MCP-1 for monocyte chemotactic protein 1;
MDC for macrophage-derived chemokine;
MIPlalpha for macrophage inflammatory protein lalpha;
MIPlbeta for macrophage inflammatory protein lbeta;
MSC for mesenchymal stem cells;
NDRI for National Disease Research Interchange in Philadelphia, PA
NHDF for Normal Human Dermal Fibroblasts;
NPE for Neural Progenitor Expansion media;
- 15 -PBMC for Peripheral blood mononuclear cell;
PBS for phosphate buffered saline;
PDGFbb for platelet derived growth factor;
PDGFr/alpha for platelet derived growth factor receptor alpha;
PD-L2 for programmed - death ligand 2;
PO (orpo) for "per os" (by mouth);
PPDC for postpartum-derived cell;
Raines (or RANTES) for regulated on activation, normal T cell expressed and secreted;
rhGDF-5 for recombinant human growth and differentiation factor 5;
SC for subcutaneously;
SDF-lalpha for stromal-derived factor I alpha;
SHH for sonic hedgehog;
SOP for standard operating procedure;
TARC for thymus and activation-regulated chemokine;
TCP for Tissue culture plastic;
TGFbeta2 for transforming growth factor beta2;
TGFbeta-3 for transforming growth factor beta-3;
TIMP1 for tissue inhibitor of matrix metalloproteinase 1;
TPO for thrombopoietin;
TuJ1 for Bill Tubulin;
UDC for umbilicus-derived cell;
VEGF for vascular endothelial growth factor;
vFVF for von Willebrand factor;
alphaFP for alpha-fetoprotein;
Description As summarized above, the invention, in one of its aspects is generally directed to isolated postpartum-derived cells which are derived from postpartum tissue which is substantially free of blood and which are capable of self-renewal and expansion in culture and having the potential to differentiate along mesenchymal lineage, towards cardiomyogenic, angiogenic and vasculogenic phenotypes, and further towards cells such as cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g.
vascular smooth
- 16 muscle cells), as well as cells of the excitatory and conductive systems, and progenitors of the foregoing.. Other aspects provide populations comprising such cells, therapeutic cell compositions, and methods of using the therapeutic cell compositions for treatment of patients with injury or disease of the heart or circulatory system. The postpartum-derived cells have been characterized by their growth properties in culture, by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
In a first aspect, the invention provides isolated postpartum-derived cells comprising L-valine-requiring cells derived from mammalian postpartum tissue substantially free of blood.
The cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes; for example cardiomyocytes, or their progenitors.
Cells may be isolated from postpartum tissue, for example umbilicus or placenta, of any mammal of interest by the techniques provided herein. Human cells are presently preferred. The cells can be grown under a wide range of conditions, including a wide variety of culture media, and environmental conditions. The cells can be grown at least from about 35 C to about 39 C, and possibly a wider range depending on other conditions. The cells can be grown in chemically-defined media, or in medium with added mammalian serum, for example fetal bovine serum.
The cells also tolerate cryopreservation at various stages. Cells can maintained frozen, or banked at temperatures preferably below -80 C for long periods. Temperature below -90 C are also preferred and can be attained by specialized electric freezers. Temperature of -180 C and below are also preferred and can be attained in liquid- or vapor-phase nitrogen.
Tissues can also be banked prior to the isolation of the cells. Preferably such tissues are banked within a few hours or less after the completion of the pregnancy.
The cells are capable of growth in atmospheres containing oxygen from about 5%
to at least about 20% and comprise at least one of the following characteristics:
the cells have the potential for at least about 40 doublings in culture; the cells preferably are adherent, thus attachment and expansion on a coated or uncoated tissue culture vessel is preferred, wherein a coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, polylysine, vitronectin, or fibronectin. While the cells are preferably adherent and isolated as such, the cells have been grown in a spherical form in some embodiments.
Many populations of cells are present in postpartum tissue, but the cells of the invention preferably produce of at least one of tissue factor, vimentin, and alpha-smooth muscle actin;
more preferred are cells which produce each of tissue factor, vimeatin, and alpha-smooth muscle actin; production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and
- 17-HLA-A,B,C is also preferred. The cells are also characterized in their lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, 87-112, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; more preferable cells lack production of all of these surface markers. Also preferred are cells which express at least one of interleulcin 8;
reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha);
chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2);
cheinokine (C-X-C
motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3. The cells, in other embodiments, preferably also express one or more of C-type (calcium dependent, carbohydrate-recognition domain) lectin, superfamily member 2 (activation-induced); Wilms tumor 1;
aldehyde dehydrogenase 1 family, member A2; and renin; oxidized low density lipoprotein (lectin-like) receptor 1; Homo sapiens, clone IMAGE:4179671, mRNA, partial cds; protein kinase C, zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; Homo sapiens mRNA; and cDNA DKFZp547K1113 (from clone DKFZp547K1113 ). Preferred cells also have expression, which relative to a human cell that is a fibroblast, a mesenchrnal stem cell, or an iliac crest bone marrow cell, is reduced for at least one of:
short stature homeobox 2;
heat shock 27kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1);
elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens inRNA; cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein;
similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha;
procollagen C-endopeptidase enhancer, frizzled homolog 7 (Drosophila);
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280 fis, clone MANLMA1001744; cytoldne receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7;
DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3;
distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens tnRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate
- 18 -cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus ElB 19kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
and insulin-like growth factor binding protein 2, 36kDa. The skilled artisan will appreciate that the expression of a wide variety of genes is conveniently characterized on oligonucleotide arrays, for example on a Affymetrix GENECHIP.
The cells secrete a variety of biochemically active factors, such as growth factors, chemoldnes, cytokines and the like. Preferred cells secrete at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIPla, RANTES, and TIMPl; preferred cells may alternatively be characterized in their lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP lb, 1309, MDC, and VEGF, as detected by ELISA. These and other characteristics are available to identify and characterize the cells, and distinguish the cells of the invention from others known in the art.
In preferred embodiments, the cell comprises two or more of the foregoing characteristics. More preferred are those cells comprising, three, four, or five or more of the characteristics. Still more preferred are those isolated postpartum cells comprising six, seven, or eight or more of the characteristics. Still more preferred presently are those cells comprising all nine of the claimed characteristics.
Also presently preferred are cells that produce at least two of tissue factor, vimentin, and alpha-smooth muscle actin. More preferred are those cells producing all three of the proteins tissue factor, vimentin, and alpha-smooth muscle actin.
The skilled artisan will appreciate that cell markers are subject to vary somewhat under vastly different growth conditions, and that generally herein described are characterizations in Growth Medium, or variations thereof. Postpartum-derived cells that produce of at least one, two, three, or four of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C
are preferred. More preferred are those cells producing five, six, or seven of these cell surface markers. Still more preferred are postpartum cells that can produce all eight of the foregoing cell surface marker proteins.
Similarly, postpartum cells that lack of production of at least one, two, three, four of the proteins CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry are presently preferred. Cells lacking production of at least five, six, seven or eight or more of these markers are also preferred. More preferred are
-19-cells which lack production of at least nine or ten of the cell surface markers. Most highly preferred are those cells lacking production of all eleven of the foregoing identifying proteins.
Presently preferred cells produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C, and do not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
Presently, it is preferred that postpartum-derived cells express at least one, two or three of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2);
chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3. More preferred are those cells which express four or five, and still more preferred are cell capable of expressing all six of the foregoing genes.
In some embodiments, the cells preferably also express two, three, four or more of C-type (calcium dependent, carbohydrate-recognition domain) lectin, superfamily member 2 (activation-induced); Wilms tumor 1; aldehyde dehydrogenase 1 family, member A2; renin;
oxidized low density lipoprotein (lectin-like) receptor 1; Homo sapiens, clone IMAGE:4179671, mRNA, partial cds; protein kinase C, zeta; hypothetical protein DKFZp564F013;
dovvnregulated in ovarian cancer 1; Homo sapiens mRNA; and cDNA DKFZp547K1113 (from clone DKFZp547K1113). In other embodiments, it is preferred that the cells express five, six, seven or eight of the foregoing. Also preferred are those cells expressing genes corresponding to nine, ten or even all of the foregoing sequences.
For some embodiments, preferred are cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, have reduced expression for at least one of the genes corresponding to: short stature homeobox 2; heat shock 271cDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1);
elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA;
cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B;
= dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleulcin 11 receptor, alpha;
procollagen C-endopeptidase enhancer, frizzled homolog 7 (Drosophila);
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJ12280
- 20-fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7;
DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3;
distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type FE); biglycan; fibronectin 1;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus E1B 191cDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit Vila polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
insulin-like growth factor binding protein 2, 36kDa. More preferred are cells that have, relative _ to human fibroblasts, mesenchymal stem cells, or iliac crest bone marrow cells, reduced expression of at least 5, 10, 15 or 20 genes corresponding to those listed above. Presently more preferred are cell with reduced expression of at least 25, 30, or 35 of the genes corresponding to the listed sequences. Also more preferred are those postpartum-derived cells having expression that is reduced, relative to that of a human fibroblast, a mesenchymal stem cell, or an iliac crest bone marrow cell, of genes corresponding to 35 or more, 40 or more, or even all of the sequences listed.
Secretion of certain growth factors and other cellular proteins can make cells of the invention particularly useful. Preferred postpartum-derived cells secrete at least one, two, three or four of MCP-1, IL-6, 1L-8, GCP-2, HGF, KGF, FGF, 11B-EGF, BDNF, TPO, MIPla, RANTES, and TIMPl. Cells which secrete more than five, six, seven or eight of the listed proteins are also useful and preferred. Cells which can secrete at least nine, ten, eleven or more of the factors are more preferred, as are cells which can secrete twelve or more, or even all thirteen of the proteins in the foregoing list.
While secretion of such factors is useful, cells can also be characterized by their lack of secretion of factors into the medium. Postpartum-derived cells that lack secretion of at least one, two, three or four of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, 1VIDC, and VEGF, as detected by ELISA, are presently preferred for use. Cells that are characterized in their lack secretion of five or six of the foregoing proteins are more preferred. Cells which lack secretion of all seven of the factors listed above are also preferred.
-21-The postpartum-derived cells are preferably isolated in the presence of one or more enzyme activities. A broad range of digestive enzymes for use in cell isolation from tissue are known in the art, including enzymes ranging from those considered weakly digestive (e.g.
TM
deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin). For example, collagenases are known to be useful for isolating various cells from tissues. Deoxyribonucleases can digest single-stranded DNA and can minimize cell-clumping during isolation. Enzymes can be used alone or in combination. Serine protease are preferably used in a sequence following the use of other enzymes as they may degrade the other enzymes being used. The temperature and time of contact with serine proteases must be monitored.
Serine proteases may be inhibited with alpha 2 microglobulin in serum and therefore the medium used for digestion is preferably serum-free. EDTA and DNase are commonly used and may improve yields or efficiencies. Preferred methods involve enzymatic treatment with for example TM TM
collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral TM
protease dispase are used in the dissociating step. Presently preferred are mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as-trypsin, chymotrypsin, or elastase), and deoxyribonucleases. More preferred are enzyme activites selected from metalloproteases, neutral proteases and mucolytic activities. Presently preferred are cells that are TM
isolated in the presence of one or more activities of collagenase, hyaluronidase and dispase.
More preferred are those cells isolated in the presence of a collagenase from Clostridium TM
histolyticum, and either of the protease activities, dispase and thermolysin.
Still more preferred TM
are cells isolated with collagenase and dispase enzyme activities. Also preferred are such cells TM
isolated in the presence of a hyaluronidase activity, in addition to collagenase and dispase activity. The skilled artisan will appreciate that many such enzyme treatments are known in the art for isolating cells from various tissue sources.Also useful for isolation of certain cells of the invention are commercial enzyme preparations such as blends of enzymes, for example, LEBERASE Blendzymes available from Roche Diagnostics. The skilled artisan will appreciate the methods for optimizing enzyme use during isolation, and information on such optimization procedures is available from the manufacturers of commercial enzymes.
Preferred are those methods which can result in homogenous populations or nearly homogeneous populations of cells.
The cells also preferably comprise a normal karyotype, which is maintained as the cells are passaged. Karyotyping is particularly useful for identifying and distinguishing neonatal from
- 22 -maternal cells derived from placenta. Methods for karyotyping are available and known to those of skill in the art.
Among cells that are presently preferred for use with the invention in several of its aspects are postpartum cells having the characteristics described above and more particularly those wherein the cells have normal karyotypes and maintain normal karyotypes with passaging, and during cryopreservation and subsequent thawing and use, and farther wherein the cells express each of the markers CD10, CD13, CD44, CD73, CD90, PDG-Fr-alpha, and HLA-A,B,C, wherein the cells produce the immunologically-detectable proteins which correspond to the listed markers. Still more preferred are those cells which in addition to the foregoing do not produce proteins corresponding to any of the markers CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
Certain prior art cells having the potential to differentiate along lines leading to various phenotypes are unstable and thus can spontaneously differentiate. Presently preferred for use with the invention are cells which do not spontaneously differentiate, for example along cardiomyogenic, angiogenic, hemangiogenic, or vascualogenic lines_ Preferred cells when grown in Growth Medium are substantially stable with respect to the cell markers produced on their surface, and with respect to the expression pattern of various genes, for example as deteHnined using oligonucleotide arrays, such as an Affymetrix GENECHIP.. The cells remain substantially constant in their biochemical, genetic, and immunological characteristics, for example, their cell surface markers, over passaging, and through multiple population doublings.
In another of its several aspects, the invention provides populations of cells comprising the cells described above. Cell populations are useful in connection -with the methods of the invention, as well as in connection with making the therapeutic cell compositions and cell lysates in larger amounts than isolated cells can provide.
Preferred populations comprise from about 1% postpartum-derived cells to about 10%
postpartum cells. More preferred populations comprise at least about 10%
postpartum-derived cells. More preferred populations comprise at least about 25% postpartum-derived cells. Also, some preferred populations comprise about 50% postpartum-derived cells. Such populations may be useful for coculture or other cultures wherein the cells are equally populous and divide at the same rate, or where the population is adjusted to about 50% of each culture after expansion of the cultures in coculture or separately. More preferred for some applications are populations comprising at least about 65% postpartum-derived cells. Populations that comprising at least 90% postpartum-derived cells are highly preferred for certain aspects of the invention. More preferred populations comprise substantially only postpartum-derived cells.
-23 -The populations may comprise a clonal cell line of postpartum-derived cells.
Such populations are particularly useful wherein a cell clone with highly desirable functionality is isolated. Both neotal and maternal clones are useful and are provided herein_ Methods of isolating clonal cell lines from cultured cells are known in the art.
The invention also provides cell lysates, soluble cell fractions and membrane-enriched cell fractions prepared from the populations of the postpartum cells. Such lysates and fractions have many utilities. Use of cell lysates, and more particularly soluble cell fractions, in vivo allows the beneficial intracellular milieu to be used in a patient allogeneic patient without stimulating allogeneic lymphocytes, or generating other adverse immunological responses, or triggering rejection. Methods of lysing cells are well-known in the art and include various means of mechanical disruption, enzymatic disruption, or chemical disruption, or combinations thereof.
Such cell lysates may be prepared from cells directly in their Growth Medium and thus containing secreted growth factors and the like, or may be prepared from cells washed free of medium in, for example, PBS or another solution. For making lysates from cells directly in the growth medium it is preferred that cells are grown in serum from the species in which the lysates are to be used, in some embodiments, washed cells may be preferred. Washed cells may be resuspended at concentrations greater than the original population density if preferred. Cell lysates prepared from populations of postpartum-derived cells may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, enriched, partially purified, combined with pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example phan-naceutically useful protein compositions. Cell lysates may be used in vitro or in vivo, alone or, for example, with syngeneic or autologous live cells. The lysates, if introduced in vivo, may be introduced locally at a site of treatment, or remotely to provide, for example, needed cellular growth factors to a patient. Preferably, the lysates are not immunogenic, and more preferably they are immunologically tolerated in a broad population of syngeneic and allogeneic recipients without adverse immunological consequences or reaction. Cell lysates of the invention are useful from cells at any stage or age which have been grown under conditions for growth and expansion, for example on Growth Medium. Even senescent cells are useful for the preparation of lysate and can provide certain factors which are biologically useful. Nonviable or even dead or killed cells have utility for preparing lysates, and cellular fractions. Also useful are lysates from cells which have been exposed to factors which tend to induce them along a mesenchymal pathway, particularly towards cardiomyogenic, angiogenic, hemangiogenic, and vascalogenic lines. Cell lysates from differentiated cells, or cells more committed than the PPDCs are also desirable. For
-24 -example, lysates from cells with characteristics of cardiomyoblasts, caroliomyocytes, angioblasts, hemangioblasts and the like, or their progenitors are also useful and contemplated for use herewith.
Also provided herein are populations of cells incubated in the presence of one or more factors which stimulate stem cell differentiation along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway. Such factors are known in the art and the skilled artisan will appreciate that determination of suitable conditions for differentiation can be accomplished with routine experimentation. Optimization of such conditions can be accomplished by statistical experimental design and analysis, for example, response surface methodology allows simultaneous optimization of multiple variables, for example biological culture conditions.
Presently preferred factors include, but are not limited to factors, such as growth factors, chemokines, cytokines, cellular products, demethylating agents, and other stimuli which are now known or later determined to stimulate differentiation, for example of stem cells, along cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathways or lineages.
Presently preferred for inducing or stimulating differentiation along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway are cells incubated in the presence of factors comprising at least one of a demethylation agent, a member of BMP, FG-F, TAK, GATA, Csx, NK, MEF2, ET-1, and Wnt factor families, Hedgehog, Csx/Nkx-2.5, and anti-Wnt factors. DNA
methylation is known to silence certain genes, preventing their expression, demethylation may allow expression of such genes, including some required for differentiation.
Preferred demethylation agents include inhibitors of DNA methyltransferases or inhibitors of histone deacetylase, or inhibitors of a repressor complex.
Presently preferred demethylation agents comprise at least one o f 5-azacytidine, 5-aza-2'-deoxycytidine, DMSO, chelerythrine chloride, retinoic acid or salts thereof, 2-amino-4-(ethylthio)butyric acid, procainamide, and procaine. Inclusion of such factors tend to induce the cells to differentiate along mesenchymal lines, toward a cardiomyogenic pathway, as determined, for example, by the expression of at least one of cardiomyosin, skeletal myosin, or GATA4; or by the acquisition of a beating rhythm, spontaneous or otherwise induced; or by the ability to integrate at least partially into a patient's cardiac muscle without inducing arrhythmias.
In preferred embodiments herein, cells induced with one or more factors as identified above may become cardiomyogenic, angiogenic, hemangiogenic, or vasculogenic cells, or progenitors or primitive relatives thereof. Preferably at least some of the cells can integrate at least partially into the patient's heart or vasculature tree, including but not limited to heart muscle, vascular and other structures of the heart, blood vessels, and the like. More preferred are
-25 -differentiated cells acquiring two or more of the indicia of cardionlyogenic, cells or their progenitors, and able to fully integrate into a patient's heart or vasculature. Still more preferred are those cells which when placed into a patient, result in no increase in arrhythmias, heart defects, blood vessel defects or other anamolies related to the patient's circulatory system or health. Also preferred are those cells which can stimulate stem cells naturally present in the patient's cardiac muscle, blood vessels, blood and the like to themselves differentiate into for example, cardiomyocytes, or at least along cardiomyogenic, angiogenic, hemangiogenic, or vasculogenic lines. Equally preferred are PPDCs which can support the stem cells naturally present in the patient's cardiac muscle, blood, blood stream and the like to grow and expand and be available for later differentiation.
The populations can be provided therapeutically to a patient, for example with a disease of the heart or circulatory system. Common examples, not intended to limit the invention, include congestive heart failure due to atherosclerosis, cardiomyopathy, or cardiac injury, such as from myocardial infarction or wound (acute or chronic). In presently preferred embodiments, the population comprises about 50% postpartum-derived cells, while in other preferred embodiments the population is a substantially homogeneous population of postpartum-derived cells. In other embodiments the population comprises at least about 1, 10, 20, 25, 30, 33, 40, 60, 66, 70, 75, 80, or 90% postpartum-derived cells.
Co-cultures comprising the cells or cultures of the invention with other mammalian cells are also provided herein. Preferably these co-cultures comprise another mammalian cell line whose growth or therapeutic potential, for example, is improved by the presence of the umbilicus-derived cells. Human cell lines are particular preferred. Such co-cultures are useful for therapeutic application in vitro or in vivo.
Also provided herein are therapeutic compositions comprising a postpartum-derived cell and another therapeutic agent, factor, or bioactive agent, such as a pharmaceutical compound.
Such bioactive agents include, but are not limited to, IGF, LIF, PDGF, EGF, FGF, as well as antithrombogenic, antiapoptotic agents, anti-inflammatory agents, immunosuppressive or immunomodulatory agents, and antioxidants. Such therapeutic compositions can further comprise one or more additional cell types in addition to the PPDCs and the bioactive component.
Thus, in conjunction with therapeutic cells, other biologically active molecules, such as antithrombogenic agents, anti-apoptotic agents, and anti-inflammatory agents may be useful and may be administered in sequence with, or coadministered with the the cells, individually or in combinations or two or more such compounds or agents. For example, anti-apoptotic agents
- 26 -may be useful to minimize programmed cell death. Such agents include but are not limited to EPO, EPO derivatives and analogs, and their salts, TPO, IGF-I, IGF-II, hepatocyte growth factor (HGF), and caspase inhibitors. Anti-inflammatory agents include but are not limited to P38 MAP kinase inhibitors, statins, IL-6 and IL-1 inhibitors, Pemirolast, Tranilast, Remicade, Sirolimus, nonsteroidal anti-inflammatory compounds, for example, Tepoxalin, Tolmetin, and Suprofen.
Other bioactive factors or therapeutic agents which can be coadministered with the therapeutic cells of the invention include, for example, antithrombogenic factorsõ
immunosuppressive or immunomodulatory agents, and antioxidants. Examples of immunosuppressive and immudulatory agents include calcineurin inhibitors, for example cyclosporine, Tacrolimus, mTOR inhibitors such as Sirolimus or Everolimus;
anti-proliferatives such as azathioprine and mycophenolate mofetil; corticosteroids for example prednisolone or hydrocortisone; antibodies such as monoclonal anti-IL-2Ra receptor antibodies, Basiliximab, Daclizumab; polyclonal anti-T-cell antibodies such as anti-thymocyte globulin (ATG), anti-lymphocyte globulin (ALG), and the monoclonal anti-T cell antibody OKT3.
Antithrombogenic compounds which can be therapeutically provided in conjunction with the cells of the invention include, for example, heparin, heparin derivatives, urokinase, and PP ack (dextrophenylalanine proline arginine chloromethylketone); antithrombin compounds, platelet receptor antagonists, anti-thrombin antibodies, anti-platelet receptor antibodies, aspirin, dipyridamole, protamine, hirudin, prostaglandin inhibitors, and platelet inhibitors. Antioxidants are well known in the art of course and any pharamacueitcally acceptable antioxidant may be administered in conjunction with the cells of the invention including probucol; vitamins A, C, and E, coenzyme Q-10, glutathione, L cysteine, N-acetylcysteine, or antioxidant derivative, analogs or salts of the foregoing.
In addition to the above, compositions derived from the cells are provided herein. Cell lysates, soluble cell fractions and membrane-enriched cell fractions are provided herein, as described above in detail. Extracellular matrices derived from the cells, for example, comprising basement membranes are also useful and are provided herein. Cell lysates, soluble cell fractions, membrane-enriched cell fractions and extracellular matrix derived from the cells can all be administered to patients as appropriate, or coadministered with the cells of the invention, with or without additional cells or cell types.
Compositons of the invention also include conditioned culture media as provided herein.
Such media have first been used to grow the cells or cultures of the invention, which during growth secrete one or more useful products into the medium. Conditioned medium from these
- 27 -novel cells are useful for many purposes, including for example, supporting the growth of other mammalian cells in need of growth factors or trophic factors secreted into the media by the cells and cultures of the invention, and promoting, for example, angiogenesis.
Methods of preparing and storing conditioned media are known in the art and primarily involve removal of the cells, for example by centrifugation.
The invention provides in another of its aspects therapeutic cell compositions comprising a pharmaceutically-acceptable carrier and postpartum-derived cells derived from mammalian postpartum tissue substantially free of blood. The cells are capable of self-renewal and expansion in culture and have the potential to differentiate along mesenchymal lineage, towards cardiomyogenic, angiogenic and vasculogenic phenotypes, and further towards cells such as cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g. vascular smooth muscle cells), as well as cells of the excitatory and conductive systems, and progenitors of the foregoing. The cells are capable of growth in an atmosphere containing oxygen from about 5% to at least about 20%. The cells also require L-valine for growth; have the potential for at least about 40 doublings in culture; attach and expand on a coated or uncoated tissue culture vessel, wherein a coated tissue culture vessel is coated with gelatin, laminin, or fibronectin; produce tissue factor, vimentin, and alpha-smooth muscle actin; produce each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C; and do not produce any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry. In preferred embodiments the cells are derived from human tissue.
The therapeutic cell compositions provided can be provided therapeutically in a patient with a disease of the heart or circulatory system, such as a cardiomyopathy, or other heart disease, or a cardiac injury. In certain embodiments, the therapeutic cell compositions comprise cells induced to differentiate along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway or lineage. The therapeutic cell compositions can comprise cells or cell products that stimulate adult stem cells present in the heart, blood, blood vessels and the like, to divide, or differentiate, or both.
The therapeutic cell compositions are provided, for example, by injection. In certain embodiments, the therapeutic cell compositions are provided by intracardiac injection. In other embodiments, the injection may be onto the surface of the heart, into an adjacent area, or even to a more remote area. In preferred embodiments, the cells can home to the diseased or injured area. Particularly preferred are cells that can be injected intravenously and locate appropriately to the desired site of action, for example, cardiomyocytes or their progenitors preferably have the ability to locate and home to the heart muscle or it structures.
- 28 -The therapeutic cell compositions can also be provided in the form of a matrix-cell complex. Matrices include biocompatible scaffolds, lattices, self-assembling structures and the like, whether bioabsorbable or not, liquid, gel, or solid. Such matrices are known in the arts of therapeutic cell treatment, surgical repair, tissue engineering, and wound healing. Preferably the matrices are pretreated with the therapeutic cells. More preferably the matrices are populated with cells in close association to the matrix or its spaces. The cells can adhere to the matrix in some embodiments, in others the cells are entrapped or contained within the matrix spaces. Most preferred are those matrix-cell complexes were the cells are growing in close association with the matrix and when used therapeutically, in growth of the patient's cells is stimulated and supported, and proper angiogenesis is similarly stimulated or supported. The matrix-cell compositions can be introduced into a patients body in any way known in the art, including but not limited to implantation, injection, surgical attachment, transplantation with other tissue, injection, and the like. In some embodiemtns, the matrices form in vivo, or even more preferably in situ, for example in situ polymerizable gels can be used in accordance with the invention.
Examples of such gels are known in the art.
In some embodiments, the cells of the invention, or co-cultures thereof, may be seeded onto such three-dimensional matrices, such as scaffolds and implanted in vivo, where the seeded cells may proliferate on or in the framework or help establish replacement tissue in vivo with or without cooperation of other cells.
Growth of PPDCs or co-cultures thereof on the three-dimensional framework preferably results in the formation of a three-dimensional tissue, or foundation therefor, which can be utilized in vivo, for example for repair of damaged or diseased tissue. For example, the three-dimensional scaffolds can be used to form tubular structures, for example for use in repair of blood vessels; or aspects of the circulatory system or coronary structures.
In accordance with one aspect of the invention, PPDCs or co-cultures thereof are inoculated, or seeded on a three-dimensional framework or matrix, such as a scaffold, a foam or hydrogel. The framework may be configured into various shapes such as generally flat, generally cylindrical or tubular, or can be completely free-form as may be required or desired for the corrective structure under consideration. In some embodiments, the PPDCs grow on the three dimensional structure, while in other embodiments, the cells only survive, or even die, however in doing so they stimulate or promote ingrowth of new tissue, for example, and preferably vascularization. PPDCs may be co-administered with myocytes, myoblasts, vascular endothelial cells, dermal fibroblasts, keratinocytes, and other soft tissue type progenitors, including stem cells. When grown in this three-dimensional system, the proliferating cells
-29 -mature and segregate properly to form components of adult tissues analogous to counterparts found naturally in vivo.
For example, but not by way of limitation, the matrix may be designed such that the matrix structure: (1) supports the PPDCs or co-cultures thereof without subsequent degradation;
(2) supports the PPDCs or co-cultures thereof from the time of seeding until the tissue transplant is remodeled by the host tissue; (2) allows the seeded cells to attach, proliferate, and develop into a tissue structure having sufficient mechanical integrity to support itself in vitro, at which point, the matrix is degraded. A review of matrix design is provided by Hutmacher, J.
Biomat. Sci.
Polymer Edn., 12(1):107-124 (2001).
The matrices, scaffolds, foams and self-assembling systems contemplated for use herein can be implanted in combination with any one or more cells, growth factors, drugs, or other components, such as bioactive agents that promote healing, or in growth of tissue, or stimulate vascularization or innervation thereof or otherwise enhance or improve the therapeutic outcome or the practice of the invention, in addition to the cells of the invention.
The cells of the invention can be grown freely in culture, removed from the culture and inoculated onto a three-dimensional framework. Inoculation of the three-dimensional framework with a concentration of cells, e.g., approximately 106 to 5 x 107 cells per milliliter, preferably results in the establishment of the three-dimensional support in relatively shorter periods of time.
Moreover in some application it may be preferably to use a greater or lesser number of cells depending on the result desired.
In some embodiments, it is useful to re-create in culture the cellular microenvironment found in vivo, such that the extent to which the cells are grown prior to implantation in vivo or use in vitro may vary. PPDCs or co-cultures thereof may be inoculated onto the framework before or after forming the shape desired for implantation, e.g., ropes, tubes, filaments, and the like. Following inoculation of the cells onto the framework, the framework is preferably incubated in an appropriate growth medium. During the incubation period, the inoculated cells will grow and envelop the framework and may for example bridge, or partially bridge any interstitial spaces therein. It is preferable, but not required to grow the cells to an appropriate degree which reflects the in vivo cell density of the tissue being repaired or regenerated. In other embodiments, the presence of the PPDCs, even in relatively low numbers on the framework encourages ingrowth of the other healthy cells to facilitate healing for example of a wounded or necrotic tissue..
Examples of matrices, for example scaffolds which may be used for aspects of the invention include mats (woven, knitted, and more preferably nonwoven) porous or semiporous
-30-foams, self assembling peptides and the like. Nonwoven mats may, for example, be formed using fibers comprised of natural or synthetic polymers. In a preferred embodiment, absorbable copolymers of glycolic and lactic acids (PGA/PLA), sold under the tradename VICRYL
(Ethicon, Inc., Somerville, NJ) are used to form a mat. Foams, composed of, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, fonned by processes such as freeze-drying, or lyophilization, as discussed in U.S. Patent No.
6,355,699, can also serve as scaffolds. Gels also form suitable matrices, as used herein. Examples include in situ polymerizable gels, and hydrogels, for example composed of self-assembling peptides. These materials are frequently used as supports for growth of tissue. In situ-forming degradable networks are also suitable for use in the invention (see, e.g., Anseth, K.S.
et al., 2002, J.
Controlled Release 78: 199-209; Wang, D. et al., 2003, Biomaterials 24: 3969-3980; U.S. Patent Publication 2002/0022676 to He et al.). These materials are formulated_ as fluids suitable for injection, then may be induced by a variety of means (e.g., change in temperature, pH, exposure to light) to form degradable hydrogel networks in situ or in vivo.
According to a preferred embodiment, the framework is a felt, which can be composed of a multifilament yarn made from a bioabsorbable material, e.g., PGA, PL,A, PCL
copolymers or blends, or hyaluronic acid. The yarn is made into a felt using standard textile processing techniques consisting of crimping, cutting, carding and needling. In another preferred embodiment the cells of the invention are seeded onto foam scaffolds that may be composite structures. In addition, the three-dimensional framework may be molded into a useful shape, such as a specific structure in the body to be repaired, replaced, or augmented.
The framework may be treated prior to inoculation of the cells of the invention in order to enhance cell attachment. For example, prior to inoculation with the cells of the invention, nylon matrices could be treated with 0.1 molar acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon. Polystyrene could be similarly treated using sulfuric acid.
In addition, the external surfaces of the three-dimensional framework may be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma coating the framework 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), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, among others.
In some embodiments, the scaffold is comprised of 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
-31 -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 poly-urethaneurea silicones, such as PURSPAN (The Polymer Technology Group, Inc., Berkeley, CA). These materials can be further treated to render the scaffold non-thrombogenic. Such treatments include anti-thrombotic agents such as heparin, and treatments which alter the surface charge of the material such as plasma coating.
Different proportions of the various types of collagen, for example, deposited on the framework can affect the growth of tissue-specific or other cells which may be later inoculated onto the framework or which may grow onto the structure in vivo. For example, for three-dimensional skin culture systems, collagen types I and III are preferably deposited in the initial matrix. Alternatively, the framework can be inoculated with a mixture of cells which synthesize the appropriate collagen types desired. Thus, depending upon the tissue to be cultured, the appropriate collagen type to be inoculated on the framework or produced by the cells seeded thereon may be selected. For example, the relative amounts of collagenic and elastic fibers present in the framework can be modulated by controlling the ratio of collagen-producing cells to elastin-producing cells in the initial inoculum. For example, since the inner walls of arteries are rich in elastin, an arterial scaffold should contain a co-culture of smooth muscle cells which secrete elastin.
The seeded or inoculated three-dimensional framework of the invention can be used in a variety of applications. These include but are not limited to transplantation or implantation of either the cultured cells obtained from the matrix or the cultured matrix itself in vivo. The three-dimensional scaffolds may, according to the invention, be used to replace or augment existing tissue, to introduce new or altered tissue, to modify artificial prostheses, or to join together biological tissues or structures. For example, and not by way of limitation, specific embodiments of the invention include but are not limited to, flat structures and tubular three-dimensional tissue implants for repair or regeneration, for example, of cardiac muscle, its structures, and those of the entire vascular tree, including for example the endovascular structures of the brain and intracranium.
PPDCs can be inoculated onto a flat scaffold. The scaffold is preferably incubated in culture medium prior to implantation. Two or more flat frameworks can be laid atop another and sutured together to generate a multilayer framework.
For example and not by way of limitation, the three-dimensional framework can also be used to construct single and multi-layer tubular tissues in vitro that can serve as a replacement for damaged or diseased tubular tissue in vivo.
-32-A scaffold can be cut into a strip (e.g., rectangular in shape) of which the width is approximately equal to the inner circumference of the tubular organ into which it will ultimately be inserted. The cells can be inoculated onto the scaffold and incubated by floating or suspending in liquid media. At the appropriate stage of confluence, the scaffold can be rolled up into a tube by joining the long edges together. The seam can be closed by suturing the two edges together using fibers of a suitable material of an appropriate diameter.
According to the invention, a scaffold can be formed as a tube, inoculated with PPDCs, and suspended in media in an incubation chamber. In order to prevent cells from occluding the lumen, one of the open ends of the tubular framework can be affixed to a nozzle. Liquid media can be forced through this nozzle from a source chamber connected to the incubation chamber to create a current through the interior of the tubular framework. The other open end can be affixed to an outflow aperture which leads into a collection chamber from which the media can be recirculated through the source chamber. The tube can be detached from the nozzle and outflow aperture when incubation is complete. This method is described by Ballermann, B. J., et al., Int.
Application No. WO 94/25584 and in U.S. Application Ser. No. 08/430,768.
In general, two three-dimensional frameworks can be combined into a tube in accordance with the invention using any of the following methods.
Two or more flat frameworks can be laid atop another and sutured together.
This two-layer sheet can then be rolled up, and, as described above, joined together and secured.
One tubular scaffold that is to serve as the inner layer can be inoculated with PPDCs and incubated. A second scaffold can be grown as a flat strip with width slightly larger than the outer circumference of the tubular framework. After appropriate growth is attained, the flat framework can be wrapped around the outside of the tubular scaffold followed by closure of the seam of the two edges of the flat framework and, preferably, securing the flat framework to the inner tube.
Two or more tubular meshes of slightly differing diameters can be grown separately. The framework with the smaller diameter can be inserted inside the larger one and secured.
For each of these methods, more layers can be added by reapplying the method to the double-layered tube. The scaffolds can be combined at any stage of growth of the PPDCs, and incubation of the combined scaffolds can be continued when desirable.
The lumenal aspect of the tubular construct can be comprised of or treated with materials that render the lumenal surface of the tubular scaffold 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
- 33 materials such as basement membrane proteins such as laminin and Type IV
collagen, synthetic materials such as ePTFE, and segmented polyurethaneurea silicones, such as PLJRSPAN (The Polymer Technology Group, Inc., Berkeley, CA). These materials can be further treated to render the lumenal surface of the tubular scaffold non-thrombogenic. Such treatments include anti-thrombotic agents such as heparin, and treatments which alter the surface charge of the material such as plasma coating.
In conjuction with the above, the cells, cell lysates and fractions, and therapeutic compositions of the invention can be used in conjunction with implantable devices. For example the cells, cell lysates and cell fractions can be coadminstered with, for example stents, artificial valves, ventricular assist devices, Guglielmi detachable coils and the like.
As the devices may constitute the dominant therapy provided to an individual in need of such therapy, the cells and the like may be used as supportive or secondary therapy to assist in, stimulate, or promote proper healing in the area of the implanted device. The cells, lysates, cell fractions and therapeutic compositions of the invention may also be used to "pretreat" certain implantable devices, to minimize problems when they are used in vivo. Such pretreated devices, including coated devices may be better tolerated by patients receiving them, with decrease risk of local or systemic infection, or for example, restenosis or further occulision of blood vessels.
The therapeutic cell compositions, in certain embodiments also comprise cells that express at least one of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; , and tumor necrosis factor, alpha-induced protein 3; or which have reduced expression, relative to a human cell that is a fibroblast, a rnesenchymal stem cell, or an iliac crest bone marrow cell, for at least one of: short stature homeobox 2; heat shock 27kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1);
elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Ifonzo sapiens mRNA; cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme horneobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein;
Similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleukin 11 receptor, alpha;
procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosoplzila);
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); ixoquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Honza sapiens cDNA FLJ12280
-34--fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7;
DKFZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; early growth response 3;
distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus ElB 19kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit Vila polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
insulin-like growth factor binding protein 2, 36kDa.
Preferred therapeutic cell compositions also comprise cells which secrete at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIPla, RANTES, and TIMPl; and do not secrete at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
In yet another of its aspects, the invention provides methods for treating a patient with a heart disease or injury comprising administering a therapeutic postpartum-derived cell composition to a patient with a disease or injury of the heart or circulatory system; and evaluating the patient for improvements in cardiac function. In certain preferred embodiments the heart disease is a cardiomyopathy, either idiopathic or with a known cause, and either ischemic or nonischemic in nature. While patients with any heart or circulatory disease will benefit from such therapy, patients with myocardial infarction caused by any condition may benefit by receiving the therapeutic cell compositions of the invention as discussed below. In other preferred embodiments, the disease of the heart or circulatory system comprises one or more of angioplasty, aneurysm, angina (angina pectoris), aortic stenosis, aortitis, arrhythmias, arteriosclerosis, arteritis, asymmetric septal hypertrophy (ASH), atherosclerosis, Athletic Heart Syndrome, atrial fibrillation and flutter, bacterial endocarditis, Barlow's Syndrome (mitral valve prolapse), bradycardia, Buerger's Disease (thromboangiitis obliterans), cardiomegaly, cardiomyopathy, carditis, carotid artery disease, coarctation of the aorta, congenital heart
- 35 -diseases (congenital heart defects), congestive heart failure (heart failure), coronary artery disease, Eisenmenger's Syndrome, embolism, endocarditis, erythromelalgia, fibrillation, fibromuscular dysplasia, heart block, heart murmur, hypertension, hypotension, idiopathic infantile arterial calcification, Kawasaki Disease (mucocutaneous lymph node syndrome, mucocutaneous lymph node disease, infantile polyarteritis), metabolic syndrome, microvascular angina, myocardial infarction (heart attacks), myocarditis, paroxysmal atrial tachycardia (PAT), periarteritis nodosa (polyarteritis, polyarteritis nodosa), pericarditis, peripheral vascular disease, phlebitis, pulmonary valve stenosis (pulmonic stenosis), Raynaud's Disease, renal artery stenosis, renovascular hypertension, rheumatic heart disease, septal defects, silent ischemia, syndrome X, tachycardia, Takayasu's Arteritis, Tetralogy of Fallot, transposition of the great vessels, tricuspid atresia, truncus arteriosus, valvular heart disease, varicose ulcers, varicose veins, vasculitis, ventricular septal defect, Wolff-Parkinson-White Syndrome, and endocardial cushion defect.
In still other preferred embodiments, the disease of the heart or circulatory system comprises one or more of acute rheumatic fever, acute rheumatic pericarditis, acute rheumatic endocarditis, acute rheumatic myocarditis, chronic rheumatic heart diseases, diseases of the mitral valve, mitral stenosis, rheumatic mitral insufficiency, diseases of aortic valve, diseases of other endocardial structures, ischemic heart disease (acute and subacute), angina pectoris, diseases of pulmonary circulation (acute pulmonary heart disease, pulmonary embolism, chronic pulmonary heart disease), kyphoscoliotic heart disease, pericarditis, myocarditis, endocarditis, endomyocardial fibrosis, endocardial fibroelastosis, atrioventricular block, cardiac dysrhythmias, myocardial degeneration, diseases of the circulatory system including cerebrovascular disease, occlusion and stenosis of precerebral arteries, occlusion of cerebral arteries, diseases of arteries, arterioles and capillaries (atherosclerosis, aneurysm) diseases of veins and lymphatics, and other diseases of circulatory system.
Measurement of improvement in patients receiving the therapeutic compositions provided herein can include any means known in the art, but preferred improvements include improvements in hemodynamic measurements including but not limited to chest cardiac output (CO), cardiac index (CI), pulmonary artery wedge pressures (PAWP), and cardiac index (CI), %
fractional shortening (%FS), ejection fraction (EF), left ventricular ejection fraction (LVEF); left ventricular end diastolic diameter (LVEDD), left ventricular end systOlic diameter (LVESD), contractility (e.g. dP/dt), pressure-volume loops, measurements of cardiac work, an increase in atrial or ventricular functioning; an increase in pumping efficiency, a decrease in the rate of loss of pumping efficiency, a decrease in loss of hemodynamic functioning; and a decrease in complications associated with cardiomyopathy. Biochemical measurements of improvement are
- 36 -also contemplated herein, for example production of certain cellular products or factors. The presence or absence of biological molecules, for example particular enzymes (or their activities), mRNAs, transcription factors, proteins, modified proteins, lipids, sterols, or the like may be shown to correlate with improvement in cardiac or circulatory health and the use of these measurements of improvement are also contemplated for use herein. Also contemplated herein as a indications of improvement are histological changes deemed beneficial or for example, indicia of angiogenesis or improved vascularization.
In some presently preferred embodiments, the methods comprise inducing the therapeutic postpartum-derived cells to differentiate along mesenchymal lineage, towards cardiomyogenic, angiogenic and vasculogenic phenotypes, or even further towards cells such as cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g. vascular smooth muscle cells), or towards cells of the excitatory and conductive systems, and progenitors or more primitive relatives of the foregoing.. Such cells are discussed above, and the methods and factors for differentiating the cells, assessing the induction of cells to differentiate, and the uses of such cells for the therapeutic compositions is analogous. Also the therapeutic cell compositions can integrate into the patient's heart, or alternatively can provide support for growth or stimulation to differentiate for naturally present cardiac stem cells present.
Therapeutic cells can be coadministered with cell lysates, or with other allogeneic, syngeneic or autologous cells. The survival of the cells delivered in administering the therapeutic cell compositions is not determinative of the success or results of their use, rather the improvement in cardiac or circulatory health is outcome determinative. Thus, the cells need not integrate and beat with the patient's heart, or into blood vessels, but rather the indicia of improvements in cardiac or circulatory health in the patient before and after treatment preferably include at least one of objective measurements of cardiac or circulatory health, and subjective assessment (including self-assessment) of the patient's condition. A successful treatment could thus comprise treatment of a patient with a cardiomyopathy with a therapeutic cell composition comprising the PPDCs, in the presence or absence of another cell type. For example, and not by way of limitation, the PPDCs preferably at least partially integrate, multiply, or survive in the patient. In other preferred embodiments, the patient experiences benefits from the therapy, for example from the ability of the PPDCs to support the growth of other cells, including stem cells or progenitor cells present in the heart, from the tissue ingrowth or vascularization of the tissue, and from the presence of beneficial cellular factors, chemokines, cytokines and the like, but the cells do not integrate or multiply in the patient. In another embodiment, the patient benefits from the therapeutic treatment with the PPDCs, but the cells do not survive for a prolonged period in
- 37 -the patient. In one embodiment, the cells gradually decline in number, viability or biochemical activity, in other embodiments, the decline in cells may be preceded by a period of activity, for example growth, division, or biochemical activity. In other embodiments, senescent, nonviable or even dead cells are able to have a beneficial therapeutic effect.
The administering is preferably in vivo by transplanting, implanting, injecting, fusing, delivering via catheter, or providing as a matrix-cell complex, or any other means known in the art for providing cell therapy.
Patients with myocardial infarction caused by any condition may benefit by receiving the therapeutic cell compositions of the invention. Such treatment is preferably provided within a reasonable therapeutic window after the cardiac event. Presently, it is preferred that treatment with the cells or compositions of the invention be initiated within 30 days of the myocardial infarction. Treatment within 1-21 days is preferred. It is also comtemplated herein that beneficial effects of certain applications, for example by intravenous injection where the cells home to the damaged site, will allow treatment far more rapidly. For example, it is presently contemplated that treatment in close temporal relation with the myocardial infarction or similar cardiac event may be beneficial. In preferred embodiments, treatment with the therapeutic cell compositions is within twenty four hours of the cardiac event. Also preferred is treatment within 12, 8, or even four hours. More preferably treatment is given with two hours.
Treatment within one hour of the event is more preferred, with treatment within 30 minutes, or even 15 minutes most preferred. Also provided herein are kits for use in the treatment of myocardial infarction.
The kits provide the therapeutic cell composition which can be prep ared in a pharmaceutically acceptable fonn, for example by mixing with a pharmaceutically acceptable carrier, and an applicator, along with instructions for use. Ideally the kit can be used in the field, for example in a physician's office, or by an emergency care provider to be applied to a patient diagnosed as having had a myocardial infarction or similar cardiac event.
The invention also provides in another aspect, methods for treating a patient with a disease of the heart or circulatory system comprising administering a therapeutic postpartum-derived cell composition to a patient with a disease of the heart or circulatory system; and evaluating the patient for improvements in cardiac function, wherein the administering is with a population of another cell type. Administration of cocultures, mixed populations or other nonclonal populations are preferred. Other cell types which can be coadministered are stem cells in certain embodiments, while in others, myoblasts, myocytes, cardiomyoblasts, cardiomyocytes, or progenitors of myoblasts, myocytes, cardiomyoblasts, or cardioniyocytes are used.
-38-Also provided herein are methods for treating a patient with a disease of the heart or circulatory system comprising administering a therapeutic postpartum-derived cell composition to a patient with a disease of the heart or circulatory system; and evaluating the patient for improvements in cardiac function, wherein the therapeutic cell composition is administered as a matrix-cell complex. In certain embodiments, the matrix is a scaffold, preferably bioabsorbable, comprising at least the postpartum-derived cells.
Kits for the therapeutic application of the populations and cocultures of the invention are also provided. Where used for treatment of cardiomyopathy, or other scheduled treatment, the kits include a therapeutic cell composition, with or without a matrix, and with or without a coculture present. The kits also optionally include a means of administering the cells, for example by injection, and a pharmaceutically-acceptable carrier for the cells, if required. The kits include instructions for use of the cells. Kits prepared for field hospital use, such as for military use may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute cardiac injuries.
The invention also provides for banking of tissues, cells, populations and therapeutic cell compositions of the invention. As discussed above the cells are readily cryopreserved. The invention therefore provides methods of cryopreserving the cells in a bank, wherein, the cells are stored frozen and associated with a complete characterization of the cells based on immunological, biochemical and genetic properties of the cells. The cells so frozeia can be used for autologous, syngeneic, or allogeneic therapy, depending on the requirements of the procedure and the needs of the patient. Preferably, the information on each cryopreserved sample is stored in a computer which is searchable based on the requirements of the surgeon, procedure and patient with suitable matches being made based on the characterization of the cells or populations. Preferably, the cells of the invention are grown and expanded to the desired quantity of cells and therapeutic cell compositions are prepared either separately or as cocultures, in the presence or absence of a matrix or support. While for some applications it Inay be preferable to use cells freshly prepared, the remainder can be cryopreserved and banked by freezing the cells and entering the information in the computer to associate the computer entry with the samples. Even where it is not necessary to match a source or donor with a recipient of such cells, for immunological purposes, the bank system makes it easy to match, for example, desirable biochemical or genetic properties of the banked cells to the therapeutic needs. Upon matching of the desired properties with a particle banked sample, the sample is retrieved and readied for therapeutic use. Cell lysates prepared as described herein may also be cryopreserved and banked in accordance with the present invention.
-39-In another aspect of the invention, kits for the growth and maintenance, the isolation and the use of the umbilical-derived cells are provided. The cells, cell lysates, soluble cell fractions, membrane fractions and matrices can conveniently be employed as parts of kits, for example, for a kit for culture or implantation. The invention provides a kit including the UDCs and additional components, including instructions for growth or maintenance, isolation, or use of the cells or cell fractions, together with for example, matrix (e.g., a scaffold) material, hydrating agents (e.g., physiologically-compatible saline solutions, prepared cell culture media), cell culture substrates (e.g-., culture dishes, plates, vials, etc.), cell culture media (whether in liquid or dehydrated faun), antibiotic compounds, hormones, and the like. Kits for giowth can for example include all of the components of the Growth Medium as used herein, including serum, for example fetal bovine serum. While the kit can include any such components, preferably it includes all ingredients necessary for its intended use. If desired, the kit also can include cells (typically cryopreserved), which can be seeded into the lattice as described herein. Kits for isolation will contain everything required to practice the isolation methods as provided herein, except for the umbilicus tissue which should be obtained fresh or frozen from a tissue bank at the time of isolation. The surgical equipment for dissociating the tissue, preferred enzymes, or choices of enzymes in stable form are provided, as are the buffers and medium, cell strainers and the like, as required or preferred for the method as disclosed above. Detailed instructions with optional steps and lists of suppliers of optional or alternative materials are abs conveniently provided. Control cells can be included for comparison of the cells isolated to, for example the UDC cultures deposited with the ATCC.
Kits for utilizing the umbilicus-derived cells preferably contain populations of the cells, or therapeutic compositions comprising the cells, components and products, or fractions or conditioned media derived from the cells as described above. In some embodiments, the kits may include one or more cell populations, including at least UDCs and a pharmaceutically acceptable carrier (liquid, semi-solid or solid). The populations in some embodiments are homogenous or even clonal cell lines of UDCs. In other embodiments, the kits include other cell lines for use in coculture. Therapeutic application kits preferably include additional bioactive agents as desired for example anithrombogenic agents, anti-inflammatory agents, antiapoptotic agents, and immunosuppressive or immunomodulatory compounds. The kits also optionally may include a means of administering the cells, for example by injection. The kits further may include instructions for use of the cells. Kits prepared for field hospital use, such as for military use, may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute injuries.
Kits for assays and in vitro methods as described herein may contain one or more of (1) LTDCs or fractions,
- 40 -components or products of UDCs, (2) reagents for practicing the in vitro method, (3) other cells or cell populations, as appropriate, for example for cocultures and (4) instructions for conducting the in vitro method. Kits for the preparation of cell-derived componenets can include both the components required for growth of the cells and the components required for preparing the cell fraction of interest, along with instructions for obtaining the desried fraction from the cells. Kits for production of and collection of conditioned media are also provided herein and include cells, medium, collection vessels, instructions, standards for assaying the secreted molecules of interest and the like.
The following examples describe several aspects of embodiments of the invention in greater detail. These examples are provided to further illustrate, not to limit, aspects of the invention described herein.

Isolation of Cells from Postpartum Tissues Summary Postpartum cells have been isolated from full- and pre-tei_in placental and umbilical cord tissues. A highly preferred way of isolating cells from these tissues is by using a combination of TM
digestive enzymes. Particularly preferred are collagenase, hyalusonidase and dispase. This combination results in the isolation of a cell population with good expansion and differentiation potentials. Other enzyme combinations used have yielded cell populations that can also be expanded.
Introduction Populations of cells from placental and umbilical cord tissues were isolated.
Postpartum umbilicus and placenta were obtained upon births of either fall- or pre-term pregnancies. Cells were harvested from five separate donors of umbilicus and placental tissue.
Different methods of cell isolation were tested for their ability to yield cells with: 1) the potential to differentiate into cells with different phenotypes, a characteristic common to stem cells, or 2) the potential to provide critical trophic factors useful for other cells and tissues.
Methods & Materials Umbilical cell isolation
-41-Umbilical cords were obtained from National Disease Research Interchange (NDRI
, Philadelphia, PA). The tissues were obtained following normal deliveries. The cell isolation protocol was performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, CA) in the presence of 10,000 Units of antimycotic and antibiotic per 100 milliliters of PBS
(Invitrogen Carlsbad, CA). The tissues were then mechanically dissociated in 150 cm2 tissue culture plates in the presence of 50 milliliters of medium (DMEM-Low glucose or DMEM-High glucose;
Invitrogen), until the tissue was minced into a fine pulp. The chopped tissues were transferred to 50 milliliter conical tubes (approximately 5 grams of tissue per tube). The tissue was then digested in either DMEM-Low glucose medium or DMEM-High glucose medium, each containing 10,000 Units of antimycotic and antibiotic per 100 milliliters of PBS and digestion TM
enzymes. In some experiments an enzyme mixture of collagenase and dispase was used ("C:D;"
TM
collagenase (Sigma, St Louis, MO), 500 Units/milliliter; and dispase (Invitrogen), 50 Units/milliliter in DMEM:-Low glucose medium). In other experiments a mixture of TM
collagenase, dispase and hyaluronidase ("C:D:H") was used (collagenase, 500 Units/milliliter;
dispasTMe, 50 Units/milliliter; and hyaluronidase (Sigma), 5-Units/milliliter, in DMEM:-Low glucose). The conical tubes containing the tissue, medium and digestion enzymes were incubated at 37 C in an orbital shaker (Environ, Brooklyn, NY) at 225 rpm for 2 hrs.
After digestion, the tissues were centrifuged at 150 x g for 5 minutes, the supernatant was aspirated. The pellet was resuspended in 20 milliliters of Growth Medium (DMEM:Low glucose (Invitrogen), 15 percent (v/v) fetal bovine serum (14'13S; defined bovine serum; Lot#AND18475;
Hyclone, Logan, UT), 0.001% (v/v) 2-mercaptoethanol (Sigma), 1 milliliter per 100 milliliters of antibiotic/antimycotic (10,000 Units per milliliter penicillin, 10,000 micrograms per milliliter streptomycin, 25 micrograms per milliliter amphotericin B; Invitrogen, Carlsbad, CA)). The cell suspension was filtered through a 70-micrometer nylon cell strainer (Nalge Nunc International, Rochester, NY). An additional 5 milliliters rinse comprising Growth Medium was passed through the strainer. The cell suspension was then passed through a 40-micrometer nylon cell strainer (Nalge Nunc International) and chased with a rinse of an additional 5 milliliters of Growth Medium.
The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 x g for 5 minutes. The supernatant was aspirated. and the cells were resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more.
Upon the final centrifugation supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. The number of viable cells was
- 42 -determined using trypan blue staining. Cells were then cultured -under standard conditions.
The cells isolated from umbilical cord cells were seeded at 5,000 cells/cm2 onto gelatin-coated T-75 cm2 flasks (Corning Inc., Corning, NY) in Growth Medium. After 2 days, spent medium was aspirated from the flasks. Cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with Growth Medium and allowed to grow to confluence (about 10 days from passage 0) to passage 1. On subsequent passages (from passage 1 to 2 etc), cells reached sub-confluence (75-85 percent confluence) in 4-5 days.
For these subsequent passages, cells were seeded at 5000 cells/crn2. Cells were grown in a humidified incubator with 5 percent carbon dioxide and 20 percent oxygen, at 37 C.
Placental Cell Isolation Placental tissue was obtained from NDRI (Philadelphia, PA). The tissues were from a pregnancy and were obtained at the time of a normal surgical delivery.
Placental cells were isolated as described for umbilical cell isolation.
The following description applies to the isolation of separate populations of maternal-derived and neonatal-derived cells from placental tissue.
The cell isolation protocol was performed aseptically in a laminar flow hood.
The placental tissue was washed in phosphate buffered saline (PBS; invitrogen, Carlsbad, CA) in the presence of antimycotic and antibiotic (penicillin, 10,000 Units/milliliter;
streptomycin, 10,000 micrograms/milliliter; amphotericin B, 25 micrograms /milliliter; Invitrogen) to remove as much blood and debris as practical. The placental tissue was then dissected into three sections:
neonatal aspect, the villous region, the maternal aspect.
The separated sections were individually washed several times in PBS with antibiotic/antimycotic to further remove blood and debris. In this manner, substantially all the blood was removed. Each section was then mechanically dissociated in 150 cm2 tissue culture plates in the presence of 50 milliliters of DMEM:Low glucose (Ixivitrogen), to a fine pulp. The pulp was transferred to 50 milliliter conical tubes. Each tube contained approximately 5 grams of tissue. The tissue was digested in either DMEM-Low glucose DMEM-High glucose medium containing 10,000 Units of antimycotic and antibiotic per 100 milliliters of PBS and digestion TM
enzymes. In some experiments an enzyme mixture of collagenase and dispase ("C:D") was used containing collagenase (Sigma, St Louis, MO) at 500 Units/milliliter and dispaserm(Invitrogen) at 50 Units/milliliter in DMEM:-Low glucose medium. In other experiments, a mixture of collagenase, dispasrand hyaluronidase (C:D:H) was used (collagenase, 500 Units/milliliter;
-43 -= = CA 02530422 2014-04-07 dispaser,m50 Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter in DMEM:-Low glucose). The conical tubes containing the tissue, medium, and digestion enzymes were incubated for 2 h at 37 C in an orbital shaker (Environ, Brooklyn, NY) at 225 rpm.
After digestion, the tissues were centrifuged at 150 x g for 5 minutes, the resultant supernatant was aspirated off. The pellet was resuspended in 20 milliliters of Growth Medium.
The cell suspension was filtered through a 70 micrometer nylon cell strainer (Nalge Nunc International, Rochester, NY), and rinsed with 5 milliliters of Growth Medium.
The total cell suspension was passed through a 40 micrometer nylon cell strainer (Nalge Nunc International) and rinsed with an additional 5 milliliters of Growth Medium.
The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 x g for 5 minutes. The supern.atant was aspirated and the cell pellet was resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more. .
After the final centrifugation, supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. A cell count was determined using the trypan blue Exclusion test. Cells were cultured under standard conditions.
=
LIBERASE Cell Isolation Cells were isolated from postpartum tissues in DlVfEM-Low glucose medium with L1BERASE (2.5 milligrams per milliliter, Blendzyme 3; (Roche Applied Sciences, Indianapolis, ENT)) and hyaluronidase (5 Units/milliliter, Sigma). Digestion of the tissue and isolation of the cells was as described for other digestions above, with the L1BERASE/hyaluronidase mixture used instead of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE resulted in the isolation of cell populations from postpartum tissues that expanded readily.
Cell isolation using other enzyme combinations Procedures were compared for isolating cells from the umbilical cord using differing enzyme combinations. Enzymes compared for digestion included: i) collagenase;
ii) dispaseTiii) TM
hyaluronidase; iv) collagenase:dispase mixture (C:D); v) collagenase:hyaluronidase mixture (C:H); vi) dispas-rme:hyaluronidase mixture (D:H); and vii) collagenase:dispase:hyaluronidase mixture (C:D:H). Differences in cell isolation utilizing these different enzyme digestion conditions were observed (Table 1-1).
Isolation of cells from residual blood in the cords
- 44-Other attempts were made to isolate pools of cells from umbilical cords by different approaches. In one instance umbilical cord was sliced and washed with Growth Medium to dislodge the blood clots and gelatinous material. The mixture of blood, gelatinous material and Growth Medium was collected and centrifuged at 150 x g. The pellet was resuspended and seeded onto gelatin coated flasks in Growth Medium. Cell populations that readily expanded were isolated.
Isolation of cells from Cord Blood Cells were also isolated from cord blood samples attained from NDRI. The isolation protocol used here was that of International Patent Application WO 03/025149 by Ho et al. Samples (50 milliliter and 10.5 milliliters, respectively) of umbilical cord blood (NDRI, Philadelphia PA) were mixed with lysis buffer (filter-sterilized 155 millimolar ammonium chloride, 10 millimolar potassium bicarbonate, 0.1 millimolar EDTA buffered to pH 7.2 (all components from Sigma, St.
Louis, MO)). Cells were lysed at a ratio of 1:20 cord blood to lysis buffer.
The resulting cell suspension was vortexed for 5 seconds, and incubated for 2 minutes at ambient temperature. The lysate was centrifuged (10 minutes at 200 x g). The cell pellet was resuspended in complete minimal essential medium (Gibco, Carlsbad CA) containing 10 percent fetal bovine serum (Hyclone, Logan UT), 4 millimolar glutamine (Mediatech Herndon, VA), 100 Units penicillin per 100 milliliters and 100 micrograms streptomycin per 100 milliliters (Gibco, Carlsbad, CA).
The resuspended cells were centrifuged (10 minutes at 200 x g), the supernatant was aspirated, and the cell pellet was washed in complete medium. Cells were seeded directly into either T75 flasks (Corning, NY), T75 laminin-coated flasks, or T175 fibronectin-coated flasks (both Becton Dickinson, Bedford, MA).
Isolation of postpartum cells using different enzyme combinations and growth conditions To determine whether cell populations can be isolated under different conditions and expanded under a variety of conditions immediately after isolation, cells were digested in Growth Medium with or without 0.001 percent (v/v) 2-mercaptoethanol (Sigma, St. Louis, MO), using the C:D:H enzyme combination, according to the procedures provided above. Placental cells so isolated were seeded under a variety of conditions. All cells were grown in the presence of penicillin/streptomycin. (Table 1-2).
Isolation of postpartum cells using different enzyme combinations and growth conditions
- 45 -In all conditions tested, cells attached and expanded well from about passage 0 to 1 (Table 1-2). Cells in conditions 5-8 and 13-16 proliferated well up to 4 passages after seeding at which point they were cryopreserved. All cells were banked for later investigation.
Results Cell isolation using different enzyme combinations The combination of C:D:H enzymes provided the best cell yield following isolation, and generated cells which expanded for many more generations in culture than the other conditions (Table 1). An expandable cell population was not attained using collagenase or hyaluronidase alone. No attempt was made to determine if this result is specific to the collagenase that was tested.
Isolation of postpartum cells using different enzyme combinations and growth conditions Cells attached and expanded well from about passage 0 to 1 under all conditions tested for enzyme digestion and growth (Table 1-2). Cells in experimental conditions 5-8 and 13-16 proliferated well up to 4 passages after seeding, at which point they were cryopreserved. All cells were banked for further investigation.
Isolation of cells from residual blood in the cords Nucleated cells attached and grew rapidly. These cells were analyzed by flow cytometry and were similar to cells obtained by enzyme digestion.
Isolation of cells from Cord Blood The preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.
Discussion and Conclusion Populations of cells can be isolated from umbilical and placental tissue most efficiently using the enzyme combination collagenase (a metalloprotease), dispasTMe (a neutral protease) and hyaluronidase (a mucolytic enzyme which breaks down hyaluronic acid). LIBERASE

Blendzyme, which is a commercial blend of collagenase and another protease may also be used.
In the present study Blendzyme 3 which contains collagenase (4 Wunsch units/g) and thermolysin (1714 casein Units/g) was also used together with hyaluronidase to isolate cells.
- 46 -=
Cells isolated with these enzymes expand readily over many passages, for example, when cultured in Growth Medium on gelatin coated plastic.
Cells were also isolated from residual blood in the cords, but not cord blood.
The presence of cells in blood clots washed from the tissue, that adhere and grow under the conditions used, may be due to cells being released during the dissection process. Other explanations may include the migration of cells from the matrix.
Recommendation Use of C:D:H enzyme combinations for isolation of cell populations from postpartum tissues is preferred. Cells isolated using this combination of enzymes have been extensively characterized and have many desirable properties. LIBERASE extracted cells and cells treated with other enzyme combination cells provide useful cells with expansion potential. It may be useful to choose a process or method for cell isolation that helps minimize handling and transfer of the tissue. Such methods may include mechanical digestion for example with a blender, tissue homogenizer, and the like.
Reference sl. HO, Tony, W. ;KOPEN, Gene, C. ;RIGHTER, William, F.
;RUTKOWSKI, J., Lynn ;HERRING, W., Joseph ;RAGAGLIA, Vanessa ;WAGNER, Joseph W02003025149 A2 CELL POPULATIONS WHICH CO-EXPRESS CD49C AND CD90, NEURONYX, INC.
Application No. US0229971 US, Filed 20020920, A2 Published 20030327, A3 Published Table 1-1: Isolation of cells from umbilical cord tissue using varying enzyme combinations Enzyme Digest Cells Isolated Cell Expansion Collagenase X X
Dispase TM + h) Hyaluronidase X X
Collagenase:DispaseN -H- (<3 h) -H-Collagenase:Hyaluronidase -H- (<3 h) Dispaellyaluronidase + (>10 h) Collagenase:Dispasnlyaluronidase +-H- (<3 h) -H-+
Key: + = good, -H- = very good, +-H- = excellent, .X = no success
-47 -Table 1-2: Isolation and culture expansion of postpartum cells under varying conditions:
Condition Medium 15% FBS BME Gelatin 20% 02 Growth. Factors 1 DMEM-Lg Y Y Y Y N
2 DMEM-Lg Y Y Y N (5%) N
3 DMEM-Lg Y Y N Y N
4 DMEM-Lg Y Y N N (5%) N
DMEM-Lg N (2%) Y N (Laminin) Y EGF/FGF (20 ng/rnilliliter) 6 DMEM-Lg N (2%) Y N (Laminin) N (5%) EGF/FGF (20 ng/rnilliliter) 7 \ DMEM-Lg N (2%) Y N (Fibrone) Y PDGF/VEGF
8 DMEM-Lg N (2%) Y N (Fibrone) N (5%) PDGF/VEGF
9 DMEM-Lg Y N Y Y N
DMEM-Lg Y N Y N (5%) N
11 DMEM-Lg Y N N Y N
12 DMEM-Lg Y N N N (5%) N
13 DMEM-Lg N (2%) N N (Laminin) Y EGF/FGF (20 ng/rnilliliter) 14 DMEM-Lg N (2%) N N (Laminin) N (5%) EGF/FGF (20 ng/rnilliliter) DMEM-Lg N (2%) N N (Fibrone) Y PDGF/VEGF
16 DMEM-Lg N (2%) N N (Fibrone) N (5%) PDGF/VEGF

Growth Characteristics of Postpartum Cells Summary Commercially viable cell products must be able to be produced in sufficient qu_antities to provide therapeutic treatment to patients in need of the treatment. Postpartum cells can be expanded in culture for such purposes. Comparisons were made of the growth of postpartum cells in culture to that of other cell populations including mesenchymal stem cells. The data
- 48 -demonstrate that postpartum cell lines as developed herein can expand for greater than 40 doublings to provide sufficient cell numbers, for example, for pre-clinical banks. Furthermore, these postpartum cell populations can be expanded well from low- or high-density seeding. This study has demonstrated that mesenchymal stem cells, in contrast, cannot be expanded to obtain large quantities of cells.
Introduction The cell expansion potential of postpartum cells was compared to other populations of isolated stem cells. The art of cell expansion to senescence is referred to as Hayflick's limit (Hayflick L. The longevity of cultured human cells. J. Am. Geriatr. Soc.
22(1):1-12, 1974;
Hayflick L. The strategy of senescence. Gerontologist 14(1):37-45), 1974).
Senescence is defined as the point at which cell division stops completely, i.e., when the cell loses its ability to proliferate and expand. Postpartum-derived cells are highly suited for therapeutic use because they can be readily expanded to sufficient cell numbers.
Materials and Methods Gelatin-coating flasks Tissue culture plastic flasks were coated by adding 20 milliliters of a 2%
(w/v) gelatin (Type B: 225 Bloom; Sigma, St Louis, MO) solution each to T75 flasks (Corning, Corning, NY) for 20 minutes at room temperature. After removing the gelatin solution, 10 milliliters phosphate-buffered saline (PBS) (Invitrogen, Carlsbad, CA) were added and then aspirated.
Comparison of expansion potential of postpartum cells vs. other stem cell and non-stem cell populations For comparison of growth expansion potential the following cell populations were utilized; i) Mesenchymal stem cells (MSC; Cambrex, Walkersville, MD); ii) Adipose-derived cells (US6555374 Bl; U.S. Patent Application US20040058412); iii) Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, MD); iv) Umbilical-derived cells; and vi) Placental-derived cells. Cells were initially seeded at 5,000 cells/cm2 on gelatin-coated T75 flasks in Growth Medium.
For subsequent passages, cell cultures were treated as follows: After trypsinization, viable cells were counted after trypan blue staining. Cell suspension (50 microliters) was combined with trypan blue (50 ml, Sigma, St. Louis MO). Viable cell numbers were estimated using a hemocytometer.
- 49 -Following counting, cells were seeded at 5,000 cells/cm2 onto gelatin-coated T
75 flasks in 25 ml of fresh Growth Medium. Cells were grown under standard atmospheric conditions (5 percent carbon dioxide) at 37 C in the presence of 20 percent oxygen and 75 percent nitrogen (v/v). The Growth Medium was changed twice per week When cells reached about 85 percent confluence they were passaged; this process was repeated until the cells reached senescence.
At each passage, cells were trypsinized and counted. The viable cell yield, population doubling [ln (cell final/cell initial)/1n2] and doubling time (time in culture (h)/population doubling) were calculated. For the purposes of determining optimal cell expansion, the total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e. expansion factor = cell final/cell initial).
Expansion of potential of cell banks at low density The expansion potential of cells banked at passage 10 was also tested. A
different set of conditions was used. Normal dermal skin fibroblasts (cc-2509 lot # 9F0844;
Cambrex, Walkersville, MD), umbilical-derived cells, and placenta-derived cells were tested. These cell populations had been banked at passage 10 previously, having been cultured at 5,000 cells/cm2 at each passage to that point. The effect of cell density on the cell populations following cell thaw at passage 10 was determined. Cells were thawed under standard conditions, counted using trypan blue staining. Thawed cells were then seeded at 1000 cells/cm2 in Growth Medium.
Cells were grown under standard atmospheric conditions at 37 C. Growth Medium was changed twice a week and cells were passaged as they reached about 85% confluence.
Cells were subsequently passaged until senescence, i.e., until they could not be expanded any further. Cells were trypsinized and counted at each passage. The cell yield, population doubling (ln (cell final/cell initial)/1n2) and doubling time (time in culture (h)/population doubling). The total cell yield per passage was determined by multiplying total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor = cell final/cell initial).
Expansion of postpartum cells at low density from initial cell seeding The expansion potential of freshly isolated postpartum cell cultures under low cell seeding conditions was tested in another experiment. Umbilical and placental cells were isolated as described herein. Cells were seeded at 1000 cells/cm.2 and passaged as described above until senescence. Cells were grown under standard atmospheric conditions at 37 C.
Growth Medium was changed twice per week. Cells were passaged as they reached about 85%
confluence. At each passage, cells were trypsinized and counted by trypan blue staining. The cell yield,
-50-population doubling (In (cell final/cell initial)/1n2) and doubling time (time in culture (h)/population doubling) were calculated for each passage. The total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e. expansion factor = cell final/cell initial). Cells were grown on gelatin and non¨
gelatin coated flasks.
Expansion of Clonal Neonatal Placental Cells Cloning was used in order to expand a population of neonatal cells successfully from placental tissue. Following isolation of three differential cell populations from the placenta (as described herein), these cell populations were expanded under standard growth conditions and then karyotyped to reveal the identity of the isolated cell populations. Since these cells were isolated from a mother who delivered a boy it was very simple to distinguish between the male and female chromosomes by performing metaphase spreads. These experiments demonstrated that cells isolated from the neonatal aspect were primarily karyotype-positive for neonatal phenotpye, and cells isolated from the maternal aspect were primarily karyotype-positive for maternal cells, those cells isolated from the villous region were karyotype-positive for both neonatal and maternal phenotypes. Subcloning of populations derived from neonatal and maternal aspects is required to ensure that clonal populations are obtained for both neonatal and maternal cells.
Expansion of cells in low oxygen culture conditions It has been demonstrated that low 02 cell culture conditions can improve cell expansion in certain circumstances (Csete, Marie; Doyle, John; Wold, Barbara J.; McKay, Ron; Studer, Lorenz. Low oxygen culturing of central nervous system progenitor cells.
U520040005704).
order to determine if cell expansion of postpartum-derived cells could be improved by altering cell culture conditions, cultures of umbilical-derived cells were grown in low oxygen conditions_ Cells were seeded at 5000 cells/cm2 in Growth Medium on gelatin coated flasks.
Cells were initially cultured under standard atmospheric conditions through passage 5, at which point they were transferred to low oxygen (5% 02) culture conditions.
Other growth conditions In other experiments cells were expanded on non-coated, collagen-coated, fibronectin-coated, laminin-coated and Matrigel-coated plates. Cultures have been demonstrated to expand well on these different matrices.
- 51 -Results Comparison of expansion potential of postpartum cells vs. other stein cell and non-stein cell populations Both umbilical-derived and placenta-derived cells expanded for greater than 40 passages generating cell yields of > 1E17 cells in 60 days. In contrast, MSCs and fibroblasts senesced after <25 days and <60 days, respectively. Although both adipose-derived and omental cells expanded for almost 60 days they generated total cell yields of 4.5E12 and 4.24E13 respectively.
Thus, when seeded at 5000 cells/cm2 under the experimental conditions utilized, postpartum-derived cells expanded much better than the other cell types grown under the same conditions (Table 1).
Expansion of potential of cell banks at low density Umbilical-derived, placental-derived and fibroblast cells expanded for greater than 10 passages generating cell yields of > 1E11 cells in 60 days (Table 2). After 60 days under these conditions the fibroblasts became senescent whereas the umbilical-derived and placental-derived cell populations senesced after 80 days, completing >50 and > 40 population doublings respectively.
Expansion of postpartum cells at low density from initial cell seeding Placental-derived cells were expanded at low density (1,000 cells/cm2) on gelatin-coated and uncoated plates or flasks. Growth potential of these cells under these conditions was good.
The cells expanded readily in a log phase growth. The rate of cell expansion was similar to that observed when placental-derived cells were seeded at 5000 cells/cm2 on gelatin-coated flasks in Growth Medium. No differences were observed in cell expansion potential between culturing on either uncoated flasks or gelatin-coated flasks. However, cells appeared phenotypically much smaller on gelatin-coated flasks and more larger cell phenotypes were observed on uncoated flasks.
Expansion of Clonal Neonatal and Maternal Placental Cells The expansion of a clonal cell populations of placental¨derived cells isolated from the neonatal and maternal aspects of the placenta are studied. Populations derived from neotal and maternal aspects are serially diluted and then seeded onto gelatin-coated plates in Growth Medium for expansion at 1 cell/well in 96-well gelatin coated plates. From this initial cloning,
- 52 -expansive clones are identified, trypsinized and reseeded in 12 well gelatin coated plates in Growth Medium and then subsequently passaged into T25 gelatin coated flasks at 5,000 cells/cm2 in Growth Medium. Subcloning is then performed to ensure that a clonal population of cells had been identified. For subcloning experiments cells are trypsinized and reseeded at 0.5 cells/well. The subclones that grow well are then expanded in gelatin-coated T25 flasks at 5,000 cells/cm2. Cells are subsequently passaged at 5,000 cells/cm2 in T75 flasks.
Karyotypiag confirms that clones so-derived are neotal or maternal in nature.
Expansion of cells in low oxygen culture conditions Cells expanded well under the reduced oxygen conditions, however, culturing under low oxygen conditions does not appear to have a significant effect on cell expansion for postpartum-derived cells. These results are preliminary in the sense that any ultimate conclusions to be made regarding the effect of reduced oxygen should include data from experiments on growing cells in low oxygen from initial isolation. Standard atmospheric conditions have already proven_ successful for growing sufficient numbers of cells, and low oxygen culture is compatible with, but not required for, the growth of postpartum-derived cells.
Discussion and Conclusions The current cell expansion conditions of growing isolated postpartum-derived cells at densities of about 5000 cells/cm2, in Growth Medium on gelatin-coated or uncoated flasks, under standard atmospheric oxygen, are sufficient to generate large numbers of cells at passage 11.
Furthermore, the data suggest that the cells can be readily expanded using lower density culture conditions (e.g. 1000 cells/cm2). Postpartum-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for growth. Presently, culturing postpartum-derived cells under standard atmospheric conditions is preferred for generating large pools of cells. However, when the culture conditions are altered, postpartum-derived cell expansion can likewise be altered. This strategy may be used to enhance the proliferative and differentiative capacity of these cell populations.
Under the conditions utilized, while the expansion potential of MSC and adipose-derived cells is limited, postpartum-derived cells expand readily to large numbers.
Recommendations:
- 53 -In order to optimize expansion and scale-up of postpartum-derived cell cultures additional work with new methods for cell expansion, media conditions, extracellular matrix conditions for cellular attachment and cell density would be useful. However, with all these changes the cell potential would have to be re-determined.
References 1) Hayflick L. The longevity of cultured human cells. J Am Geriatr Soc. 1974 Jan;22(1):1-12.
2) Hayflick L. The strategy of senescence. Gerontologist. 1974 Feb;14(1):37-45.
3) Patent US20040058412 4) Patent US20040048372 6) Csete, Marie; (Ann Arbor, MI) ; Doyle, John; (South Pasadena, CA) ; Wold, Barbara J.; (San Marino, CA) ; McKay, Ron; (Bethesda, MD) ; Studer, Lorenz; (New York, NY). Low oxygen culturing of central nervous system progenitor cells. US20040005704.
Table 1: Growth characteristics for different cell populations grown to senescence Cell Type Senescence Total Population Total Cell Doublings Yield MSc 24d 8 4.72E7 Adipose-derived 57 d 24 4.5 E12 Fibroblasts 53 d 26 2.82 E13 Umbilical 65 d 42 6.15 E17 Placenta 80 d 46 2.49 E19 Table 2: Growth characteristics for different cell populations using low density growth expansion from passage 10 till senescence Cell Type Senescence Total Population Total Cell Doublings Yield Fibroblast (P10) 80 d 43.68 2.59 Ell Umbilical (P10) 80d 53.6 1.25 E14 Placental (P10) 60 d 32.96 6.09 E12
- 54 -Growth of Postpartum cells in Medium containing D-Valine Summary Culture media containing D-valine instead of the L-valine isoform reportedly selectively inhibit the growth of fibroblast-like cells in culture. To determine whether postpartum-derived cells can grow in medium containing D-valine, cells derived from placenta and umbilical cord were grown in medium containing D-valine for 4 weeks. The cells did not proliferate and eventually died. Medium containing D-valine is not suitable for selectively growing postpartum¨
derived cells. L-valine is required for postpartum-derived cell proliferation and long-term viability.
Introduction It has been reported that medium containing D-valine instead of the normal L-valine isoform can be used to selectively inhibit the growth of fibroblast-like cells in culture (Hongpaisan, 2000; Sordillo et al., 1988). It was not previously known whether postpartum-derived cells can grow in medium containing D-valine.
Methods & Materials Placenta-derived cells (P3), fibroblasts (P9) and umbilical-derived cells (P5) were seeded at 5 x 103 cells/cm2 in gelatin-coated T75 flasks (Corning, Corning, NY).
After 24 hours the medium was removed and the cells were washed with phosphate buffered saline (PBS) (Gibco, Carlsbad, CA) to remove residual medium. The medium was replaced with a Modified Growth Medium (DMEM with D-valine (special order Gibco), 15% (v/v) dialyzed fetal bovine serum (Hyclone, Logan, UT), 0.001% (v/v) betamercaptoethanol (Sigma), penicillin/streptomycin (Gibco)).
Results Placenta-derived, umbilical-derived, and fibroblast cells seeded in the D-valine-containing medium did not proliferate, unlike cells seeded in Growth Medium containing dialyzed serum. Fibroblasts cells changed morphologically, increasing in size and changing shape. All of the cells died and eventually detached from the flask surface after 4 weeks.
Discussion and Conclusion
- 55 -Postpartum-derived cells require L-valine for cell growth and to maintain long-term viability. L-valine should therefore not be removed from the Growth Medium for postpartum-derived cells.
References Hongpaisan J. (2000) Inhibition of proliferation of contaminating fibroblasts by D-valine in cultures of smooth muscle cells from human myometrium. Cell Biol Int. 24:1-7.
Sordillo LM, Oliver SP, Akers RM. (1988) Culture of bovine mammary epithelial cells in D-valine modified medium: selective removal of contaminating fibroblasts. Cell Biol Int Rep.12:355-64.

Karyotype Analysis of PPDCs Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type. Cells used in cell therapy should have a normal chromosome number (46) and structure. To identify postpartum placental and umbilical cord cell lines that are homogeneous and free from cells of non-postpartum tissue origin, karyotypes of cell samples were analyzed.
Materials and Methods PPDCs from postpartum tissue of a male neonate were cultured in Growth Media.
Postpartum tissue from a male neonate (X,Y) was selected to allow distinction between neonatal-derived cells and maternal derived cells (X,X). Cells were seeded at 5,000 cells per square centimeter in Growth Medium in a T25 flask (Corning, Coming, NY) and expanded to 80%
confluence. A T25 flask containing cells was filled to the neck with Growth Medium. Samples were delivered to a clinical cytogenetics lab by courier (estimated lab to lab transport time is one hour). Chromosome analysis was performed by the Center for Human & Molecular Genetics at the New Jersey Medical School, Newark, NJ. Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two). A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells w-ere counted and analyzed when a heterogeneous karyotype number (four) was identified.
-56-Results All cell samples sent for chromosome analysis were interpreted by the cytogenetics laboratory staff as exhibiting a normal appearance. Three of the sixteen cell lines analyzed exhibited a heterogeneous phenotype (XX and XY) indicating the presence of cells derived from both neonatal and maternal origins (Table 4-1). Cells derived from tissue Placenta-N were isolated from the neonatal aspect of placenta. At passage zero, this cell line appeared homogeneous XY. However, at passage nine, the cell line was heterogeneous (XX/XY), indicating a previously undetected presence of cells of maternal origin.
Table 4-1. Karyotype results of PPDCs.
[ 1 [issue passage Metaphase cells counted Metaphase cells analyzed mber of karyotype ISCN Karyotype Placenta 22 20 5 2 46, XX
Umbilical 23 20 5 2 46, XX
Umbilical 6 20 b 2 46, XY
Placenta 2 20 5 2 46, )0( , Umbilical 3 20 5 2 46,)0( Placenta-N 0 20 5 2 _ 46, XY
Placenta-V 0 20 5 2 46, XY
Placenta-M 0 21 5 4 46, XY[18]/46,XX[3]
Placenta-M 4 20 5 2 46,)0( Placenta-N 9 25 b 4 46, XY151140,XX[20]
Placenta-N Cl 1 20 b 2 46, XY
Placenta-N C3 1 20 6 4 46, XY[21/46,X)q1 8]
Placenta-N C4 1 20 6 2 46, XY
Placenta-N C15 1 20 5 2 46, XY
Placenta-N C20 1 20 5 2 46, XY
Key: N- Neonatal aspect; V- villous region; M- maternal aspect; C- clone Summary. Chromosome analysis identified placenta- and umbilical cord-derived PPDCs whose karyotypes appear normal as interpreted by a clinical cytogenetic laboratory.
Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.
= EXAMPLE 5 Evaluation of Postpartum-Derived Cell Surface Markers by Flow Cytometry Summary Characterization of cell surface protein expression, or "markers" by flow cytometry of cultured cell lines labeled with fluorescent monoclonal antibodies enables the determination of a
- 57 -cell line's identity. Placental and umbilicus-derived postpartum cells were characterized by flow cytometry for the expression of cell surface markers CD10, CD13, CD31, CD34, CD44, CD45, CD73, CD90, CD117, CD141, PDGFr-alpha, HLA-A,B,C and HLA-DR, DP,DQ. Both placenta- and umbilicus-derived postpartum cells are positive for the expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A, B, C and negative for the expression of CD31, CD34, CD44, CD45, CD73, CD90, CD117, CD141and HLA-DR, DP, DQ. This expression pattern was consistent across variables such as cell donor, passage, culture vessel surface coating, and digestion enzymes used in isolation. This expression pattern was also consistent in cells isolated from the maternal aspect, neonatal aspect and villous region of the placenta.
Introduction Characterization of cell surface proteins or "markers" by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions.
Postpartum cell lines isolated from the placenta and umbilicus were characterized (by flow cytometry) providing a profile for the identification of these cell lines.
Materials and Methods Media Cells were cultured in Growth Media.
Culture Vessels Cells were cultured in plasma-treated T75, T150, and T225 tissue culture flasks (Corning, Corning, NY) until confluent. The growth surfaces of the flasks were coated with gelatin by incubating 2% (w/v) gelatin (Sigma, St. Louis, MO) for 20 minutes at room temperature.
Antibody Staining Adherent cells in flasks were washed in phosphate buffered saline (PBS);
(Gibco, Carlsbad, MO) and detached with Trypsin/EDTA (Gibco, Carlsbad, MO). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of lx107 per milliliter. In accordance to the manufacture's specifications, antibody to the cell surface marker of interest (see below) was added to one hundred microliters of cell suspension and the mixture was incubated in the dark for 30 minutes at 4 C. After incubation, cells were washed with PBS
- 58 -and centntugect to remove unbouna antibody. Cells were resuspended in 500 microliters PBS
and analyzed by flow cytometry.
Flow Cytometty Analysis Flow cytometry analysis was performed with a FACScalibur instrument (Becton Dickinson, San Jose, CA).
Antibodies to Cell Surface Markers The following antibodies to cell surface markers were used.
Antibody Manufacture Catalog Number CD10 BD Pharmingen (San Diego, CA) 555375 CD13 BD Pharmingen 555394 CD31 BD Pharmingen 555446 CD34 BD Pharmingen 555821 CD44 BD Pharmingen 555478 CD45RA BD Pharmingen 555489 CD73 BD Pharmingen 550257 CD90 BD Pharmingen 555596 CD117 BD Pharmingen 340529 CD141 BD Phanningen 559781 PDGFr-alpha BD Pharmingen 556002 HLA-A, B, C BD Pharmingen 555553 HLA-DR, DP, DQBD Pharmingen 555558 IgG-FITC Sigma (St. Louis, MO) F-6522 IgG-PE Sigma P-4685 Placenta and Umbilicus Comparison Placenta cells were compared to umbilicus at passage 8.
Passage to Passage Comparison Placenta and umbilicus were analyzed at passages 8, 15, and 20.
Donor to Donor Comparison To compare differences among donors, placenta cells from different donors were compared to each other, and umbilical from different donors were compared to each other.
Surface Coating Comparison
-59-Placenta cultured on gelatin-coated flasks was compared to placenta cultured on uncoated flasks. Umbilicus cultured on gelatin-coated flasks was compared to umbilicus cultured on uncoated flasks.
Digestion Enzyme Comparison Four treatments used for isolation and preparation of cells were compared.
Cells isolated from placenta by treatment with 1) collagertase; 2) co1lagenase/dispase;13) collagenase/hyaluronidase; and 4) collagenase/hyaluronidase/dispasTMe were compared.
Placental Layer Comparison Cells isolated from the maternal aspect of placental tissue were compared to cells isolated from the villous region of placental tissue and cells isolated from the neonatal fetal aspect of placenta.
Results Placenta vs. Umbilicus Comparison Placental- and umbilical-derived cells analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for detectable expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values comparable to the IgG control. Variations in florescence values of positive curves was accounted for. The mean (i.e. CD13) and range (i.e. CD90) of the positive curves showed some variation, but the curves appear normal, confirming a homogenous population. Both curves individually exhibited values greater than the IgG
control.
Passage to Passage Comparison- Placenta Placenta-derived cells at passages 8, 15, and 20 analyzed by flow cytometry all were positive for expression of CD 10, CD13, CD44, CD73, CD90, PDGFr-alpha and 131.,A-A, B, C, as reflected in the increased value of fluorescence relative to the IgG control.
The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ
having fluorescence values consistent with the IgG control. Variations in florescence detection values of the positive curves was accounted for. While range (i.e. CD10) of the positive curves varied, the curves were normal, confirming a homogenous population, and each curves individually exhibited values greater than the IgG control.
- 60 -Passage to Passage Comparison- Umbilicus Umbilical cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were within expected ranges. While the means (i.e. CD13) of the positive curves varied all curves individually exhibited values greater than the IgG control.
Donor to Donor Comparison- Placenta Placenta-derived cells isolated from separate donors analyzed by flow cytorrietry each expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence value consistent with the IgG control. Variations in florescence detection values of positive curves are within expected ranges. While the range (i.e. CD44) of the positive curves varied, the curves appeared normal, confirming a homogenous population, and both curves individually exhibit values greater than the IgG control.
Donor to Donor Comparison- Umbilicus Umbilical-derived cells isolated from separate donors analyzed by flow cytometry each showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG
control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ
with fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were accounted for. While the mean (i.e. CD10) of the positive curves varied, both curves individually exhibited values greater than the IgG control.
The Effect of Surface Coating with Gelatin on Placenta-derived Cells Placenta-derived cells expanded on either gelatin-coated or uncoated flasks analyzed by flow cytometry all expressed of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG
control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ
- 61 -indicated by fluorescence values consistent with the IgG control. Variations in florescence detection values of positive curves were noted. While the mean (i.e. PDGFr-alpha) of the positive curves varied both curves individually exhibited values greater than the IgG control.
The Effect of Surface Coating with Gelatin on Umbilicus-derived Cells Umbilical cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
Does the Enzyme Digestion Procedure Used for Preparation and Isolation of the Cells -Effect the Cell Surface Marker Profile?
Placenta cells isolated using various digestion enzymes analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD3 1, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control. Variations in the CD13 mean florescence value were noted. While the CD13 mean fluorescence values of the collagenase-treated cells was less than the other CD13 curves, the collagenase-treated curve appeared normal, confirming a homogenous population, and it individually exhibited values greater than the IgG control.
Placental Layer Comparison Cells isolated from the maternal, villous, and neonatal layers of the placenta, respectively, analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased value of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG
control. Variations in florescence detection values of positive curves were noted. While the mean and range (i.e. CD10, CD73) of the positive curves varied, both curves appeared normal, confirming a homogenous population, and all curves individually exhibit values greater than the IgG control.
Conclusions
- 62 -Analysis of placenta- and umbilicus-derived postpartum cells by flow cytometry has established of an identity of these cell lines. Placenta- and umbilicus-derived postpartum cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A,B,C and negative for CD31, CD34, CD45, CD117, CD141and HLA-DR, DP, DO. This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, digestion enzymes, and placental layer. Some variation in individual fluorescence value histogram curve means and ranges were observed, but all positive curves under all conditions tested were normal and expressed fluorescence values greater than the IgG
control, thus confirming that the cells comprise a homogenous population which has positive expression of the markers.

Analysis of Postpartum Tissue-Derived Cells using Oligonucleotide Arrays Oligonucleotide arrays were used to compare gene expression profiles of umbilicus- and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.
Materials and Methods Isolation and Culture of Cells Postpartum tissue-derived cells. Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, PA) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 1. Cells were cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37 C with 5% CO2.
Fibroblasts. Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, MD; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines 'were cultured in DMEM/F12 medium (Invitrogen, Carlsbad, CA) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen). The cells were grown on standard tissue-treated plastic.
Human Mesenchyinal Stein Cells (hMSC). hMSCs were purchased from Cambrex Incorporated (Walkersville, MD; Lot numbers 2F1655, 2F1656 and 2F1657) and cultured according to the manufacturer's specifications in MSCGM Media (Cambrex). The cells were grown on standard tissue cultured plastic at 37 C with 5% CO2.
- 63 -Human Ileac Crest Bone Marrow Cells (ICBM). Human ileac crest bone marrow was received from NDRI with patient consent. The marrow was processed according to the method outlined by Ho, etal. (W003/025149). The marrow was mixed with lysis buffer (155 millimolar NH4C1, 10 millimolar KHCO3, and 0.1 millimolar EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500 x g. The supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 millimolar glutamine. The cells were centrifuged again and the cell pellet was resuspended in fresh medium. The viable mononuclear cells were counted using trypan-blue exclusion (Sigma, St. Louis, MO). The mononuclear cells were seeded in tissue-cultured plastic flasks at 5 x 104 cells/cm2. The cells were incubated at 37 C with 5% CO2 at either standard atmospheric 02 or at 5% 02. Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture.
The adherent cells were maintained in culture.
Isolation of tuRNA and GENECHIP Analysis. Actively growing cultures of cells were removed from the flasks with a cell scraper in cold phosphate buffered saline (PBS). The cells were centrifuged for 5 minutes at 300 x g. The supernatant was removed and the cells were resuspended in fresh PBS and centrifuged again. The supernatant was removed and the cell pellet was immediately frozen and stored at -80 C. Cellular niRNA was extracted and transcribed into cDNA. cDNA was then transcribed into cRNA and biotin-labeled.
The biotin-labeled cRNA was hybridized with Affymetrix GENECH1P HG-U133A oligonucleotide arrays (Affyrnetrix, Santa Clara CA). The hybridizations and data collection were performed according to the manufacturer's specifications. The hybridi7ation and data collection was performed according to the manufacturer's specifications. Data analyses were performed using "Significance Analysis of Microarrays" (SAM) version 1.21 computer software (Tusher, V.G. et al., 2001, Proc. Natl. Acad. Sci. USA 98: 5116-5121).
Results Fourteen different populations of cells were analyzed in this study. The cells along with passage information, culture substrate, and culture media are listed in Table 6-1. =
- 64 -Table 6-1. Cells analyzed by the microarray study. The cells lines are listed by their identification code along with passage at the time of analysis, cell growth substrate, and growth media.
Cell Population Passage Substrate Media Umbilical (022803) 2 Gelatin DMEM, 15% FBS,I3ME
Umbilical (042103) 3 Gelatin DMEM, 15% FBS, f3ME
Umbilical (071003) 4 Gelatin DMEM, 15% FBS, f3ME
Placenta (042203) 12 Gelatin DMEM, 15% FBS, f3ME
Placenta (042903) 4 Gelatin DMEM, 15% FBS,13ME
Placenta (071003) 3 Gelatin DMEM, 15% FBS, pmE
ICBM (070203) (5% 02) 3 Plastic MEM 10% FBS
ICBM (062703) (std 02) 5 Plastic MEM 10% FBS
ICBM (062703 )(5% 02) 5 Plastic MEM 10% FBS
hMSC (Lot 2F1655) 3 Plastic MSCGM
hMSC (Lot 2F1656) 3 Plastic MSCGM
hMSC (Lot 2F1657) 3 Plastic MSCGM
hFibroblast (9F0844) 9 Plastic DMEM-F12, 10% FBS
hFibroblast (CCD39SK) 4 Plastic DMEM-F12, 10% FBS
The data were evaluated by Principle Component Analysis. Analysis revealed 290 genes that were expressed in different relative amounts in the cells tested. This analysis provided relative comparisons between the populations.
Table 6-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs. The Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types. The Euclidean distance is inversely proportional to similarity between the expression of the 290 genes.
Table 6-2. The Euclidean Distances for the Cell Pairs.
The Euclidean distance was calculated for the cell types using the 290 genes that were expressed differently between the cell types.
Similarity between the cells is inversely proportional to the Euclidean distance.
Cell Pair Euclidean Distance ICBM-hMSC 24.71 Placenta-umbilical 25.52 ICBM-Fibroblast 36.44 ICBM-placenta 37.09 Fibroblast-MSC 39.63 ICBM-Umbilical 40.15 Fibroblast-Umbilical 41.59 MSC-Placenta 42.84 MSC-Umbilical 46.86 ICBM-placenta 48.41
- 65 -Tables 6-3, 6-4, and 6-5 show the expression of genes increased in placenta-derived cells (Table 6-3), increased in. umbilical cord-derived cells (Table 6-4), and reduced in umbilical cord and placenta-derived cells (Table 6-5).
Table 6-3. Genes which are specifically increased in expression in the placenta-derived cells as compared to the other cell lines assayed.
Genes Increased in Placenta-Derived Cells NCBI Accession Probe Set ID Gene Name Number C-type (calcium dependent, carbohydrate-recognition domain) 209732 at AF070642 lectin, superfamily member 2 (activation-induced) 206067_s_at Wilms tumor 1 NM_024426 207016_s_at aldehyde dehydrogenase 1 family, member A2 AB015228 206367_at Renin NM_000537 210004_at oxidized low density lipoprotein (lectin-like) receptor 1 214993_at Homo sapiens, clone IMAGE:4179671, mRNA, partial cds 202178_at protein kinase C, zeta NM 002744 209780_at hypothetical protein DKFZp564F013 AL136883 204135_at downregulated in ovarian cancer 1 NM 014890 Homo sapiens mRNA; cDNA DKFZp547K1113 (from clone 213542 at A1246730 DKFZp547K1113) Table 6-4. Genes which are specifically increased in expression in umbilical cord -derived cells as compared to the other cell lines assayed.
Genes Increased in Umbilicus-Derived Cells NCBI Accession Probe Set ID Gene Name Number 202859_x_at Interleukin 8 NM 000584 211506_s_at Interleukin 8 AF043337 210222_s_at reticulon 1 BC000314 chemokine (C-X-C motif) ligand 1 (melanoma growth 204470_at NM 001511 stimulating activity chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic 206336 at NM 002993 protein 2) 207850_at Chemokine (C-X-C motif) ligand 3 NM 002090 203485_at reticulon 1 NM_021136 202644_s_at tumor necrosis factor, alpha-induced protein 3 NM 006290
- 66 -Table 6-5. Genes which were decreased in expression in the umbilical cord and placenta cells as compared to the other cell lines assayed.
Genes Decreased in Umbilicus- and Placenta-Derived Cells Probe Set Gene name NCBI Accession ID Number _ 210135_s_at short stature homeobox 2 AF022654.1 205824_at heat shock 27kDa protein 2 N IVI
_001541.1 209687_at chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1) = U19495.1 203666_at chennokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1) N IV1 _000609.1 212670_at elastin (supravalvular aortic stenosis, Williams-Beuren syndrome) 213381Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone N91149 _at DKFZp586M2022) 206201_s_at mesenchyme homeobox 2 (growth arrest-specific homeobox) NI
hil _005924.1 205817_at Sine oculis homeobox homolog 1 (Drosophila) N M
_005982.1 209283_at crystallin, alpha B AF007162.1 212793_at dishevelled associated activator of nnorphogenesis 2 213488_at DKFZP586132420 protein AL050143.1 209763_at similar to neuralin 1 AL049176 205200_at Tetranectin (plasminogen binding protein) NI M_003278.1 205743_at src homology three (SH3) and cysteine rich domain NI
M_003149.1 200921_s_at B-cell translocation gene 1, anti-proliferative NI
M_001731.1 206932_at cholesterol 25-hydroxylase NI M_003956.1 204198_s_at runt-related transcription factor 3 AA541630 219747_at hypothetical protein FLJ23191 NI M_024574.1 204773_at Interleukin 11 receptor, alpha NJ M_004512.1 202465_at Procollagen C-endopeptidase enhancer NI M_002593.2 203706_s_at Frizzled homolog 7 (Drosophila) NI M_003507.1 212736_at hypothetical gene BC008967 BE299456 214587¨ at Collagen, type VIII, alpha 1 BE877796 201645_at Tenascin C (hexabrachion) NI M_002160.1 210239_at iroquois homeobox protein 5 U90304.1 203903_s_at Hephaestin NI M _014799.1 205816_at integrin, beta 8 NI M _002214.1 203069_at synaptic vesicle glycoprotein 2 NI M_014849.1
- 67 -213909_at Horno sapiens cDNA
FLJ12280 fis, clone MAMMA1001744 AU147799 206315_at cytokine receptor-like factor 1 NM
_004750.1 204401 at potassium intermediate/small conductance calcium-activated channel, NM 002250.1 ¨ subfamily N, member 4 ¨
216331_at integrin, alpha 7 AK022548.1 209663_s_at integrin, alpha 7 AF072132.1 213125_at DKFZP586L151 protein 202133_at transcriptional co-activator with PDZ-binding motif (TAZ) AA081084 206511_s_at Sine oculis homeobox homolog 2 (Drosophila) NM
_016932.1 213435_at KIAA1034 protein AB028957.1 206115_at early growth response 3 NM
_004430.1 213707_s_at distal-less homeobox 5 NM
_005221.3 218181_s_at hypothetical protein FLJ20373 NM_017792.1 aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid 209160 at AB018580.1 dehydrogenase, type II) 213905_x_at Biglycan 201261_x_at Biglycan BC002416.1 202132_at transcriptional co-activator with PDZ-binding motif (TAZ) AA081084 214701_s_at fibronectin 1 AJ276395.1 213791_at Proenkephalin NM
_006211.1 205422_s_at lntegrin, beta-like 1 (with EGF-like repeat domains) NM
_004791.1 Homo sapiens mRNA full length insert cDNA clone EUROIMAGE
214927 _at AL359052.1 206070_s_at EphA3 AF213459.1 212805_at KIAA0367 protein AB002365.1 natriu retie peptide receptor C/guanylate cyclase C (atrionatriuretic 219789 at A1628360 peptide receptor C) 219054_at hypothetical protein FLJ14054 NM _024563.1 Homo sapiens mRNA; cDNA DKFZp56413222 (from clone 213429 _at DKFZp564B222) 204929_s_at vesicle-associated membrane protein 5 (myobrevin) NM_ 006634.1 201843_s_at EGF-containing fibulin-like extracellular matrix protein 1 NM _004105.2 221478_at BCL2/adenovirus El B
19kDa interacting protein 3-like AL132665.1 201792_at AE binding protein 1 NM
_001129.2 204570_at cytochrome c oxidase subunit Vila polypeptide 1 (muscle) NM _001864.1 201621_at neuroblastoma, suppression of tumorigenicity 1 NM
_005380.1 202718_at Insulin-like growth factor binding protein 2, 36kDa NM_ 000597.1
- 68 -Tables 6-6, 6-7, and 6-8 show the expression of genes increased in human fibroblasts (Table 6-6), ICBM cells (Table 6-7), and MSCs (Table 6-8).
Table 6-6. Genes which were increased in expression in fibroblasts as compared to the other cell lines assayed.
Genes increased in fibroblasts dual specificity phosphatase 2 KIAA0527 protein Homo sapiens cDNA: FLJ23224 fis, clone ADSU02206 dynein, cytoplasmic, intermediate polypeptide 1 ankyrin 3, node of Ranvier (ankyrin G) inhibin, beta A (activin A, activin AB alpha polypeptide) ectonucleotide pyrophosphatase/phosphodiesterase 4 (putative function) KIAA1053 protein microtubule-associated protein lA
zinc finger protein 41 HSPC019 protein Homo sapiens cDNA: FLJ23564 fis, clone LNG10773 Homo sapiens mRNA; cDNA DKFZp564A072 (from clone DKFZp564A._072) LIM protein (similar to rat protein kinase C-binding enigma) inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase complex-associated protein hypothetical protein FLJ22004 Human (clone CTG-A4) mRNA sequence ESTs, Moderately similar to cytokine receptor-like factor 2; cytokine receptor CRL,2 precursor [Homo sapiens]
transforming growth factor, beta 2 hypothetical protein MGC29643 antigen identified by monoclonal antibody MRC OX-2 putative X-linked retinopathy protein Table 6-7. Genes which were increased in expression in the ICBM-derived cells as compared to the other cell lines assayed.
Genes Increased In ICBM Cells 'cardiac ankyrin repeat protein =MHC class I region ORF
=integrin, alpha 10 'hypothetical protein FI,J22362 =LTDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 3 (Ga1NAc-T3) -interferon-induced protein 44 =SRY (sex determining region Y)-box 9 (campomelic dysplasia, autosomal sex-reversal) "keratin associated protein 1-1 -hippocalcin-like 1 -jagged 1 (Alagille syndrome) =proteoglycan 1, secretory granule
- 69 -Table 6-8. Genes which were increased in expression in the MSC
cells as compared to the other cell lines assayed.
Genes Increased In MSC Cells =interleukin 26 -maltase-glucoamylase (alpha-glucosidase) 'nuclear receptor subfamily 4, group A, member 2 =v-fos FBJ murine osteosarcoma viral oncogene homolog -hypothetical protein DC42 'nuclear receptor subfamily 4, group A, member 2 =FBJ murine osteosarcoma viral oncogene homolog B
=WNT1 inducible signaling pathway protein 1 =MCF.2 cell line derived transforming sequence 'potassium channel, subfamily K, member 15 'cartilage paired-class homeoprotein 1 -Homo sapiens cDNA FLJ12232 fis, clone MAMMA1001206 -Homo sapiens cDNA FLJ34668 fis, clone LIVER2000775 =jun B proto-oncogene .B-cell CLL/Iymphoma 6 (zinc finger protein 51) 'zinc finger protein 36, C3H type, homolog (mouse) Summary. The present study was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas.
The study also included two different lines of dermal fibroblasts, three lines of mesenchyinal stem cells, and three lines of ileac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed on an GENECHIP oligonucleotide array that contained oligonucleotide probes for 22,000 genes.
The analysis revealed that transcripts for 290 genes were present in different amounts in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells. Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord.
The expression of selected genes has been confirmed by PCR (see Example 7).
These results demonstrate that the postpartum-derived cells have a distinct gene expression profile, for example, as compared to bone marrow-derived cells and fibroblasts.

Cell Markers in Postpartum-Derived Cells Summary
- 70 -To examine cells derived from the human placenta and the human umbilical cord, their gene expression profiles were compared to those of cells derived from other sources using the Affymetrix GENECHIP. Through this technique we identified 6 genes that were highly expressed in postpartum cells: oxidized LDL receptor 1, interleukin-8, renin, reticulon, chemokine receptor ligand 3 (CXC ligand 3) and granulocyte chemotactic protein 2 (G-CP-2).
Four of these genes (oxidized LDL receptor 1, renin, reticulon and IL-8) were differentially regulated at the mRNA level in postpartum cells. IL-8 was found to also be differentially regulated at the protein level.
We also investigated the expression of vimentin and alpha-smooth muscle actin., which has previously been associated with stroma-derived cells. Immediately after isolation (passage 0), cells derived from the human placenta stained positive for both alpha-smooth muscle actin and vimentin. This pattern was also observed in cells at passage 11. The result suggests that vimentin and alpha-smooth muscle actin expression in cells is preserved with passaging in the Growth Medium.
Cells derived from the human umbilical cord at passage 0 were probed for the expression of vimentin and alpha-smooth muscle actin proteins and were positive for alpha-smooth muscle actin and vimentin with the potential staining pattern preservation through passage 11.
The mRNA data at least partially verify the data obtained from the microarray experiments.
Introduction Similarities and differences in cells derived from the human placenta and the human umbilical cord were assessed by comparing their gene expression profiles with those o f cells derived from other sources (using an Affymetrix GENECHIP). Six "signature"
genes were identified: oxidized LDL receptor 1, interleukin-8, renin, reticulon, chemokine receptc.r ligand 3 (CXC ligand 3), and granulocyte chemotactic protein 2 (GCP-2). These "signature" genes were expressed at relatively high levels in postpartum-derived cells.
The present studies were conducted to verify the microarray data and detemiine correlations between gene and protein expression, as well as to establish a series of reliable assays for detection of unique identifiers for placenta- and umbilical cord-derived cells.
Methods & Materials Cells
- 71 -Placenta-derived cells (three isolates, including one isolate predominately neonatal as identified by karyotyping analysis), umbilical cord-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) grown in Growth Medium_ in gelatin-coated T75 flasks. Mesenchymal Stem Cells (MSCs) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, MD).
For IL-8 experiment, cells were thawed from liquid nitrogen and plated in gelatin-coated flasks at 5,000 cells/cm2, grown for 48 hours in Growth Medium and then grown for further 8 hours in 10 milliliters of serum starvation medium [DMEM ¨low glucose (Gibco, Carlsbad, CA), penicillin/streptomycin (Gibco, Carlsbad, CA) and 0.1% (w/v) Bovine Serum Albumin (BSA;
Sigma, St. Louis, MO)]. After this treatment RNA was extracted and the supernatants were centrifuged at 150 x g for 5 minutes to remove cellular debris. Supernatants were then frozen at -80 C for ELISA analysis.
Cell culture for ELISA assay.
Postpartum cells derived from placenta and umbilical cord, as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium in gelatin¨coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen. Cells were thawed and transferred to 15m1 centrifuge tubes. After centrifugation at 150 x g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 ml culture medium and counted. Cells were grown in a 75 cm2 flask containing 15 ml of Growth Medium at 375,000 cells/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation, medium was collected at the end of incubation, centrifuged at 14,000 x g for 5 minutes (and stored at -20 C).
To estimate the number of cells in each flask, 2 milliliters of tyrpsin/EDTA_ (Gibco, Carlsbad, CA) was added each flask. After cells detached from the flask, trypsin activity was neutralized with 8 milliliters of Growth Medium. Cells were transferred to a 15 milliliters centrifuge tube and centrifuged at 150 x g for 5 minutes. Supernatant was removed and 1 milliliter Growth Medium was added to each tube to resuspend the cells. Cell number was estimated using a hemocytometer.
ELISA assay The amount of IL-8 secreted by the cells into serum starvation medium was analyzed using ELISA assays (R&D Systems, Minneapolis, MN). All assays were tested according to the instructions provided by the manufacture.
- 72 -Total RNA isolation RNA was extracted from confluent postpartum-derived cells and fibroblasts or for IL-8 expression from cells treated as described above. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, MO) according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, CA). RNA was extracted according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, CA) and subjected to DNase treatment (2.7 U/sample) (Sigma St. Louis, MO). RNA was eluted with 50 microliters DEPC-treated water and stored at -80 C.
Reverse transcription RNA was also extracted from human placenta and umbilical cord. Tissue (30 mg) was suspended in 700 microliters of buffer RLT containing beta-mercaptoethanol.
Samples were mechanically homogenized and the RNA extraction proceeded according to manufacturer's specification. RNA was extracted with 50 microliters of DEPC-trcatcd water and stored at -80 C. RNA was reversed transcribed using random hexamers with the TaqMaii reverse transcription reagents (Applied Biosystems, Foster City, CA) at 25 C for 10 minutes, 37 C for 60 minutes, and 95 C for 10 minutes. Samples were stored at -20 C.
= .
Genes identified by cDNA microarray as uniquely regulated in postpartum cells (signature genes ¨ including oxidized LDL receptor, interleukin-8, renin and reticulon), were further investigated using real-time and conventional PCR.
Real-time PCR
PCR was performed on cDNA samples using AssaysonDemandTM gene expression products: oxidized LDL receptor (Hs00234028); renin (Hs00166915); reticulon (Hs00382515);
CXC ligand 3 (Hs00171061); GCP-2 (Hs00605742); IL-8 (Hs00174103); and GAPDH
(Applied Biosystems, Foster City, CA) were mixed with cDNA and TaqMaiTuniversal PCR
master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, CA) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems, Foster City, CA). Thermal cycle conditions were initially 50 C for 2 minutes and 95 C
for 10 min, followed by 40 cycles of 95 C for 15 seconds and 60 C for 1 minute. PCR data were analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI
Prism 7700 Sequence Detection System).
- 73 -Conventional PCR
Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Massachusetts, USA) to confirm the results from real-time PCR. PCR was performed using 2 microliters of cDNA solution, lx AmpliTa4mGold universal mix PCR reaction buffer (Applied Biosystems, Foster City, CA) and initial denaturation at 94 C
for 5 minutes.
Amplification was optimized for each primer set. For IL-8, CXC ligand 3, and reticulon (94 C
for 15 seconds, 55 C for 15 seconds and 72 C for 30 seconds for 30 cycles);
for renin (94 C for 15 seconds, 53 C for 15 seconds and 72 C for 30 seconds for 38 cycles); for oxidized LDL
receptor and GAPDH (94 C for 15 seconds, 55 C for 15 seconds and 72 C for 30 seconds for 33 cycles). Primers used for amplification are listed in Table 7-1. Primer concentration in the final PCR reaction was 1 p.M except for GAPDH which was 0.5 M. GAPDH primers were the same as real-time PCR, except that the manufacturer's TaqMagprobe was not added to the final PCR
reaction. Samples were run on 2% (w/v) agarose gel and stained with ethidium bromide (Sigma, St. Louis, MO). Images were captured using a 667 Universal Twinpack film (VWR
International, South Plainfield, NJ) using a focal-length PolaroidTM camera (VWR
International, South Plainfield, NJ).
Table 7-1: Primers used Primer name Primers Oxidized LDL receptor S: 5'- GAGAAATCCAAAGAGCAAATGG-3 (SEQ ID NO:1)' A: 5'-AGAATGGAAAACTGGAATAGG -3' (SEQ ID NO:2) Renin S: 5'-TCTTCGATGCTTCGGATTCC -3' (SEQ ID NO:3) A: 5'-GAATTCTCGGAATCTCTGTTG -3' (SEQ ID NO:4) Reticulon S: 5'- TTACAAGCAGTGCAGAAAACC-3' (SEQ ID NO:5) A: 5'- AGTAAACATTGAAACCACAGCC-3' (SEQ ID NO:6) Interleukin-8 S: 5'- TCTGCAGCTCTGTGTGAAGG-3' (SEQ ID NO:7) A: 5'-eITCAAAAACTTCTCCACAACC- 3' (SEQ ID NO:8) Chemokine (CXC) ligand 3 S: 5'- CCCACGCCACGCTCTCC-3' (SEQ ID NO:9) A: 5'-TCCTGTCAGTTGGTGCTCC -3' (SEQ ID NO:10) Inzniunofluorescence Postpartum cells were fixed with cold 4% (w/v) paraformaldehyde (Sigma-Aldrich, St.
Louis, MO) for 10 minutes at room temperature. One isolate each of umbilical-and placental-derived cells at passage 0 (PO) (directly after isolation) and passage 11 (P11) (two isolates of Placenta-derived, two isolates of Umbilical cord-derived cells) and fibroblasts (P11) were used.
Inununocytochemistry was performed using antibodies directed against the following epitopes:
vimentin (1:500, Sigma, St. Louis, MO), desmin (1:150; Sigma - raised against rabbit; or 1:300;
- 74 -Chemicon, Temecula, CA - raised against mouse,), alpha-smooth muscle actin (SMA; 1:400;
Sigma), cytokeratin 18 (CK18; 1:400; Sigma), von Willebrand Factor (vWF;
1:200; Sigma), and CD34 (human CD34 Class HI; 1:100; DAKOCytomation, Carpinteria, CA). In addition, the following markers were tested on passage 11 postpartum cells: anti¨human GROalpha - PE
(1:100; Becton Dickinson, Franklin Lakes, NJ), anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA), anti-human oxidized LDL receptor 1 (ox-LDL RI; 1:100; Santa Cruz Biotech), and anti-human NOGA-A (1:100; Santa Cruz, Biotech).
Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3%
(v/v) TritoiT(TritonTmX-100; Sigma, St. Louis, MO) for 30 minutes to access intracellular antigens. Where the epitope of interest was located on the cell surface (CD34, ox-LDL R1), TM
Triton X-100 was omitted in all steps of the procedure in order to prevent epitope loss.
Furthennore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL
RI, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout.
Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. The primary antibody solutions were removed and the cultures were washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, OR) and/or goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG-FITC (1:150, Santa Cruz Biotech). Cultures were then washed and 10 jaM DAPI
(Molecular Probes) applied for 10 minutes to visualize cell nuclei.
Following immunostaining, fluorescence was visualized using an appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY).
In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution (no 1* control). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, CA). For triple-stained = samples, each image was taken using only one emission filter at a time.
Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, CA).
Preparation of cells for FAGS analysis Adherent ce14 in flasks were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, CA) and detached with Trypsin/EDTA (Gibco, Carlsbad, CA). Cells were harvested, centrifuged, and re-suspended 3% (v/v) FBS in PBS at a cell concentration of lx1 07 per
- 75 -_ milliliter. One hundred microliter aliquots were delivered to conical tubes.
Cells stained for intracellular antigens were permeablized with Perm/Wash buffer (BD Pharmingen, San Diego, CA). Antibody was added to aliquots as per manufactures specifications and the cells were incubated for in the dark for 30 minutes at 4 C. After incubation, cells were washed with PBS
and centrifuged to remove excess antibody. Cells requiring a secondary antibody were resuspended in 100 microliters of 3% FBS. Secondary antibody was added as per manufactures specification and the cells were incubated in the dark for 30 minutes at 4 C.
After incubation, cells were washed with PBS and centrifuged to remove excess secondary antibody. Washed cells were resuspended in 0.5 milliliters PBS and analyzed by flow cylometry.
The following antibodies were used: oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROa (555042;
BD Phanningen, Bedford, MA), Mouse IgG1 kappa, (P-4685 and M-5284; Sigma), Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.).
FAGS analysis Flow cytometry analysis was performed with FACScalibur (B ecton Dickinson San Jose, CA).
Results Results of real-time PCR for selected "signature" genes perfoimed on cDNA from cells derived from human placentas, adult and neonatal fibroblasts and Mesenchymal Stem Cells (MSCs) indicate that both oxidized LDL receptor and renin were expressed at higher level in the placenta-derived cells as compared to other cells. The data obtained from real-time PCR were analyzed by the AACT method and expressed on a logarithmic scale. Levels of reticulon and oxidized LDL receptor expression were higher in umbilical cord-derived cells as compared to other cells. No significant difference in the expression levels of CXC ligand 3 and GCP-2 were found between postpartum cells and controls. The results of real-time PCR were confirmed by conventional PCR. Sequencing of PCR products further validated these observations. No significant difference in the expression level of CXC ligand 3 was found between postpartum cells and controls using conventional PCR CXC ligand 3 primers listed in Table 7-1.
The expression of the cytokine, IL-8 in postpartum cells is elevated in both Growth Medium-cultured and serum-starved postpartum-derived cells. All real-time PCR
data were validated with conventional PCR and by sequencing PCR products.
- 76 -When supernatants of cells grown in serum-free medium were examined for the presence of IL-8, the highest amounts were detected in media derived from umbilical cells and some isolates of placenta cells (Table 7-2). No IL-8 was detected in medium derived from human deinial fibroblasts.
Table 7-2: IL-8 protein expression measured by ELISA
Cell type IL-8 hFibro ND
Placenta Isolate 1 ND
UMBC Isolate 1 2058.42+144.67 Placenta Isolate 2 ND
UMBC Isolate 2 2368.86+22.73 Placenta Isolate3 (normal 02) 17.27+8.63 Placenta Isolate 3 (low02, W/O 264.92+9.88 BME) Results of the ELISA assay for interleukin-8 (11-8) performed on placenta-and umbilical cord-derived cells as well as human skin fibr-oblasts. Values are presented here are pg/million cells, n=2, sem.
ND: Not Detected Placenta-derived cells were also examined for the expression of oxidized LDL
receptor, GCP-2 and GROalpha by FACS analysis. Cells tested positive for GCP-2. Oxidized LDL
receptor and GRO were not detected by this method.
Placental cells were also tested for the expression of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells derived from the human placenta were fixed with 4% parafonnaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Cells stained positive for both alpha-smooth muscle actin and vimentin. This pattern was preserved through passage 11. Only a few cells (<5%) at passage 0 stained positive for cytokeratin 18.
Cells derived from the human umbilical cord at passage 0 -were probed for the expression of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells -were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin.
Umbilical cord-derived cells were positive for alpha-smooth muscle actin and vimentin, with the staining pattern consistent through passage 11.
- 77 -Placenta-derived and umbilical cord-derived cells at passage 1 1 were also investigated by immunocytochernistry for the expression of GROalpha, GCP-2, oxidized LDL
receptor 1 and reticulon. Complete results of that experiment are still pending.
Discussion and Conclusion Thus far concordance between gene expression levels measured by microan-ay and PCR
(both real-time and conventional) has been established for four genes:
oxidized LDL receptor 1, renin, reticulon, and IL-8. The expression of these genes was differentially regulated at the mRNA level in postpartum cells, with IL-8 also differentially regulated at the protein level. The presence of oxidized LDL receptor was not detected at the protein level by FACS analysis in cells derived from the placenta. Differential expression of GCP-2 and CXC
ligand 3 was not confirmed at the mRNA level, however GCP-2 was detected at the protein level by FACS
analysis in the placenta-derived cells. Differences between these data and that obtained from the microarray experiment may be due to differences in the sensitivity of the methodologies.
Immediately after isolation (passage 0), cells derived from the human placenta stained positive for both alpha-smooth muscle actin and vimentin. This pattern was also observed in cells at passage 11. These results suggest that vimentin and alpha-smooth muscle actin expression may be preserved in cells with passaging, at least in the Growth Medium used here.
Cells derived from the human umbilical cord at passage 0 were probed for the expression of alpha-smooth muscle actin and vimentin, and were positive for both. The staining pattern was preserved through passage 11.
In conclusion, the complete mRNA data at least partially verify the data obtained from the microan-ay experiments. As additional protein experiments are complete, the relationships between mRNA and protein expression will be more comprehensively understood.

Immunohistochemical Characterization of PPDC Phenotype The phenotypes of cells found within human postpartum tissues, namely umbilical cord and placenta, were analyzed by immunohistochemistry.
Materials & Methods
- 78 -Tissue Preparation. Human umbilical cord and placenta tissue was harvested and immersion fixed in 4% (w/v) parafon-naldehyde overnight at 4 C.
annunohistochemistry was performed using antibodies directed against the following epitopes (see Table 8-1): vimentin (1:500; Sigma, St. Louis, MO), desmin (1:150, raised against rabbit; Sigma; or 1:300, raised against mouse; Chemicon, Temecula, CA), alpha-smooth muscle actin (SMA; 1:400;
Sigma), cytokeratin 18 (CK18; 1:400; Sigma), von Willebrand Factor (vWF; 1:200;
Sigma), and CD34 (human CD34 Class III; 1:100; DAKOCytomation, Carpinteria, CA) _ In addition, the following markers were tested: anti-human GROalpha - PE (1:100; Becton Dickinson, Franklin Lakes, NJ), anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA), anti-human oxidized LDL
receptor 1 (ox-LDL R1; 1:100; Santa Cruz Biotech), and anti-human NOGO-A
(1:100; Santa Cruz Biotech). Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, CA) on a dry ice bath containing ethanol.
Frozen blocks were then sectioned (10 !um thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
Table 8-1: Summary of Primary Antibodies Used Antibody Concentration Vendor Vimentin 1:500 Sigma, St. Louis, MO
Desmin (rb) 1:150 Sigma Desmin (m) 1:300 Chemicon, Temecula, CA
alpha smooth muscle actin 1:400 Sigma (SMA) Cytokeratin 18 (CK18) 1:400 Sigma von Willebrand factor 1:200 Sigma (vWF) CD34 III 1:100 DakoCytomation, Carpinteria, CA
GROalpha - PE 1:100 BD, Franklin Lakes, NJ
GCP-2 1:100 Santa Cruz Biotech Ox-LDL R1 1:100 Santa Cruz Biotech NOGO-A 1:100 Santa Cruz Biotech Immunohistochemistry. Immunohistochemistry was performed similar to previous studies (e.g., Messina, et al. (2003) Exper. Neural. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing
- 79 -
80 PCT/US2004/020957 PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100;
Sigma) for 1 hour to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (CD34, ox-LDL R1), Triton was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL R1, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout the procedure. Primary antibodies, diluted in blocking solution, were then applied to the sections for a period of 4 hours at room temperature.
Primary antibody solutions were removed, and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG-Texas Red (1:250; Molecular Probes, Eugene, OR) and/or goat anti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG-FITC (1:150; Santa Cruz Biotech). Cultures were washed, and 10 tiM DAPI (Molecular Probes) was applied for 10 minutes to visualize cell nuclei.
Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY).
Positive staining was represented by fluorescence signal above control staining. Representative images were captured using a digital color video camera and hnagePro software (Media Cybernetics, Carlsbad, CA). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, CA).
Results Umbilical Cord Characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord.
In particular, vWF
and CD34 expression were restricted to blood vessels contained within the cord. CD34+ cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery &
vein, but not contained with the vessels themselves. CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.
Placenta Characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 were all observed within the placenta and regionally specific.
GROalpha, GCP-2, ox-LDL R1, and NOGO-A Tissue Expression. None of these markers were observed within umbilical cord or placental tissue.

Summary. Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD34 are expressed in cells within human umbilical cord and placenta.
Example 9 Secretion of Trophic Factors by Postpartum Cells Derived From Placenta and Umbilical Cord.
The secretion of selected trophic factors from placenta- and umbilical cord-derived PPDCs was measured. Factors were selected that have angiogenic activity (i.e., hepatocyte growth factor (HGF) (Rosen et al. (1997) Ciba Found. Symp. 212:215-26), monocyte chemotactic protein 1 (MCP-1) (Salcedo et al. (2000) Blood 96;34-40), interleukin-8 (IL-8) ( Li et al. (2003) J. Immunol. 170:3369-76), keratinocyte growth factor (KGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) (Hughes et al.
(2004) Ann.
Thorac. Surg. 77:812-8), matrix metalloproteinase 1 (TIMP1), angiopoietin 2 (ANG2), platelet derived growth factor (PDGF-bb), thrombopoietin (TPO), heparin-binding epidermal growth factor (HB-EGF), stromal-derived factor lalpha (SDF-lalpha)), neurotrophic/neuroprotective activity (brain-derived neurotrophic factor (BDNF) (Cheng et al. (2003) Dev.
Biol. 258;319-33), interleukin-6 (IL-6), granulocyte chemotactic protein-2 (GCP-2), transforming growth factor beta2 (TGFbeta2)), or chemokine activity (macrophage inflammatory protein lalpha (MIP1a), macrophage inflammatory protein lbeta (MIP lb), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), 1309, thymus and activation-regulated chemokine (TARC), Eotaxin, macrophage-derived chemokine (MDC), IL-8).
Methods & Materials Cell culture. PPDCs derived from placenta and umbilical cord as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium on gelatin-coated T75 flasks. Cells were cryopreserved at passage 11 and stored in liquid nitrogen. After thawing of the cells, Growth Medium was added to the cells followed by transfer to a 15 milliliters centrifuge tube and centrifugation of the cells at 150 x g for 5 minutes. The supernatant was discarded. The cell pellet was resuspended in 4 milliliters Growth Medium, and cells were counted. Cells were seeded at 375,000 cells/75 cm2 flask containing 15 milliliters of Growth Medium and cultured for 24 hours. The medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin/streptomycin (Gibco)) for 8 hours. Conditioned serum-free media was collected at the
- 81 -2A71 g I I-end of incubation by centrifugation at 14,000 x g for 5 minutes and stored at -20 C. To estimate the number of cells in each flask, cells were washed with phosphate-buffered saline (PBS) and detached using 2 milliliters trypsin/EDTA (Gibco). Trypsin activity was inhibited by addition of 8 milliliters Growth Medium. Cells were centrifuged at 150 x g for 5 minutes.
Supernatant was removed, and cells were resuspended in 1 milliliter Growth Medium. Cell number was estimated using a hemocytometer.
ELISA assay. Cells were grown at 37 C in 5% carbon dioxide and atmospheric oxygen.
Placenta-derived PPDCs (101503) also were grown in 5% oxygen or beta-mercaptoethanol (BME). The amount of MCP-1, IL-6, VEGF, SDF-I alpha , GCP-2 , 1L-8, and TGF-beta2 produced by each cell sample was measured by an ELISA assay (R&D Systems, Minneapolis, MN). All assays were performed according to the manufacturer's instructions.
Values presented are pg/ml/million cells (n=2, sem).
SEARCHLIGHT Multiplexed ELISA assay. Chemokines (MIPI a, MIP1b, MCP-1, Rantes, 1309, TARC, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, TIMPI , ANG2, PDGF-bb, TPO, HB-EGF were measured using SEARCHLIGHT Proteome Arrays (Pierce Biotechnology Inc.). The Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to 16 proteins per well. The arrays are produced by spotting a 2 x 2, 3 x 3, or 4 x 4 pattern of four to 16 different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture chemiluminescent signal generated at each spot within each well of the plate The amount of signal generated in each spot is proportional to the amount of target protein in the standard or sample.
Results ELISA assay. MCP-1 and 1L-6 were secreted by placenta- and umbilical cord-derived .PPDCs and dermal fibroblasts (Table 9-1). SDF-lalpha was secreted by placenta-derived PPDCs cultured in 5% 02 and by fibroblasts. (3CP-2 and IL-8 were secreted by umbilical-derived PPDCs and by placenta-derived PPDCs cultured in the presence of BME or 5% 02.
GCP-2 also was secreted by human fibroblasts. TGF-beta2 was not detectable by ELISA assay.
Table 9-1. ELISA assay results MCP-1 VEGF SDF-1 a GCP-2 IL-8 TGF-Fibroblast 17+1 61+3 29+2 19+1 21+1 ND ND
Placenta (042303) 60T3 - 41+2 Ni)_ ¨
____________________________________________________ ND ND ND
Umbilical (022803) 1150+74 4234+289 ND ND ¨
160+11 2058+145 ND
¨Placenta (071003) 125+16 10+1 ND ND ND NI) ND
Umbilical (071003) 2794+84 1356+43 _ ND _ ND
2184+98 2369+23 ND
-82-_ CO A71 r s. !OM' I I
= Placenta (101503) BME 21+10 67+3 ND ND 44+9 -17+9 ND
Placenta (101503) 5% 02, W/O 77+16 339+21 ND 1149+137 54+2 265+10 ND
BME
Key: ND: Not Detected.
SEARCHLIGHT Multiplexed ELISA assay. TIMI'l, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1b, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were secreted from umbilical cord-derived PPDCs (Tables 9-2 and 9-3). TIIV1131, TPO, KGF, HGF, FIBEGF, BDNF, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were secreted from placenta-derived PPDCs (Tables 9-2 and 9-3). No Ang2, VEGF, or PDGF-bb were detected.
Table 9-2. SEARCHLIGHT Multiplexed FI ISA assay results TIMP1 ANG2 PDGFbb "FPO KGF HGF FGF VEGF HBEGF BDNF
hFB 19306.3 ND ND 230.5 5.0 ND - ND 27.9 -1.3 ND
PI 24299.5 ND ND 546.6 8.8 16.4 ND ND
3.81.3 ND
Ul 57718.4 ND ND 1240.0 5.8 559.3 148.7 ND 93 165.7 P3 14176.8 ND ND 568.7 5.2 10.2 ND ND
1.9 33.6 U3 21850.0 ND ND 1134.5 9.0 195.6 30.8 ND
5.4 388.6 Key: hFB (human fibroblasts), PI (placenta-derived PPDC (042303)), Ul (umbilical cord-derived PPDC (022803)), P3 (placenta-derived PPDC (071003)), 1.13 (umbilical cord-derived PPDC
(071003)). ND: Not Detected.
Table 9-3. SEARCHLIGHT' Multiplexed ELISA assay results MIP la WPM MCP1 RANTES 1309 TARC Eotaxin MDC 1L8 _ h _____________________________________ _ hFB ND ND 39.6 ND ND 0.1 ND ND 204.9 P1 79.5 ND 228.4 4.1 ND 3.8 12.2 ND
413.5 Ul ND 8.0 1694.2 ND
22.4 37.6 ND 18.9 51930.1 P3 ND ND 102.7 ND ND 0.4 ND ND 63.8 U3 ND 5.2 2018.7 41.5 11.6 21.4 ND 4.8 10515.9 Key: hFB (human fibroblasts), P1 (placenta-derived PPDC (042303)), UI
(umbilical cord-derived PPDC (022803)), P3 (placenta-derived PPDC (071003)), U3 (umbilical cord-derived PPDC
(071003)). ND: Not Detected.
Summary. Umbilical cord- and placenta-derived cells secreted a number of trophic factors. Some of these trophic factors, such as HGF, bFGF, MCP-1 and IL-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL-6, have important roles in neural regeneration.

In vitro Immunology Postpartum cell lines were evaluated in vitro for their immunological characteristics in an effort to predict. the immunological response, if any, these cells would elicit upon in vivo
- 83 -transplantation. Postpartum cell lines were assayed by flow cytometry for the expression of HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2. These proteins are expressed by antigen-presenting cells (APC) and are required for the direct stimulation of naive CD4+ T cells (Abbas & Lichinian, CELLULAR AND MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171). The cell lines were also analyzed by flow cytometry for the expression of HLA-G (Abbas & Lichtman, CELLULAR AND MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171), CD178 (Coumans, et.al., (1999) Journal of Immunological Methods 224, 185-196), and PD-L2 (Al?bas & Lichinian, CELLULAR AND MOLECULAR
IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171; Brown, et. al.
(2003) The Journal of Immunology 170, 1257-1266). The expression of these proteins by cells residing in placental tissues is thought to mediate the immuno-privileged status of placental tissues in utero. To predict the extent to which postpartum placenta- and umbilical cord-derived cell lines elicit an immune response in vivo, the cell lines were tested in a one-way mixed lymphocyte reaction (MLR).
Materials and Methods Cell Culture. Cells were cultured in Growth Media until confluent in T75 flasks (Corning, Corning, NY) coated with 2% gelatin (Sigma, St. Louis, MO).
Antibody Staining. Cells were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, CA) and detached with Trypsin/EDTA (Gibco, Carlsbad, MO). Cells were harvested, centrifuged, and re-suspended in 3% (v/v) FBS in PBS at a cell concentration of lx i07 per milliliter. AntibodY (Table 10-1) was added to one hundred microliters of cell suspension as per manufacturer's specifications and incubated in the dark for 30 minutes at 4 C.
After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were re-suspended in five hundred microliters of PBS and analyzed by flow cytometry using a FACSCalibur instrument (Becton Dickinson, San Jose, CA).
Table 10-1. Antibodies Antibody Manufacture Catalog Number HLA-DRDPDQ BD Pharmingen (San Diego, CA) 555558 CD80 BD Pharmingen (San Diego, CA) 557227 CD86 BD Pharmingen (San Diego, CA) 555665 B7-H2 BD Pharmingen (San Diego, CA) 552502 HLA-G Abcam (Cambridgeshire, UK) ab 7904-100 CD178 Santa Cruz (San Cruz, CA) sc-19681 PD-L2 BD Pharmingen (San Diego, CA) 557846
- 84-Mouse IgG2a Sigma (St. Louis, MO) F-6522 Mouse IgGlkappa Sigma (St. Louis, MO) P-4685 Mixed Lymphocyte Reaction. Cryopreserved vials of passage 10 umbilical cord-derived PPDCs labeled as cell line A and passage 11 placenta-derived PPDCs labeled as cell line B were sent on dry ice to CTBR (Senneville, Quebec) to conduct a mixed lymphocyte reaction using CTBR SOP no. CAC-031. Peripheral blood mononuclear cells (PBMCs) were collected from multiple male and female volunteer donors. Stimulator (donor) allogeneic PBMC, autologous PBMC, and postpartum cell lines were treated with mitomycin C.
Autologous and mitomycin C-treated stimulator cells were added to responder (recipient) PBMCs and cultured for 4 days. After incubation, [31-1]thymidine was added to each sample and cultured for 18 hours.
Following harvest of the cells, radiolabeled DNA was extracted, and [31-1]-thymidine incorporation was measured using a scintillation counter.
The stimulation index for the allogeneic donor (SIAD) was calculated as the mean proliferation of the receiver plus mitomycin C-treated allogeneic donor divided by the baseline proliferation of the receiver. The stimulation index of the postpartum cell was calculated as the mean proliferation of the receiver plus mitomycin C-treated postpartum cell line divided by the baseline proliferation of the receiver.
Results Mixed Lymphocyte Reaction-Placenta. Seven human volunteer blood donors were screened to identify a single allogeneic donor that would exhibit a robust proliferation response in a mixed lymphocyte reaction with the other six blood donors. This donor was selected as the allogeneic positive control donor. The remaining six blood donors were selected as recipients.
The allogeneic positive control donor and placenta cell lines were treated with mitomycin C and cultured in a mixed lymphocyte reaction with the six individual allogeneic receivers. Reactions were performed in triplicate using two cell culture plates with three receivers per plate (Table 10-2). The average stimulation index ranged from 1.3 (plate 2) to 3 (plate 1) and the allogeneic donor positive controls ranged from 46.25 (plate 2) to 279 (plate 1) (Table 10-3).
Table 10-2. Mixed Lymphocyte Reaction Data - Cell Line B (Placenta)
- 85 -____________________________________________________ _ LYPIV1f6Fp-Ritiferalion Assay n , , PlatelD: Plate1 1 i _____________________________________ 1 . __ . . 1 Analytical Culture Replicates number -System -I 2 -8 ' Mn SD- .C.V
Roliferation baseline of receiver 79 119 138 112.0 30.12 26.9 1M03-7769 Control of autostimulation (Mtomycin C
treated autologous cells) 241 i 272 175 229.3 49:54- --2176 MLR allogenic donor IM33-7768 (Mtomycin C treated) 2.8071-5 22852- 20021 22414.7 1525.67 6.8 MLR w ith cell line (Mtomycin C treated cell type B) 664 r--66-0- 160-0---77T-b--- 281.2T- 3676-SI (donor) F.. 1 ij 200 --ST-(6-61111h6) 7 ______________ -r-Proliferation baseline of receiver 206 134 262 200.7 64.17 32.0 IM03-7770 Control of autostimulation (Mtomycin C
treated autologous cells) 109T - 602 624 -739-3Y -367:38 -4T:6 MLR allogenic donor IM33-7768 (Mtomycin C treated) -46605 I 43729 MLR w ith cell line (Mtomycin C treated cell type B) 533 2582 2376 1830.3 1128.24 61.6 SI (donor) 1 221 - --s-rra-ii line) 0-Proliferation baseline of receiver 15/ : 87 1528 124.0 35.17 28.4 Control of autostimulation (rvitomycin C treated autologous cells) 208 i 188 L13 3f 8. Ci -186781 56.4 MLR allogenic donor IN/33-7768 (Mtomycin C treated) 24497-r-34348 31388 30077.7 5054:53 16.8 MLR w ith cell line (Mtomycin C treated cell type B) -89.1 I 643.
a 622.6 20:70 -4.8 SI (donor) ji 243 _________ SI(be)ltine) I l.....____ i 5 Proliferation baseline of receiver 56 I 98 51 68.3 25.81 37.8 im 03-7772 Control of autostimulation (Mtomycin C
treated autologous cells) -133- - 120 21.3 -165:3 -5086 3-2:4 MLR allogenic donor IM03-7768 (Mtomycin C tre.ated) -14222- ' 20076- "22168 18822:0 ' 4118.75 -- -21:9- -MLR w ith cell line (Mtomycin C treated cell type B) a a a a a a SI (donor) 275 --81 (cell-litfe) 1 a __ IM 03-7768 Proliferation baseline of receiver 84 . 242 208 178.0 83.16 46.7 (allogenic donor) Control of autostimulation (Mtomycin treated autologous cells) 361 1 617 --304 42773- --186:71- ---86:0--I .
Proliferation baseline of receiver 126 , 124 143 131.0 10.44 8.0 Cell line type B __ __ --comtrol of autostimulation (Mtomycin treated autologous cells) 622 . 1676-487- - -704:7- -29-4-.65- -377f-t Plate ID: Plate 2 Analytical Culture Replicates number System 1 2 3 Mean SD CV
Proliferation baseline of receiver 908 181 330 473.0 384.02 81.2 IM03-7773 Control of autostimulation (Mitomycin C
treated autologous cells) 269 405 572 415.3 151.76 36.5 MLR allogenlc donor IM03-7768 (Mitomycin C treated) 29151 28691 28315 28719.0 418.70 1.5 MLR with cell line (Mitomycin C treated cell type B) 567 732 905 734.7 169.02 23.0 SI (donor) 61 SI (cell line) 2 Proliferation baseline of receiver 893 1376 185 818.0 599.03 73.2 IM03-7774 Control of autostimulation (Mitomycin C
treated autologous cells) 261 381 568 403.3 154.71 38.4 MLR allogenic donor IM03-7768 (Mitomycin C treated) 53101 42839 48283 48074.3 5134.18 10.7 MLR with cell line (Mitomycin C treated cell type B) 515 789 294 532.7 247.97 46.6 SI (donor) 59 SI (cell line) 1 Proliferation baseline of receiver 1272 300 544 /0 .3 505.69 71.7 IM03-7775 Control of autostImulation (Mitomycin C
treated autologous cells) 232 199 484 305.0 155.89 51.1 MLR allogenic donor IM03-7768 (Mitomycin C treated) 23554 10523 28965 21014.0 9479.74 45.1 MLR with cell line (Mitomycin C treated cell type 13) 768 924 563 751.7 181.05 24.1 SI (donor) 30 SI (cell line) 1 Proliferation baseline of receiver 1530 13/ 1046 904.3 /0/.22 78.2 I MO3-7776 Control of autostimulation (Mitomycin C
treated autologous cells) 420 218 394 344.0 109.89 31.9 MLR allogenic donor IM03-7768 (Mitomycin C treated) 28893 32493 34746 32044.0 2952.22 9.2 MLR with cell line (Mitomycin C treated cell type B) a a a a a a SI (donor) 35 SI (cell line) a
- 86 -Table 10-3. Average stimulation index of placenta cells and an allogeneic donor in a mixed lymphocyte reaction with six individual allogeneic receivers.
Average Stimulation Index Recipient Placenta Plate 1 (receivers 1-3) 279 Plate 2 (receivers 4-6) 46.25 1.3 Mixed Lymphocyte Reaction- Umbilicus. Six human volunteer blood donors were screened to identify a single allogeneic donor that will exhibit a robust proliferation response in a mixed lymphocyte reaction with the other five blood donors. This donor was selected as the allogeneic positive control donor. The remaining five blood donors were selected as recipients.
The allogeneic positive control donor and placenta cell lines were mitomycin C-treated and cultured in a mixed lymphocyte reaction with the five individual allogeneic receivers. Reactions were performed in triplicate using two cell culture plates with three receivers per plate (Table 10-4). The average stimulation index ranged from 6.5 @late 1) to 9 @late 2) and the allogeneic donor positive controls ranged from 42.75 (plate 1) to 70 (plate 2) (Table 10-5).
Table 10-4. Mixed Lymphocyte Reaction Data- Cell Line A (Umbilicus) DPM for Proliferation Assay Plate ID: Plate1 Analytical Culture Replicates number System 1 2 3 Mean SD
CV
Proliferation baseline of receiver 1074 406 391 623.7 390.07 62.5 IM04-2478 Control of autostimulation (Mitomycin C treated autologous cells) 672 510 1402 861.3 475.19 55.2 MLR allogenlc donor 1M04-2477 (Mitomycin C treated) 43777 48391 38231 43466.3 5087.12 11.7 MLR with cell line (Mitomycin C treated cell type A) 2914 5622 6109 4881.7 1721.36 35.3 SI (donor) 70 SI (cell line) Proliferation baseline of receiver 530 508 52/ 521./
11.93 2.3 IM04-2479 Control of autostimulatIon (Mitomycin C treated autologous cells) 701 567 1111 793.0 283.43 35.7 MLR allogenic donor 1M04-2477 (Mitomycin C treated) 25593 24732 22707 24344.0 1481.61 6.1 MLR with cell line (Mitomycin C treated cell type A) 5086 3932 1497 3505.0 1832.21 52.3 SI (donor) 47 SI (cell line) 7 Proliferation baseline of receiver 1192 854 1330 1125.3 244.90 21.8 IM04-2480 Control of autostimulation (Mitomycln C treated autologous cells) 2963 993 2197 2051.0 993.08 48.4 MLR allogenic donor 1M04-2477 (Mitomycin C treated) 25416 29721 23757 26298.0 3078.27 11.7 MLR with cell line (Mitomycin C treated cell type A) 2596 5076 3426 3699.3 1262.39 34.1 SI (donor) 23 SI (cell line) 3 Proliferation baseline of receiver 695 451 55 567.0 122.44 21.6 IM04-2481 Control of autostimulation (Mitomycln C treated autologous cells) 738 1252 464 818.0 400.04 48.9 MLR allogenic donor IM04-2477 (Mitomycin C treated) 13177 24885 15444 17835.3 6209.52 34.8 MLR with cell line (Mitomycin C treated cell type A) 4495 3671 4674 4280.0 534.95 12.5 SI (donor) 31 SI (cell line) 8
- 87 -Plate ID: Plate 2 Analytical Culture Replicates number System 1 2 3 Mean SD
CV
Proliferation baseline of receiver 432 533 274 413.0 130.54 31.6 IM04-2482 Control of autostimulation (Mitomycin C treated autologous cells) 1459 633 598 896.7 487.31 54.3 MLR allogenic donor 1M04-2477 (Mitomycin C treated) 24286 30823 31346 28818.3 3933.82 13.7 MLR with cell line (MItomycin C treated cell type A) 2762 1502 6723 3662.3 2724.46 74.4 SI (donor) /0 SI (cell line) 9 IM04-2477 Proliferation baseline of receiver 312 419 349 360.0 54.34 15.1 (allogenic donor) Control of autostimulation (Mitomycin treated autoiogous cells) 567 604 374 515.0 123.50 24.0 Cell line type A
Proliferation baseline of receiver 5101 3735 2973 3936.3 1078.19 27.4 Control of autostimulation (Mitomycin treated autologous cells) 1924 4570 2153 2882.3 1466.04 50.9 Table 10-5. Average stimulation index of umbilical cells and an allogeneic donor in a mixed lymphocyte reaction with five individual allogeneic receivers.
Average Stimulation Index Recipient Umbilicus Plate 1 (receivers 1-4) 42.75 6.5 Plate 2 (receiver 5) 70 Antigen Presenting Cell Markers - Placenta. Histograms of placenta cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, as noted by fluorescence value consistent with the IgG control, indicating that placental cell lines lack the cell surface molecules required to directly stimulate CD4+ T cells.
Immuno-modulating Markers - Placenta. Histograms of placenta cells analyzed by flow cytometry show positive expression of PD-L2, as noted by the increased value of fluorescence relative to the IgG control, and negative expression of CD178 and HLA-G, as noted by fluorescence value consistent with the IgG control.
Antigen Presenting Cell Markers - Umbilicus. Histograms of umbilical cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, as noted by fluorescence value consistent with the IgG control , indicating that umbilical cell lines lack the cell surface molecules required to directly stimulate CD4+ T cells.
Immuno-modulating Markers- Umbilicus. Histograms of umbilical cells analyzed by flow cytometry show positive expression of PD-L2, as noted by the increased value of fluorescence relative to the IgG control, and negative expression of CD178 and HLA-G, as noted by fluorescence value consistent with the IgG control.
-88-Summary. In the mixed lymphocyte reactions conducted with placental cell lines, the average stimulation index ranged from 1.3 to 3, and that of the allogeneic positive controls ranged from 46.25 to 279. In the mixed lymphocyte reactions conducted with umbilical cell lines the average stimulation index ranged from 6.5 to 9, and that of the allogeneic positive controls ranged from 42.75 to 70. Placental and umbilical cell lines were negative for the expression of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as measured by flow cytometry. Placental and umbilical cell lines were negative for the expression of immuno-modulating proteins HLA-G and CD178 and positive for the expression of PD-L2, as measured by flow cytometry. Allogeneic donor PBMCs contain antigen-presenting cells expressing HLA-DR, DP, DQ, CDS 0, CD86, and B7-H2, thereby allowing for the stimulation of naive CD4+ T cells. The absence of antigen-presenting cell surface molecules on placenta- and umbilical cord-derived cells required for the direct stimulation of naïve CD4+ T
cells and the presence of PD-L2, an imm-uno-modulating protein, may account for the low stimulation index exhibited by these cells in a MLR as compared to allogeneic controls.
Example 11 Plasma Clotting Assay Cell therapy may be injected systemically for certain applications where cells are able to target the site of action. It is important that injected cells not cause thrombosis, which may be fatal. Tissue factor, a membrane-bound procoagulant glycoprotein, is the initiator of the extrinsic clotting cascade, which is the predominant coagulation pathway in vivo. Tissue factor also plays an important role in embryonic vessel formation, for example, in the formation of the primitive vascular wall (Brodsky et al. (2002) Exp. Nephrol. 10:299-306). To determine the potential for PPDCs to initiate clotting, umbilical cord and placenta-derived PPDCs were evaluated for tissue factor expression and their ability to initiate plasma clotting.
Methods & Materials Human Tissue factor. SIMPLASTIN, a human tissue factor (Organon Tekailca Corporation, Durham, NC), was reconstituted with 20 milliliters distilled water. The stock solution was serially diluted (1:2) in eight tubes. Normal human plasma (George King Bio-Medical, Overland Park, KS) was thawed at 37 C in a water bath and then stored in ice before use. To each well of a 96-well plate was added 100 microliters phosphate buffered saline (PBS), microliters diluted SIMPLASTIN (except a blank well), 30 microliters 0.1M
calcium chloride, and 100 microliters of normal human plasma. The plate was immediately placed in a
- 89 -temperature-controlled microplate reader and absorbance measured at 405 nanometers at 40 second intervals for 30 minutes.
J-82 and postpartum cells. J-82 cells (ATCC, MD) were grown in Iscove's modified Dulbecco's medium (IMDM; Gibco, Carlsbad, CA) containing 10% (v/v) fetal bovine serum (FBS; Hyclone, Logan UT), 1 millimolar sodium pyruvate (Sigma Chemical, St.
Louis, MO), 2 millimolar L-Glutamine (Mediatech Herndon, VA), 1 x non-essential amino acids (Mediatech Herndon, VA). At 70% confluence, cells were transferred to wells of 96-well plate at 100,000, 50,000 and 25,000 cells/well. Postpartum cells derived from placenta ana umbilical cord were cultured in Growth Medium in gelatin-coated T75 flasks (Coming, Corning, NY).
Placenta-derived cells at passage 5 and umbilical cord-derived cells at passages 5 and 18 were transferred to wells at 50,000 cells/well. Culture medium was removed from each well after centrifugation at 150 x g for 5 minutes. Cells were suspended in PBS without calcium and magnesium. Cells incubated with anti-tissue factor antibody cells were incubated with 20 micrograms/milliliter CNTO 859 (Centocor, Malvern, PA) for 30 minutes. Calcium chloride (30 microliters) was added to each well. The plate was immediately placed in a temperature-controlled microplate reader and absorbance measured at 405 nanometers at 40 second intervals for 30 minutes.
Antibody Staining. Cells were washed in PBS and detached from the flask with Trypsin/EDTA (Gibco Carlsbad, CA). Cells were harvested, centrifuged, and re-suspended 3%
(v/v) FBS in PBS at a cell concentration of 1x107 permilliliter. Antibody was added to 100 microliters cell suspension as per the manufacturer's specifications, and the cells were incubated in the dark for 30 minutes at 4 C. After incubation, cells were washed with PBS and centrifuged at 150 x g for 5 minutes to remove unbound antibody. Cells were re-suspended in 100 microliters of 3% FBS and secondary antibody added as per the manufacturer's instructions.
Cells were incubated in the dark for 30 minutes at 4 C. After incubation, cells were washed with PBS and centrifuged to remove unbound secondary antibody. Washed cells were re-suspended in 500 microliters of PBS and analyzed by flow cytometry.
Flow Cytometry Analysis. Flow cytometry analysis was performed with a FACSCalibur instrument (Becton Dickinson, San Jose, CA).
Results Flow cytometry analysis revealed that both placenta- and umbilical cord-derived postpartum cells express tissue factor. A plasma clotting assay demonstrated that tissue factor was active. Both placenta- and umbilical cord-derived cells increased the clotting rate as indicated by the time to half maximal absorbance (T V2 to max; Table 11-1).
Clotting was
- 90 -observed with both early (P5) and late (P18) cells. The T 1/2 to max is inversely proportional to the number of J82 cells. Preincubation of umbilical cells with CNTO 859, an antibody to tissue factor, inhibited the clotting reaction, thereby showing that tissue factor was responsible for th_e clotting.
Table 11-1. The effect of human tissue factor (SIMPLASTIN), placenta-derived cells (Pla), and umbilical cord-derived cells (Umb) on plasma clotting was evaluated. The time to half maximal absorbance (T 1/2 to max) at the plateau in seconds was used as a measurement unit.
SIMPLASTIN Dilution T1/2 to max (seconds) 1:2 61 1:4 107 1:8 147 1:16 174 1:32 266 1:64 317 1:128 378 0 (negative control) 1188 J-82 cells 100,000 122 50,000 172 25,000 275 Pla P5 50,000 757 Umb P5 50,000 833 Umb P18 50,000 443 Summary. Placenta- and umbilical cord-derived PPDCs express tissue factor. The addition of an antibody to tissue factor can inhibit tissue factor. Tissue factor is normally found on cells in a conformation that is inactive but is activated by mechanical or chemical (e.g., LPS) stress (Sakariassen et al. (2001) Thromb. Res. 104:149-74; Engstad et al.
(2002) Int.
Immunopharmacol. 2:1585-97). Thus, minimization of stress during the preparation process of PPDCs may prevent activation of tissue factor. In addition to the thrombogenic activity, tissue factor has been associated with angiogenic activity. Thus, tissue factor activity may be beneficial when umbilical cord- or placenta-derived PPDCs are transplanted in tissue but should be inhibited when PPDCs are injected intravenously.
-91-Figure 11-1. Plasma clotting with J82 cells and umbilical cells without (A) and with (B) CNTTO
859, anti-human tissue factor, 20 pg/milliliters. Antibody treatment showed that coagulation was due to tissue factor on umbilical cells.

Differentiation of Postpartum Cells to the Cardiomyocyte Phenotype There is a tremendous need for therapy that will slow the progression of and/or cure heart disease, such as ischemic heart disease and congestive heart failure. Cells that can differentiate into cardiomyocytes that can fully integrate into the patient's cardiac muscle without anthythinias is highly desirable. Rodent mesenchymal stem cells treated with 5-azacytidine have been sh4own to express markers of cardiomyocytes (Fukuda etal. (2002) C. R. Biol. 325:1027-38). The same has not been shown for adult human stem cells. Additional factors have been used to improve stem cell differentiation including low oxygen (Storch (1990) Biochim.
Biophys. Acta 1055:126-9), retinoic acid (Wobus etal. (1997) J Mol. Cell Cardiol. 29:1525-39), DMSO
(Xu etal.
(2002) Circ. Res. 91:501-8), and chelerythrine chloride (International PCT
Publication No.
W003/025149), which effects the translocation of PKC from the cytosol to plasma membrane and is an inhibitor of PKC activity. In the present study, PPDCs (P10) were treated with 5-azacytidine either alone or in combination with DMSO or chelerythrine chloride and markers of cardiomyocytes measured by real-time PCR.
Methods & Materials Cells. Cryopreserved umbilical cord-derived cells (P10) and placenta-derived cells (1324) were grown in Growth Medium in gelatin-coated flasks. Cells were seeded at 5 x 104 cells/well in 96-well plates in Growth Medium for 24 hours. The medium was changed to 0, 3, 10 and_ 30 p,M 5-azacytidine (Sigma, St. Louis, MO) alone or with 5 uM chelerythrine chloride (Sigma), 1% (v/v) dimethylsulfoxide (DMSO) (Sigma), or 1 1iM retinoic acid (Sigma) in MEM-alpha_ (Sigma), insulin, transferrin, and selenium (ITS; Sigma), 10% (v/v) fetal bovine serum, penicillin and streptomycin, and cells incubated at 37 C, 5% (v/v) 02 for 48 or 72 hours.
Media was then changed to MEM-alpha, insulin, transferrin, and selenium, 10% (v/v) fetal bovine serum, penicillin and streptomycin, and cells incubated at 37 C, 5% (v/v) 02 for 14 days.
RNA extraction and Reverse Transcription. Cells were lysed with 150 microliters buffer RLT containing beta-mercaptoethanol (Sigma St. Louis, MO) according to the manufacturer's instructions (RNeasy 96 kit, Qiagen, Valencia, CA) and stored at -80 C. Cell
- 92 -c3471-c lysates were thawed and RNA extracted according to the manufacturer's instructions (RNeasy 96 kit, Qiagen, Valencia, CA) with a 2.7 U/sample DNase treatment (Sigma St.
Louis, MO). RNA
was eluted with 50 microliters DEPC-treated water and stored at -80 C. RNA was reverse transcribed using random hexamers with the TaqMg reverse transcription reagents (Applied Biosystems, Foster City, CA) at 25 C for 10 minutes, 37 C for 60 minutes and 95 C for 10 minutes. Samples were stored at -20 C.
PCR. PCR was performed on cDNA samples using Assays-on-Demand rm gene expression products cardiac myosin (Hs00165276 ml), skeletal myosin (Hs0042&
600), GATA 4 (Hs00171403 ml), GAPDH (Applied Biosystems, Foster City, CA), and TaqMan7 Universal PCR
master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, CA) using a 7000 sequence detection system with ABI prism 7000 SDS software (Applied Biosystems, Foster City, CA). Thermal cycle conditions were initially 50 C for 2 minutes and 95 C for 10 minutes followed by 40 cycles of 95 C for 15 seconds and 60 C for 1 minute.
cDNA from heart and skeletal TT111RCle (Ambion Austin TX) were used as a control.
Results Analysis of RNA from cardiac muscle showed expression of cardiac myosin and GATA
4. Skeletal muscle RNA analysis showed expression skeletal myosin and cardiac myosin but not of GATA 4. Placenta-derived cells (P24) treated for 72 h with factors and grown for a further14 days expressed GATA 4, but no skeletal myosin or cardiac myosin. Umbilical cord-derived cells (P12) treated for 481i with factors and cultured for a further 14 days expressed low levels of GATA 4, but no skeletal myosin or cardiac myosin. Additional samples from placenta and umbilical cord cells that were analyzed showed expression of GATA 4 under the conditions tested.
Summary. Untreated placenta- and umbilical cord-derived cells constitutively express GATA 4, a nuclear transcription factor in cardiomyocytes, sertoli cells, and hepatocytes.

Assessment of Postpartum Cell-Based Cardiovascular Therapy in a Rodent Coronary Ligation Model Animal models of heart failure have increased our understanding of the p athophysiology of the disease and have assisted in the development of new treatments for congestive heart
-93-failure (CHF). Coronary artery occlusion, or the blocking of the vessels that supply the heart tissue, in the rat closely mimics the pathophysiology of acute myocardial infarction in humans and has been used successfully to study pharmacological interventions for CHF.
Transplantation of human cells into cardiac lesions is a potential viable therapeutic treatment for CHF.
The objective of this study was to determine the efficacy of intracardiac human cell treatment when administered 15 minutes post-coronary artery occlusion in. a rodent model of myocardial ischemia/infarction.
Methods & Materials The Charles River Worcester, MA test facility is accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care, International (AAALAC) and registered with the United States Deparnuent of Agriculture to conduct research in laboratory animals. All the conditions of testing conform to the Animal Welfare Act (9 CFR) and its amendments. The protocol was reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) at the Test Facility for compliance with regulations prior to study initiation.
The animals having characteristics identified in Table 13-1 were individually housed in micro-isolator cages on autoclaved bedding. The cages conform to standards set forth in The Guide for the Care and Use of Laboratory Animals.
Table 13-1. Animal characteristics Species: Rattus norvegicus Strain: Rnu Source: Charles River Laboratories Age at Dosing: 6-8 weeks Weight at Dosing: ¨200-250 g Number of Males (including spares): 40 + 10 Purina Certified Diet (irradiated) was provided to the animals ad libitum.
This diet was routinely analyzed by the manufacturer for nutritional components and environmental contaminants. Results of the manufacturer's analyses are on file at the Test Facility.
Autoclaved Filtered tap water was provided ad libitum. Samples of the filtered water were analyzed for total dissolved solids, hardness, specified microbiological content, and selected environmental contaminants. Results of these analyses are on file at the Test Facility.
- 94 -Environmental controls were set to maintain temperatures of 18 to 26 C (64- to 79 F) with a relative humidity of 30% to 70%. A 12:12 hour light:dark cycle was maintained. Ten or greater air changes per hour were maintained in the animal rooms. Upon receipt and prior to use on the study, the animals were held for a minimum of four days for conditioning according to the Test Facility Vendor Management Program as described in the Test Facility Standard Operating Procedure, Receipt, Conditioning, and Quarantine of Laboratory Animals.
Each animal was identified by a unique number and this number was indicated by an ear punch. Animals were randomly assigned to groups by a weight-ordered distributiDn such that individual body weights did not exceed 120% of mean weight.
The animals were anesthetized with sodium pentobarbital (40 milligrams/kilogram) and buprenorphine(0.05milligrams/kilogram) as a single cocktail given intramuscularly (IM).
Following the establishment of anesthesia, animals were intubated using an 18-16 gauge, 2-inch length angiocath, or appropriate sized angiocath, and maintained on room air respiration (supplemented with oxygen) and a positive pressure ventilator throughout the surgical procedure.
Additional anesthesia was given incrementally as needed. Preoperative antibiotic therapy was also administered, Benzathine/Procaine penicillin G, 40,000 Units/kilogram, IM. Additional antibiotic therapy was administered every 48 hours.
Electrode pads were placed around the appropriate paws of the animals to receive a useable ECG signal. Animals were positioned on a heating pad to help maintain body temperature throughout the procedure. A rectal temperature probe was inserted into the animal to monitor body temperature. Ophthalmic ointment was administered to each eye.
The surgical sites (thoracic area) were prepared for aseptic surgery by removing any excess fur, and gently wiping the area with sponges that have been soaked in 70% isopropyl alcohol, which was allowed to dry. Medi SeppsTM or similar solution was then applied to the area and also allowed to dry. The area was appropriately draped for strict aseptic surgery.
A surgical incision was made on the skin over the fourth intercostal space.
Blunt dissection through the muscle layers was used to access the thoracic cavity. A
retractor was carefully inserted into the fourth intercostal space and opened to allow access to th_e interior cavity. The pericardium was carefully opened via gentle teasing with cotton swabs dampened in sterile saline solution. A damp cotton swab was used to gently push the apex of th_e heart into the opening where a length of 6-0 silk suture was attached into the myocardium for manipulation of the heart. After a pause to allow the heart to recover, the suture placed in the apex was used to ease the heart out of the chest cavity and to place sufficient tension on the heart to allow access to the upper heart and the left anterior descending coronary artery (LAD).
Another length of 6-0
- 95 -silk suture was placed into the myocardium so as to surround the LAD. The pressure on the apical suture was released and the heart allowed to return to the interior of the chest cavity.
Once the heart rate and ECG returned to baseline values, the ligatures around the LAD
were tied off to occlude the LAD. This was a permanent occlusion with the s-uture tied off and the ends trimmed. Once the ligature was tied, the surgeon looked for the following indications of successful occlusion: change in color of the area of the heart directly below the ligature to a white/grayish white as a result of the termination of blood flow to the area and a significant change in the ECG corresponding to occlusion of the LAD. Arrhythmias may have developed within the first 10 minutes of the occlusion. The rat was monitored closely during this time period in the event that resuscitation was necessary. In the event of severe arrhythmia and failure of the rat to convert to normal sinus rhythm without assistance, aid was rendered via cardiac massage. Approximately 15 minutes following the initiation of the LAD
occlusion, the area of left ventricle made ischemic was treated with either vehicle or test article by direct injection into the ischemic myocardium. Treatment consisted of three to ten intramyocardial injections (100 microliters/injection) into the ischemic zone of myocardium.
Human cells were grown in Growth Medium in gelatin-coated T300 flasks. Cells were washed with phosphate buffered saline (PBS, Gibco, Carlsbad CA) and trypsinized using Trypsin/EDTA (Gibco, Carlsbad CA). The trypsinization was stopped by adding Growth Medium. The cells were centrifuged at 150 x g, supernatant removed, and the cell pellet was resuspended in approximately 1 milliliter Growth Medium per million cells. An aliquot of cells was removed and added to trypan blue (Sigma, St. Louis, MO). The viable cell number was estimated using a hemocytometer. The cell suspension was centrifuged and resuspended in 1 milliliters Growth containing 10% (v/v) DMSO (Hybrimax, Sigma, St. Louis, MO) per 5 million cells and transferred into Cryovials (Nalgene). The cells were cooled at approximately 1 C/min overnight in a -80 C freezer using a "Mr Frosty" freezing container (Nalgen_e, Rochester, NY).
Vials of cells were transferred into liquid nitrogen. Vials were shipped from CBAT, Somerville, NJ to Charles River, Worcester, MA on dry ice and stored at -80 C.
Approximately 1-2 hours before injection of cells into the animal, a vial of cells was thawed rapidly in a 37 C water bath.
Under aseptic conditions in a BSL2 biosafety cabinet, cells were added to 40 milliliters PBS with magnesium and calcium (Sigma St. Louis, MO) and centrifuged at 150 x g for 5 minutes before resuspending the cell pellet in 10 milliliters PBS. The cell number and viability was estimated as described above. The cells were centrifuged at 150 x g for 5 minutes and resuspended in PBS at a fmal concentration of 106 viable cells /100 microliters. The cell suspension was loaded into 1
- 96 -milliliter syringes with a 30G needle and kept on ice. Viability was assessed again up to 5 hours on ice.
Following the administration of treatment (Table 13-2) and stabilization of the heart, the surgeon began closing the surgical incision. The retractor was removed. The lungs were over-inflated for 3 ¨ 4 breaths and visually inspected as much as possible to ensure that they were fully re-inflated. This created a negative pressure necessary to prevent pneumothorax post-recovery. To evacuate fluid and excess air from the thoracic cavity after closing the cavity, an intravenous catheter (i.e., 20 gauge, 2 mm in length) was placed through the skin and muscle layers so that the tip remains in the thoracic cavity. Care was taken so that the tip did not pierce the lung or heart. The separated ribs and associated muscle was sutured together with appropriate suture. The upper layers of muscle was sutured using a simple continuous pattern.
The skin was closed with 4-0 silk using a horizontal mattress pattern. A 10 milliliter syringe was attached to the intravenous catheter that had been previously placed in the thoracic cavity and the plunger slowly pulled back to withdraw fluids and air from the cavity. At the same time, the catheter was slowly withdrawn from the entry site, thereby allowing the surrounding muscle mass and skin to seal the puncture. The surgical drape was removed and fluids (i.e., lactated Ringers solution, 25 milliliters/kilogram subcutaneously [SC] or intraperitonealty [IP]) were given.
Table 13-2 Treatment regimens Gr. No. of Dosage Dose Conc.
Level (cells/millili Route .1 Dose Time of Necropsy Test Article Treatment No. Males RegimenDay (cells/animal) ters) Administration 1 8 Vehicle 0 0 Direct 2 8 Placenta #4 (P10) injection(s) into (A) the ischemic region of the left 15 minutes Umbilical (P10) ventricle of the afr n Day 28 8 te coro (B) heart, consisting ( 1 Day) artery ligati ary on 1 million 10 million of 3 to 10 Placenta #3 (P10) intramyocardial (C) injections of 100 p1 total.
Human 8 fibroblasts 1F1853 (P10) (D) Gr. = Group; No. = Number; Conc. = Concentration
- 97 -Immediately after each rat had undergone treatment with test article and the incision sutured, the animal underwent an echocardiography (ECG) examination.
Anesthesia was maintained throughout the completion of the echo examination. Upon the completion of the echo examination, ventilation was discontinued, and the rat was returned to the recovery area to recover in a heated, oxygenated recovery cage.
A second echo examination of each surviving animal was completed at the end of the study (approximately 28 days post-treatment), prior to termination. During the second examination, the animals were anesthetized as described previously.
For each echo examination, the left thoracic area was shaved, and warmed, ultrasonic gel was applied to the skin to enhance contact with the transducer. Electrode pads were placed around the appropriate extremities to receive an ECG signal. Echocardiographic images included short axis and long axis views to allow for the determination of ventricular cavity dimensions, contractility, blood flow through vasculature, and wall thickness.
These images were saved on optical disk for further analysis. After examination, the gel medium was removed from the skin with gauze or paper towel. The rat was removed from the ventilator and placed in a warmed recovery cage until mobile.
At the conclusion of the surgical procedures, respiratory ventilation was turned off. The animals were observed for pedal reflex. The rectal probe and ECG electrodes subsequently were removed, and the animal was extubated and placed in a warmed oxygenated recovery cage.
After complete recovery from anesthesia, the animals were given buprenorphine (0.05 milligrams/kilogram, SC). Observations were made regularly until the animals showed full mobility and an interest in food and water. The animals then were placed in a clean housing cage and returned to the animal housing room. Animals were monitored for surgical incision integrity twice daily post-surgery.
Analgesics (i.e., Buprenorphine, 0.05 milligrams/kilogram SC.) were given twice daily for 4 days post-operatively and thereafter as needed. Visual indications of post-operative pain include lack of normal body postures and movement (e.g., animal remains in hunched position), antipathy, lack of eating/drinking, lack of grooming, etc.
Body weight was recorded for each animal prior to initial treatment, weekly thereafter, and on the day of necropsy. Animals found dead were weighed and necropsied.
In order for the heart to be harvested, each rat was anesthetized as was done for surgery.
The jugular vein was cannulated. The heart was arrested in diastole with KC1 infused via the jugular cannula. The heart was then removed from the thoracic cavity. A
limited necropsy was
- 98 -G=1 A' 7 4 G
.J.)4' I I
then performed on the heart after which the heart was placed in 10% neutral bu.ffered formalin.
The remainder of each carcass was then discarded with no further evaluation.
Hearts of all animals that were found dead or euthanized moribund were placed in 4%
paraformaldehyde until evaluated. The remainder of each carcass was then discarded with no further evaluation.
Histology and Image Analysis. Fixed tissues sectioned with a stainless steel coronal heart matrix (Harvard Apparatus Holliston, MA) yielded four two-millimeter thick serial tissue sections. Sections were processed and serially embedded in paraffin using routine methods.
Five-micron sections were obtained by micro tome and stained Masson's Tri-chrome for Connective Tissue (Poly Scientific Bay Shore, NY) using manufacturer's methods. Electronic photomicrographs were captured and analyzed using image analysis methods d_eveloped by Phase 3 Imaging System (Glen Mills, PA). Photomicrographs of the tri-chrome stained sections were color-metrically analyzed electronically to determine the overall area of the ventricle and free wall and the area of the differential staining.
Results There was no loss in the initial viability of cells over 5 hours in the vehicle when kept on ice. Cells were injected into the infarct with one to three needle entry points arid multiple changes in direction of needle orientation.
Echocardiography measurements taken from the infarct-treated rats were used to evaluate the results. The left ventricle fractional shortening and the ejection fraction were similarly utilized. These values were calculated as described by Sahn et al. (1978) Circzdation 58:1072-1083. The fractional shortening of the vehicle-treated animals was significantly decreased from 47.7% 8.3% at Day 0 to 23.5% 30.2% at Day 28 (p<0.05). The animals that were treated with postpartum-derived cells showed small, non-significant differences between the fractional shortening between Day 0 and 28. There was no significant difference between the fractional shortening between the groups at Day 0. Each group had eight animals at the start but some did not survive the experiment. The fibroblast-treated animals experienced greater mortality (80%) than the groups treated with PPDCs.
Hearts collected at the study termination were subjected to histological analysis. The hearts were arrested in diastole and fixed. The results were calculated from an algorithm to estimate the percentage of total heart area that comprises the infarct. The infarct size in the vehicle-treated animals was 22.9% 1 6.7% of heart area, while the infarct size in hearts treated = with placenta-derived cells (isolate 1) was 13.9% 3.7%, with umbilical cord cells was 12.5%
-99-2.5%, with placenta-derived cells (isolate 2) was 12.9% 3.4%, and with fibroblasts was 19.3%
+ 8.0%. The difference of infarct size of cell-treated animals relative to vehicle-treated animals was not statistically significant by either Student's t test or ANOVA.
Summary. The results of the present study suggest that the postpartum-derived cells may have some benefit in reducing the damage of a surgically induced myocardial infarction in rats. The vehicle-treated animals showed a significant reduction in cardiac function_ at day 28 as compared to day 0, as measured by fractional shortening, while the placenta-and umbilical cord-derived cell-treated animals showed minimal change over the 28-day study. The fibroblast-treated animals showed minimal change but only two animals survived the study.
Evaluation of infarct size suggested that there may be some modest, but not statistically significant, reduction in the infarct size in the postpartum-derived cell-treated animals as compared to the vehicle controls at Day 28. Taken together, these data establish efficacy of the postpartum¨derived cells in reducing damage from myocardial infarction.
Example 14 PPDCs Can Be Injected as a Matrix-Cell Complex Cells derived from the postpartum umbilical cord are useful for regenerative therapies.
The tissue produced by postpartum-derived cells transplanted into SCID mice with a biodegradable material was evaluated. The materials evaluated were Vicryl non-woven, 35/65 PCL/PGA foam, and RAD 16 self-assembling peptide hydrogel.
Methods & Materials Cell Culture. Umbilical cord-derived cells were grown in Growth Mediuna in gelatin-coated flasks.
Sample Preparation. One million viable cells were seeded in 15 microliters Growth Medium onto 5 mm diameter, 2.25 mm thick Vicryl non-woven scaffolds (64.33 in_g/cc;
Lot#3547-47-1) or 5 mm diameter 35/65 PCL/PGA foam (Lot# 3415-53). Cells were allowed to attach for two hours before adding more Growth Medium to cover the scaffolds.
Cells were grown on scaffolds overnight. Scaffolds without cells were also incubated in medium.
RAD16 self-assembling peptides (3D Matrix, Cambridge, MA under a material transfer agreement) was obtained as a sterile 1% (w/v) solution in water, which was mixed 1:1 with 1 x 106 cells in 10% (w/v) sucrose (Sigma, St Louis, MO), 10 millimolar HEPES in Dulbecco's
- 100 -modified medium (DMEM; Gibco) immediately before use. The final concentratia.n of cells in RAD16 hydrogel was 1 x 106 cells/100 microliters.
TEST MATERIAL (N=4/1&) 1. Vicryl non-woven + 1 x 106 umbilical cord-derived cells 2. 35/65 PCL/PGA foam + 1 x 106 umbilical cord-derived cells 3. RAD 16 self-assembling peptide + 1 x 106 umbilical cord-derived cells 4. 35/65 PCL/PGA foam 5. Vicryl non-woven Animal Preparation. The animals utilized in this study were handled and maintained in accordance with the current requirements of the Animal Welfare Act. Compliance with the above Public Laws were accomplished by adhering to the Animal Welfare regulations (9 CFR) and conforming to the current standards promulgated in the Guide for the Care and Use of Laboratory Animals, 7th edition.
Mice (Mus Musculus)/Fox Chase SCID/Male (Harlan Sprague Dawley, Inc., Indianapolis, Indiana), 5 weeks of age. All handling of the SCID mice took place -under a hood.
The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligrams/kilogram KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligrams/kilogram ROMPUN (xylazine, Mobay Corp., Shawnee, Kansas) and saline. After induction of anesthesia, the entire back of the animal from the dorsal cervical area to the dorsal lumbosacral area was clipped free of hair using electric animal clippers. The area was then scrubbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine.
Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period_ Subcutaneous Implantation Technique. Four skin incisions, each approximately 1.0 cm in length, were made on the dorsum of the mice. Two cranial sites were located transversely over the dorsal lateral thoracic region, about 5-mm caudal to the palpated inferior edge of the scapula, with one to the left and one to the right of the vertebral column.
Another two were placed transversely over the gluteal muscle area at the caudal sacro-lumbar level, about 5-mm caudal to the palpated iliac crest, with one on either side of the midline.
Implants were randomly placed in these sites. The skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected for RAD16) about 1-cm caudal to the incision.
- 101 -The appropriate test material was implanted into the subcutaneous space. The skin incision was closed with metal clips.
Animal Housing. Mice were individually housed in microisolator cages throughout the course of the study within a temperature range of 64 F - 79 F and relative humidity of 30% to 70%, and maintained on an approximate 12 hour light/12 hour dark cycle. The temperature and relative humidity were maintained within the stated ranges to the greatest extent possible. Diet consisted of Irradiated Pico Mouse Chow 5058 (Purina Co.) and water fed ad libitum.
Mice were euthanized at their designated intervals by carbon dioxide inhalation. The subcutaneous implantation sites with their overlying skin were excised and frozen for histology.
Histology. Excised skin with implant was fixed with 10% neutral buffered formalin (Richard-Allan Kalamazoo, MI). Samples with overlying and adjacent tissue were centrally bisected, paraffin-processed, and embedded on cut surface using routine methods. Five-micron tissue sections were obtained by microtome and stained with hematoxylin and eosin (Poly Scientific Bay Shore, NY) using routine methods.
=
Results There was minimal in growth of tissue into foams implanted subcutaneously in SCID
mice after 30 days. In contrast there was extensive tissue fill in foams implanted with umbilical-derived cells.
There was some tissue in growth in Vicryl non-woven scaffolds. Non-woven scaffolds seeded with umbilical cord-derived cells showed increased matrix deposition and mature blood vessels.
Summary. The purpose of this study was to determine the type of tissue formed by cells derived from human umbilical cord in scaffolds in immune deficient mice.
Synthetic absorbable non-woven/foam discs (5.0 mm diameter x 1.0 mm thick) or self-assembling peptide hydrogel were seeded with cells derived from human umbilical cord and implanted subcutaneously bilaterally in the dorsal spine region of SCID mice. The present study demonstrates that postpartum-derived cells can dramatically increase good quality tissue formation in biodegradable scaffolds.

Endothelial Network Formation: PPDCs Promote Angiogenesis In Vitro
-102-' Angiogenesis, or the formation of new vasculature, is necessary for the growth of new tissue. Induction of angiogenesis is an important therapeutic goal in many pathological conditions. The present study was aimed at identifying potential angiogenic activity of the postpartum cells in in vitro assays. The study followed a well-established method of seeding endothelial cells onto a culture plate coated with a basement membrane extract (Nicosia and Ottinetti (1990) In Vitro Cell Dev. Biol. 26(2):119-28). Treating endothelial cells on extracellular matrix with angiogenic factors will stimulate the cells to form a network that is similar to capillaries. This is a common in vitro assay for testing stimulators and inhibitors of blood vessel fonnation (Ito et al. (1996) Int. J. Cancer 67(1):148-52). The present studies made use of a co-culture system with the postpartum cells seeded onto culture well inserts. The permeable inserts allow for the passive exchange of media components between the endothelial and the postpartum culture media.
Material & Methods Cell Culture.
Postpartum tissue-derived cells. Human umbilical cords and placenta were received and cells were isolated as previously described (Example 1). Cells were cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37 C with 5%
CO2. Cells used for experiments were from about passage 4 to 12.
Actively growing postpartum cells were trypsinized, counted, and seeded onto Costar Transwelle 6.5 mm diameter tissue culture inserts (Corning, Coming, NY) at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium at 37 C with 5%
CO2.
Human mesenchymal stem cells (hMSC). hMSCs were purchased from Cambrex (Walkersville, MD) and cultured in MSCGM (Cambrex). The cultures were incubated at 37 C
with 5% CO2.
Actively growing MSCs were trypsinized, counted and seeded onto Costar Transwell 6.5 mm diameter tissue culture inserts (Corning, Corning, NY) at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium at 3'7 C with 5%
CO2.
Human umbilical vein endothelial cells (HUVEC). HUVEC were obtained from Cambrex (Walkersville, MD). Cells were grown in separate cultures in either EBM or EGM
- 103 -endothelial cell media (Cambrex). Cells were grown on standard tissue cultured plastic at 37 C
with 5% CO2. Cells used in the assay were from about passage 4 to 10.
Human coronary artery endothelial cells (HCAEC). HCAEC were purchased from Cambrex Incorporated (Walkersville, MD). The cells were maintained in separate cultures in either the EBM or EGM media formulations. Cells were grown on standard tissue culture plastic at 37 C with 5% CO2. Cells used for experiments were from about passage 4 to 8.
MatrigelTM assays. Culture plates were coated with MatrigelT/%4 according to manufacturer's specifications. Briefly, MatrigelTM (BD Discovery Labware, Bedford, MA) was thawed at 4 C and approximately 250 microliters was aliquoted and distributed evenly onto each well of a chilled 24-well culture plate (Coming). The plate was then incubated at 37 C for 30 minutes to allow the material to solidify. Actively growing endothelial cell cultures were trypsinized and counted. Cells were washed twice in Growth Media with 2% PBS
by centrifugation, resuspension, and aspiration of the supernatant. Cells were seeded onto the coated wells 20,000 cells per well in approximately 0.5 milliliters Growth Medium with 2% (v/v) PBS. Cells were then incubated for approximately 30 minutes to allow cells to settle.
Endothelial cell cultures were then treated with either 10 nanornolar human bFGF
(Peprotech, Rocky Hill, NJ) or 10 nanomolar human VEGF (Peprotech, Rocky Hill, NJ) to serve as a positive control for endothelial cell response. Transwell inserts seeded with postpartum cells were added to appropriate wells with Growth media with 2% FB S in the insert chamber.
Cultures were incubated at 37 C with 5%CO2 for approximately 24 hours. The well plate was removed from the incubator, and images of the endothelial cell cultures were collected with an Olympus inverted microscope (Olympus, Melville, NY).
Results In a co-culture system with placenta-derived cells or with umbilical cord-derived cells, HUVEC form cell networks. HUVEC cells form limited cell networlcs in co-culture experiments with hMSC and with 10 nanomolar bFGF. HUVEC cells without any treatment showed very little or no network formation. These results suggest that the postpartum cells release angiogenic factors that stimulate the HUVEC.
In a co-culture system with placenta-derived cells or with umbilical cord-derived cells, CAECs form cell networks.
Table 15-1 shows levels of known angiogenic factors released by the postpartum cells in Growth Medium. Postpartum cells were seeded onto inserts as described above.
The cells were
- 104 -cultured at 37 C in atmospheric oxygen for 48 hours on the inserts and then switched to a 2%
FBS media and returned at 37 C for 24 hours. Media were removed, immediately frozen and stored at -80 C, and analyzed by the SEARCHLIGHT multiplex ELISA assay (Pierce Chemical Company, Rockford, IL). Results shown are the averages of duplicate measurements. The results show that the postpartum cells do not release detectable levels of platelet-derived growth factor-bb (PDGF-bb) or heparin-binding epidermal growth factor (HBEGF). The cells do release measurable quantities of tissue inhibitor of metallinoprotease-1 (TIP-1), angiopoietin 2 (ANG2), thrombopoietin (TPO), keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEGF).
Table 15-1. Potential angiogenic factors released from postpartum cells.
Postpartum cells were cultured in 24 hours in media with 2% FBS in atmospheric oxygen. Media were removed and assayed by the SEARCHLIGHT multiplex ELISA
assay (Pierce). Results are the means of a duplicate analysis. Values are concentrations in the media reported in picograms per milliliter of culture media.

(pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) Placenta (P4) 91655.3 175.5 <2.0 275.5 3.0 58.3 7.5 644.6 <1.2 Placenta (P11) 1592832.4 28.1 <2.0 1273.1 193.3 5960.3 34.8 12361.1 1.7 Umbilical cord (P4) 81831.7 <9.8 <2.0 365.9 14.1 200.2 5.8 <4.0 <1.2 Media alone <9.8 25.1 <2.0 <6.4 <2.0 <3.2 <5.4 <4.0 <1.2 Table 15-2 shows levels of known angiogenic factors released by the postpartum cells.
Postpartum cells were seeded onto inserts as described above. The cells were cultured in Growth Medium at 5% oxygen for 48 hours on the inserts and then switched to a 2% FBS
medium and returned to 5% 02 incubation for 24 hours. Media were removed, immediately frozen, and stored at -80 C, and analyzed by the SEARCHLIGHT multiplex ELISA assay (Pierce Chemical Company, Rockford, IL). Results shown are the averages of duplicate measurements. The =
results show that the postpartum cells do not release detectable levels of platelet-derived growth factor-bb (PDGF-13B), or heparin-binding epidermal growth factor (HBEGF). The cells do release measurable quantities of tissue inhibitor of metallinoprotease-1 (TIMP-1), angiopoietin 2 (ANG2), thrombopoietin (TPO), keratinocyte growth factor (KGF), hepatocyte growth factor (HGF), fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF).
Table 15-2. Potential angiogenic factors released from postpartum cells.
Postpartum cells were cultured in 24 hours in media with 2% FBS in 5% oxygen.
- 105 -5.1471-c Media were removed and assayed by the SEARCHLIGHT multiplex ELISA assay (Pierce). Results are the means of a duplicate analysis. Values are concentrations in the media reported in picograms per milliliter of culture media.

(pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) (pg/mL) Placenta (P4) 72972.5 253.6 <2.0 743.1 2.5 30.2 15.1 1495.1 <1.2 Placenta (P11) 458023.1 55.1 <2.0 2562.2 114.2 2138.0 295.1 7521.3 1.8 Umbilical cord (P4) 50244.7 <9.8 <2.0 403.3 10.7 156.8 5.7 <4.0 <1.2 Media alone <9.8 25.1 <2.0 <6.4 <7.0 <3.2 <5.4 <4.0 <1.2 Summary. Postpartum cells can stimulate both human umbilical vein and coronary artery endothelial cells to form networks in an in vitro MatrigelTm assay.
This effect is similar to that with known angiogenic factors in this assay system, suggesting that the postpartum cells are useful for stimulating angiogenesis in vivo.
Biological Deposit of Postpartum-Derived Cells and Cultures Consistent with the detailed description and the written examples provided herein, examples of umbilicus-derived cells of the invention were deposited with the American Type Culture Collection (ATCC, Manassas, VA) on June 10, 2004, and assigned ATCC
Accession Numbers as follows: (1) strain designation 'UMB 022803 (P7) was assigned Accession No.
PTA-6067; and (2) strain designation UIVIB 022803 (P17) was assigned Accession No.. PTA- , 6068.
As with the umbilicus-derived cells, examples of placenta-derived cells of the invention were also deposited with the American Type Culture Collection (ATCC, Manassas, VA) and assigned ATCC Accession Numbers as follows: (1) strain designation PLA 071003 (P8) was deposited June 15, 2004 and assigned Accession No. PTA-6074; (2) strain designation. PLA
071003 (P11) was deposited June 15 , 2004 and assigned Accession No. PTA-6075;
and (3) strain designation PLA 071003 (P16) was deposited June 16, 2004 and assigned Accession No.
PTA-6079.
= - 106 -SEQUENCE LISTING
<110> Ethicon, Incorporated Harris, Ian Ross Harmon, Alexander M.
Kihm, Anthony J.
Messina, Darin J.
Mistry, Sanjay Seyda, Agnieszka Yi, Chin-Feng Gosiewska, Anna <120> POSTPARTUM-DERIVED CELLS FOR USE IN TREATMENT OF DISEASE OF THE
HEART AND CIRCULATORY SYSTEM
<130> CBAT-0233 <140> PCT/US2004/020957 <141> 2004-06-25 <150> US 60/483,264 <151> 2003-06-27 <160> 10 <170> PatentIn version 3.3 <210> 1 <211> 22 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 1 gagaaatcca aagagcaaat gg 22 <210> 2 <211> 21 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 2 agaatggaaa actggaatag g 21 <210> 3 <211> 20 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 3 tcttcgatgc ttcggattcc 20 <210.> 4 <211> 21 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 4 gaattctcgg aatctctgtt g 21 <210> 5 <211> 21 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 5 ttacaagcag tgcagaaaac c 21 <210> 6 <211> 22 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 6 agtaaacatt gaaaccacag cc 22 <210> 7 <211> 20 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 7 tctgcagctc tgtgtgaagg 20 <210> 8 <211> 22 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 8 cttcaaaaac ttctccacaa cc 22 <210> 9 <211> 17 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 9 cccacgccac gctctcc 17 <210> 10 <211> 19 <212> DNA
<213> Artificial <220>
<223> Synthetic Construct <400> 10 tcctgtcagt tggtgctcc 19

Claims (45)

1. A pharmaceutical composition for use in the treatment of a disease of the heart or circulatory system comprising a pharmaceutically acceptable carrier and isolated umbilical-derived cells, wherein the cells are capable of self-renewal and expansion in culture, have the potential to differentiate into a cell of cardiomyocyte phenotypes; and have each of the following characteristics:
potential for at least about 40 doublings in culture;
production of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
lack of production of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; and expression of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3.
2. The pharmaceutical composition of claim 1, wherein the umbilical-derived cells are isolated in the presence of one or more enzyme activities comprising metalloprotease activity, mucolytic activity and neutral protease activity.
3. The pharmaceutical composition of claim 2, wherein the enzyme activities are collagenase and neutral protease.
4. The pharmaceutical composition of claim 3, further comprising hyaluronidase.
5. The pharmaceutical composition of claim 1, wherein the cells comprise a normal karyotype.
6. The pharmaceutical composition of claim 1, wherein the cells maintain their karyotype upon passage.
7. The pharmaceutical composition of claim 1, wherein the cells further comprise one or more of the following characteristics:
secretion of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, M1P1b, RANTES, and TIMP1;
lack of secretion of TGF-beta2, ANG2, PDGFbb, MIP1a, [309, MDC, and VEGF, as detected by ELISA;
ability to expand in the presence of oxygen from about 5% to about 20%;
and increased expression of chemokine receptor (C-X-C motif) ligand 1, granulocyte chemotactic protein 2 (GCP-2), tumor necrosis factor and alpha-induced protein 3 relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell.
8. The pharmaceutical composition of any one of claims 1 to 7, wherein the cells do not spontaneously differentiate along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway when cultured in Growth Medium.
9. The pharmaceutical composition of any one of claims 1 to 7 comprising from about 1% cells to about 10% cells.
10. The pharmaceutical composition of any one of claims 1 to 7, wherein the cells comprise a clonal cell line of umbilical-derived cells.
11. The pharmaceutical composition of any one of claims 1 to 7, wherein the composition comprises umbilical-derived cells incubated in the presence of one or more factors which stimulate stem cell differentiation along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
12. The pharmaceutical composition of claim 11, wherein the factors comprise at least one of a demethylation agent, a member of BMP, FGF, TAK, GATA, Csx, NK, MEF2, ET-1, and Wnt factor families, Hedgehog, Csx/Nkx-2.5, and anti-Wnt factors.
13. The pharmaceutical composition of claim 12, wherein the demethylation agent comprises an inhibitor of DNA methyltransferase or an inhibitor of a histone deacetylase, or an inhibitor of a repressor complex.
14. The pharmaceutical composition of claim 12, wherein the demethylation agent comprises at least one of 5-azacytidine, 5-aza-2'-deoxycytidine, DMSO, chelerythrine chloride, retinoic acid or salts thereof, 2-amino-4-(ethylthio)butyric acid, procainamide, and procaine.
15. The pharmaceutical composition of any one of claims 1 to 7, wherein the cells are seeded onto a matrix to form a matrix-cell complex.
16. The pharmaceutical composition of claim 15, wherein the matrix is a scaffold.
17. The pharmaceutical composition of claim 16, wherein the scaffold is bioabsorbable.
18. The pharmaceutical composition of claim 17, wherein the scaffold further comprises at least one or more stem cells in addition to the umbilical-derived cells.
19. The pharmaceutical composition of any one of claims 1 to 18, wherein the disease of the heart or circulatory system is a cardiomyopathy.
20. The pharmaceutical composition of claim 19 wherein the cardiomyopathy is idiopathic.
21. The pharmaceutical composition of claim 20, wherein the cardiomyopathy is ischemic cardiomyopathy or nonischemic cardiomyopathy.
22. The pharmaceutical composition of claim 21, wherein the cardiomyopathy is a nonischemic cardiomyopathy selected from dilated cardiomyopathy, hypertrophic cardiomyopathy, and restrictive cardiomyopathy.
23. The pharmaceutical composition of any one of claims 1 to 22, which is for administration by injection.
24. The pharmaceutical composition of claim 23, wherein the injection is intracardiac injection.
25. The pharmaceutical composition of any one of claims 1 to 7, comprising about 50% umbilical-derived cells.
26. The pharmaceutical composition of any one of claims 1 to 7, further comprising one or more of an antithrombogenic agent, an anti-inflammatory, an immunosuppressive agent, an immunomodulatory agent, and an antiapoptotic agent.
27. The pharmaceutical composition of any one of claims 1 to 7, wherein the pharmaceutical composition is formulated for injection.
28. The pharmaceutical composition of claim 27, wherein the pharmaceutical composition is formulated for intracardiac injection.
29. The pharmaceutical composition of any one of claims 1 to 7, further comprising one or more factors which stimulate stem cell differentiation along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
30. The pharmaceutical composition of claim 29, wherein the factors comprise at least one of a demethylation agent, a member of BMP, FGF, TAK, GATA, Csx, NK, MEF2, ET-1, and Wnt factor families, Hedgehog, Csx/Nkx-2.5, and anti-Wnt factors.
31. The pharmaceutical composition of claim 30, wherein the demethylation agent comprises an inhibitor of DNA methyltransferase, an inhibitor of a histone deacetylase, or an inhibitor of a repressor complex.
32. The pharmaceutical composition of claim 30, wherein the demethylation agent comprise at least one of 5-azacytidine, 5-aza-2'-deoxycytidine, DMSO, chelerythrine chloride, retinoic acid or salts thereof, 2-amino-4-(ethylthio)butyric acid, procainamide, and procaine.
33. Use of isolated umbilical-derived cells as defined in claim 1, for the treatment of a disease of the heart or circulatory system.
34. Use of isolated umbilical-derived cells as defined in claim 1, in the manufacture of a medicament for the treatment of a disease of the heart or circulatory system.
35. Use of an effective amount of umbilical-derived cells in the treatment of a disease of the heart or circulatory system in a patient, wherein the cells are derived from umbilical cord tissue substantially free of blood, wherein the cells are capable of self-renewal and expansion in culture, have the potential to differentiate into a cell of cardiomyocyte phenotypes; and have each of the following characteristics:
potential for at least about 40 doublings in culture;
production of 0D10, CD13, 0D44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
lack of production of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; and expression of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3.
36. The use of claim 35, wherein the cells further comprise one or more of the following characteristics:

secretion of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1b, RANTES, and TIMP1;
lack of secretion of TGF-beta2, ANG2, PDGFbb, MIP1a, I309, MDC, and VEGF, as detected by EL1SA;
ability to expand in the presence of oxygen from about 5% to about 20%;
and increased expression of chemokine receptor (C-X-C motif) ligand 1, granulocyte chemotactic protein 2 (GCP-2), tumor necrosis factor and alpha-induced protein 3 relative to that of a human cell which is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell.
37. The use of claim 35, wherein the disease of the heart or circulatory system is a cardiomyopathy.
38. The use of claim 37, wherein the cardiomyopathy is idiopathic.
39. The use of claim 37, wherein the cardiomyopathy is ischemic cardiomyopathy or nonischemic cardiomyopathy.
40. The use of claim 39, wherein the cardiomyopathy is a nonischemic cardiomyopathy selected from dilated cardiomyopathy, hypertrophic cardiomyopathy, and restrictive cardiomyopathy.
41. The use of claim 35, wherein the use further comprises inducing the cells along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
42. The use of claim 41, wherein the cells are induced in vitro.
43. The use of any of claims 35 to 42, wherein the patient is syngeneic with the umbilical-derived cells.
44. The use of any of claims 35 to 42, wherein the patient is allogeneic with the umbilical-derived cells.
45. The use of any of claims 35 to 42, wherein the effective amount of umbilical-derived cells are suitable for administration in vivo by transplanting, implanting, injecting, fusing, delivering via catheter, or providing as a matrix-cell complex.
CA2530422A 2003-06-27 2004-06-25 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system Expired - Fee Related CA2530422C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US48326403P 2003-06-27 2003-06-27
US60/483,264 2003-06-27
PCT/US2004/020957 WO2005001080A2 (en) 2003-06-27 2004-06-25 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system

Publications (2)

Publication Number Publication Date
CA2530422A1 CA2530422A1 (en) 2005-01-06
CA2530422C true CA2530422C (en) 2016-01-05

Family

ID=38456561

Family Applications (7)

Application Number Title Priority Date Filing Date
CA2530533A Expired - Fee Related CA2530533C (en) 2003-06-27 2004-06-25 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
CA2530732A Expired - Fee Related CA2530732C (en) 2003-06-27 2004-06-25 Regeneration and repair of neural tissue using postpartum-derived cells
CA2530416A Expired - Fee Related CA2530416C (en) 2003-06-27 2004-06-25 Postpartum cells derived from placental tissue, and methods of making and using the same
CA2530255A Expired - Fee Related CA2530255C (en) 2003-06-27 2004-06-25 Soft tissue repair and regeneration using postpartum-derived cells
CA2530422A Expired - Fee Related CA2530422C (en) 2003-06-27 2004-06-25 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
CA2530421A Expired - Fee Related CA2530421C (en) 2003-06-27 2004-06-25 Repair and regeneration of ocular tissue using postpartum-derived cells
CA2530412A Expired - Fee Related CA2530412C (en) 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells

Family Applications Before (4)

Application Number Title Priority Date Filing Date
CA2530533A Expired - Fee Related CA2530533C (en) 2003-06-27 2004-06-25 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
CA2530732A Expired - Fee Related CA2530732C (en) 2003-06-27 2004-06-25 Regeneration and repair of neural tissue using postpartum-derived cells
CA2530416A Expired - Fee Related CA2530416C (en) 2003-06-27 2004-06-25 Postpartum cells derived from placental tissue, and methods of making and using the same
CA2530255A Expired - Fee Related CA2530255C (en) 2003-06-27 2004-06-25 Soft tissue repair and regeneration using postpartum-derived cells

Family Applications After (2)

Application Number Title Priority Date Filing Date
CA2530421A Expired - Fee Related CA2530421C (en) 2003-06-27 2004-06-25 Repair and regeneration of ocular tissue using postpartum-derived cells
CA2530412A Expired - Fee Related CA2530412C (en) 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells

Country Status (8)

Country Link
US (28) US7413734B2 (en)
EP (14) EP2338981B1 (en)
JP (7) JP4948165B2 (en)
AU (7) AU2004252568B2 (en)
CA (7) CA2530533C (en)
ES (14) ES2564044T3 (en)
PL (14) PL2322599T3 (en)
WO (7) WO2005001078A2 (en)

Families Citing this family (535)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080077251A1 (en) * 1999-06-07 2008-03-27 Chen Silvia S Cleaning and devitalization of cartilage
US8563232B2 (en) 2000-09-12 2013-10-22 Lifenet Health Process for devitalizing soft-tissue engineered medical implants, and devitalized soft-tissue medical implants produced
US20020095157A1 (en) * 1999-07-23 2002-07-18 Bowman Steven M. Graft fixation device combination
US6179840B1 (en) 1999-07-23 2001-01-30 Ethicon, Inc. Graft fixation device and method
US8147824B2 (en) 1999-08-05 2012-04-03 Athersys, Inc. Immunomodulatory properties of multipotent adult progenitor cells and uses thereof
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
AU2020902A (en) 2000-12-06 2002-06-18 Robert J Hariri Method of collecting placental stem cells background of the invention
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
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
ES2549161T3 (en) * 2001-02-14 2015-10-23 Anthrogenesis Corporation Postpartum mammalian placenta, its use and placental stem cells thereof
IL157350A0 (en) * 2001-02-14 2004-02-19 Anthrogenesis Corp Post-partum mammalian placenta, its use and placental stem cells therefrom
IL159895A0 (en) * 2001-07-20 2004-06-20 Technion Res & Dev Foundation Methods of generating human cardiac cells and tissues and uses thereof
US7677565B2 (en) 2001-09-28 2010-03-16 Shuffle Master, Inc Card shuffler with card rank and value reading capability
US20080299090A1 (en) * 2002-02-25 2008-12-04 Kansas State University Research Foundation Use Of Umbilical Cord Matrix Cells
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20080003258A1 (en) * 2002-10-16 2008-01-03 Marcum Frank D Composition and Method for Treating Rheumatoid Arthritis
US20040078090A1 (en) 2002-10-18 2004-04-22 Francois Binette Biocompatible scaffolds with tissue fragments
US7824701B2 (en) 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
KR101042448B1 (en) * 2002-11-26 2011-06-16 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
WO2004069172A2 (en) 2003-01-30 2004-08-19 The Government of the United States of America as represented by the Department of Veterans Affairs Multilineage-inducible cells and uses thereof
PT1594957E (en) 2003-02-11 2010-11-30 John E Davies Progenitor cells from wharton`s jelly of human umbilical cord
CA2515594A1 (en) * 2003-02-13 2004-08-26 Anthrogenesis Corporation Use of umbilical cord blood to treat individuals having a disease, disorder or condition
US8197837B2 (en) 2003-03-07 2012-06-12 Depuy Mitek, Inc. Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
WO2004097720A1 (en) * 2003-04-24 2004-11-11 Koninklijke Philips Electronics N.V. Non-invasive left ventricular volume determination
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
US7875273B2 (en) 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum 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
US8491883B2 (en) * 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US7413734B2 (en) 2003-06-27 2008-08-19 Ethicon, Incorporated Treatment of retinitis pigmentosa with human umbilical cord cells
US8226715B2 (en) 2003-06-30 2012-07-24 Depuy Mitek, Inc. Scaffold for connective tissue repair
US10583220B2 (en) 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
DE602004024312D1 (en) * 2003-08-12 2010-01-07 Tigenix Nv USE OF CXCL6 CHEMOKIN IN THE PREVENTION OR REPAIR OF CARTILAGE DEFECTS
US8043614B2 (en) * 2004-03-09 2011-10-25 Ahlfors Jan-Eric W Autogenic living scaffolds and living tissue matrices: methods and uses thereof
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
US7682828B2 (en) 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
US7316822B2 (en) * 2003-11-26 2008-01-08 Ethicon, Inc. Conformable tissue repair implant capable of injection delivery
CN101966183A (en) * 2003-12-02 2011-02-09 细胞基因公司 Methods and compositions for the treatment and management of hemoglobinopathy and anemia
US7901461B2 (en) * 2003-12-05 2011-03-08 Ethicon, Inc. Viable tissue repair implants and methods of use
JP2005168360A (en) * 2003-12-09 2005-06-30 Olympus Corp Method for examining biological tissue-supplying material, device, cell culture container and method for examining culturing state
US20050176139A1 (en) * 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
US20080241107A1 (en) * 2004-01-23 2008-10-02 Copland Iii John A Methods and Compositions For Preparing Pancreatic Insulin Secreting Cells
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
EP2298861B1 (en) 2004-03-22 2017-09-13 Mesoblast International Sàrl Mesenchymal stem cells and uses therefor
US8221780B2 (en) * 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
CA2563518C (en) * 2004-04-23 2014-09-02 Bioe, Inc. Multi-lineage progenitor cells
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
WO2005107807A2 (en) * 2004-05-06 2005-11-17 University Of South Florida Cerebral intraventricular transplantation as method of treating amyotrophic lateral sclerosis
US20060083732A1 (en) * 2004-06-30 2006-04-20 Arlene Gwon Hyaluronic acid in the enhancement of lens regeneration
US8802651B2 (en) * 2004-06-30 2014-08-12 Abbott Medical Optics Inc. Hyaluronic acid in the enhancement of lens regeneration
ES2527293T3 (en) 2004-08-16 2015-01-22 Cellresearch Corporation Pte Ltd Isolation of stem / progenitor cells of amniotic membrane of the umbilical cord
US20060045872A1 (en) * 2004-08-25 2006-03-02 Universidad Autonoma De Madrid Ciudad Universitaria de Cantoblanco Use of adipose tissue-derived stromal stem cells in treating fistula
US8039258B2 (en) * 2004-09-28 2011-10-18 Ethicon, Inc. Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20060069008A1 (en) * 2004-09-28 2006-03-30 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US20080038316A1 (en) * 2004-10-01 2008-02-14 Wong Vernon G Conveniently implantable sustained release drug compositions
US7473678B2 (en) 2004-10-14 2009-01-06 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and methods of use thereof
US7842304B2 (en) * 2004-10-29 2010-11-30 Nexeon Medsystems, Inc. Methods and apparatus for treating an injured nerve pathway
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US8017395B2 (en) 2004-12-17 2011-09-13 Lifescan, Inc. Seeding cells on porous supports
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
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
EP1831355A2 (en) * 2004-12-23 2007-09-12 Ethicon, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
CA2589041C (en) * 2004-12-23 2019-08-20 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
EP1838842A2 (en) * 2004-12-23 2007-10-03 Ethicon, Incorporated Treatment of osteochondral diseases using postpartum-derived cells and products thereof
WO2006088867A2 (en) * 2005-02-15 2006-08-24 Medistem Laboratories, Incorporated Method for expansion of stem cells
WO2006093276A1 (en) * 2005-03-04 2006-09-08 Kyoto University Pluripotent stem cell derived from cardiac tissue
CN101589137B (en) * 2005-03-31 2013-06-05 斯丹姆涅恩有限公司 Amnion-derived cell compositions, methods of making and uses thereof
US20060222634A1 (en) 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US8153430B2 (en) * 2005-03-31 2012-04-10 Stemnion, Inc. Methods related to surgery
WO2006110806A2 (en) * 2005-04-12 2006-10-19 The Trustees Of The University Of Pennsylvania Multipotent adult stem cells
US20100087546A1 (en) * 2005-04-20 2010-04-08 Biogenic Innovations, Llc Use of dimethyl sulfone (msm) to reduce homocysteine levels
WO2006113731A2 (en) * 2005-04-20 2006-10-26 University Of Florida Research Foundation, Inc. Bone marrow-derived neurogenic cells and uses thereof
EP2283851B1 (en) * 2005-04-25 2023-06-07 Massachusetts Institute of Technology Compositions for promoting hemostasis
AU2006202209B2 (en) * 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
AU2006255183B2 (en) 2005-06-08 2012-02-02 Centocor, Inc. A cellular therapy for ocular degeneration
GB0511723D0 (en) * 2005-06-09 2005-07-13 Smith & Nephew Placental stem cells
WO2006135899A2 (en) * 2005-06-13 2006-12-21 The Johns Hopkins University Survival, differentiation and structural intergration of human neural stem cells grafted into the adult spinal cord
US7838503B2 (en) * 2005-06-15 2010-11-23 Children's Medical Center Corporation Methods for extending the replicative lifespan of cells
US20150119794A1 (en) * 2005-08-05 2015-04-30 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
US20140336514A1 (en) * 2005-08-05 2014-11-13 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
CA2622204C (en) 2005-09-12 2016-10-11 Abela Pharmaceuticals, Inc. Systems for removing dimethyl sulfoxide (dmso) or related compounds, or odors associated with same
US8480797B2 (en) 2005-09-12 2013-07-09 Abela Pharmaceuticals, Inc. Activated carbon systems for facilitating use of dimethyl sulfoxide (DMSO) by removal of same, related compounds, or associated odors
EP1937286B1 (en) 2005-09-12 2016-03-09 Abela Pharmaceuticals, Inc. Compositions comprising dimethyl sulfoxide (dmso)
US8435224B2 (en) 2005-09-12 2013-05-07 Abela Pharmaceuticals, Inc. Materials for facilitating administration of dimethyl sulfoxide (DMSO) and related compounds
JP2007106760A (en) * 2005-09-16 2007-04-26 Kenji Yoshida Hematopoietic stem cell proliferator
EP1933852B1 (en) 2005-09-27 2018-12-19 TissueTech, Inc. Amniotic membrane preparations and purified compositions and methods of use
CN104138392A (en) * 2005-10-13 2014-11-12 人类起源公司 Immunomodulation using placental stem cells
KR20080056302A (en) * 2005-10-13 2008-06-20 안트로제네시스 코포레이션 Production of oligodendrocytes from placenta-derived stem cells
JP4921767B2 (en) * 2005-10-14 2012-04-25 株式会社カネカ Cell differentiation induction method
US11000546B2 (en) 2005-11-09 2021-05-11 Athersys, Inc. Immunomodulatory properties of MAPCs and uses thereof
US20070178067A1 (en) * 2005-11-09 2007-08-02 John Maier System and method for cytological analysis by raman spectroscopic imaging
US10117900B2 (en) * 2005-11-09 2018-11-06 Athersys, Inc. MAPC treatment of brain injuries and diseases
US20090170059A1 (en) * 2005-11-14 2009-07-02 Hans Klingemann Methods for Preparing Cord Matrix Stem Cells (CMSC) for Long Term Storage and for Preparing a Segment of umbilical cord for cryopreservation
WO2007061889A2 (en) * 2005-11-17 2007-05-31 Biomimetic Therapeutics, Inc. Maxillofacial bone augmentation using rhpdgf-bb and a biocompatible matrix
PL1971681T3 (en) * 2005-12-16 2018-01-31 Depuy Synthes Products Inc Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
AU2006327073B2 (en) * 2005-12-19 2012-08-30 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
SG10201401805YA (en) 2005-12-22 2014-08-28 Jane Ennis Viable cells from frozen umbilical cord tissue
AU2006330409B2 (en) * 2005-12-28 2012-07-19 Ethicon, Incorporated Treatment of peripheral vascular disease using postpartum-derived cells
CN105106239A (en) * 2005-12-28 2015-12-02 伊西康公司 Treatment of peripheral vascular disease using postpartum-derived cells
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
CA2633775A1 (en) 2005-12-29 2007-07-12 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
EP2471907B1 (en) * 2005-12-29 2018-09-26 Celularity, Inc. Placental stem cell populations
WO2007080590A2 (en) 2006-01-11 2007-07-19 Technion Research & Development Foundation Ltd. Human embryonic stem cell-derived connective tissue progenitors for tissue engineering
US20080286249A1 (en) * 2006-01-12 2008-11-20 Varney Timothy R Use of mesenchymal stem cells for treating genetic diseases and disorders
US20070253931A1 (en) * 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
US8871198B2 (en) * 2006-03-29 2014-10-28 Stemnion, Inc. Methods related to wound healing
US9944900B2 (en) * 2006-01-18 2018-04-17 Hemacell Perfusion Pulsatile perfusion extraction method for non-embryonic pluripotent stem cells
GB0600972D0 (en) * 2006-01-18 2006-03-01 Univ Leeds Enrichment of cells
US7875451B2 (en) * 2006-01-19 2011-01-25 The University Of Washington Formulation to improve survival of transplanted cells
JP5986346B2 (en) 2006-01-23 2016-09-06 アサーシス,インコーポレーテッド MAPC therapy without adjuvant immunosuppressive treatment
CA2640185A1 (en) * 2006-01-24 2007-08-02 Christopher J. Centeno Mesenchymal stem cell isolation and transplantation method and system to be used in a clinical setting
FR2896511B1 (en) * 2006-01-26 2012-10-26 Centre Nat Rech Scient PROCESS FOR CULTIVATION OF CELLS FROM ADIPOSE TISSUE AND THEIR APPLICATIONS
EP2311505B1 (en) * 2006-02-09 2013-11-06 BioMimetic Therapeutics, LLC Compositions and methods for treating bone
WO2007099534A2 (en) * 2006-03-01 2007-09-07 The Regenerative Medicine Institute Compostions and populations of cells obtained from the umbilical cord and methods of producing the same
EP1996698A1 (en) * 2006-03-01 2008-12-03 Cartela R&D AB Expansion and differentiation of mesenchymal stem cells
US20110171182A1 (en) * 2006-03-23 2011-07-14 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
ES2549528T3 (en) * 2006-03-23 2015-10-29 Pluristem Ltd. Methods of cell expansion and uses of cells and conditioning media produced by them for therapy
EP2019858B1 (en) * 2006-04-17 2012-06-13 BioE LLC Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
WO2007124023A2 (en) * 2006-04-21 2007-11-01 Wake Forest University Health Sciences Ink-jet printing of tissues
US9456979B2 (en) * 2006-04-27 2016-10-04 Sri International Adminstration of intact mammalian cells to the brain by the intranasal route
WO2007127873A2 (en) 2006-04-27 2007-11-08 Advanced Medical Optics, Inc. Enhancement of lens regeneration using materials comprising polysiloxane polymers
US8741643B2 (en) * 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
KR101507174B1 (en) 2006-05-05 2015-04-01 존 이. 다비스 Immune privileged and modulatory progenitor cells
WO2007133665A2 (en) * 2006-05-11 2007-11-22 Naoko Takebe Methods for collecting and using placenta cord blood stem cells
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
WO2007146105A2 (en) * 2006-06-05 2007-12-21 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
CA2654716A1 (en) * 2006-06-09 2007-12-21 Anthrogenesis Corporation Placental niche and use thereof to culture stem cells
WO2007145889A1 (en) * 2006-06-14 2007-12-21 Stemnion, Inc. Methods of treating spinal cord injury and minimizing scarring
US8475788B2 (en) * 2006-06-14 2013-07-02 Stemnion, Inc. Methods of treating spinal cord injury and minimizing scarring
US20070292401A1 (en) * 2006-06-20 2007-12-20 Harmon Alexander M Soft tissue repair and regeneration using stem cell products
US20080019949A1 (en) * 2006-06-28 2008-01-24 University Of Kansas Differentiation of stem cells from umbilical cord matrix into hepatocyte lineage cells
AU2007269712B2 (en) 2006-06-30 2013-02-07 Biomimetic Therapeutics, Llc PDGF-biomatrix compositions and methods for treating rotator cuff injuries
US9161967B2 (en) 2006-06-30 2015-10-20 Biomimetic Therapeutics, Llc Compositions and methods for treating the vertebral column
CN101511307A (en) * 2006-07-24 2009-08-19 国际干细胞公司 Synthetic cornea from retinal stem cells
US7993918B2 (en) * 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US7736485B2 (en) * 2006-08-11 2010-06-15 Aqua Resources Corporation Nanoplatelet magnesium hydroxides and methods of preparing same
US8822030B2 (en) 2006-08-11 2014-09-02 Aqua Resources Corporation Nanoplatelet metal hydroxides and methods of preparing same
US7671014B2 (en) * 2006-08-14 2010-03-02 Warsaw Orthopedic, Inc. Flowable carrier matrix and methods for delivering to a patient
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
JP5656183B2 (en) * 2006-08-22 2015-01-21 国立大学法人 東京医科歯科大学 Application of synovial mesenchymal stem cells (MSCs) to cartilage and meniscal regeneration
US20080082170A1 (en) * 2006-09-29 2008-04-03 Peterman Marc M Apparatus and methods for surgical repair
US20080078412A1 (en) * 2006-10-03 2008-04-03 Restore Medical, Inc. Tongue implant
US20080078411A1 (en) * 2006-10-03 2008-04-03 Restore Medical, Inc. Tongue implant for sleep apnea
SI2078073T1 (en) 2006-10-12 2013-11-29 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
EP1913869A3 (en) * 2006-10-19 2008-12-10 Esaote S.p.A. Diagnostic imaging method and apparatus for the anatomical region of the pelvic floor
US8562972B2 (en) 2006-10-23 2013-10-22 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
EP2086598B1 (en) * 2006-11-03 2015-05-27 BioMimetic Therapeutics, LLC Compositions and methods for arthrodetic procedures
EP2089511B1 (en) * 2006-11-13 2014-09-17 DePuy Synthes Products, LLC In vitro expansion of postpartum-derived cells using microcarriers
US20080132803A1 (en) * 2006-11-30 2008-06-05 Hyman Friedlander Method and system for doing business by mining the placental-chord complex
AU2007332799A1 (en) * 2006-12-07 2008-06-19 Teva Pharmaceutical Industries Ltd. Method of generation and expansion of tissue-progenitor cells and mature tissue cells from intact bone marrow or intact umbilical cord tissue
US20100143289A1 (en) * 2006-12-19 2010-06-10 Michael Cohen Umbilical cord stem cell secreted product derived topical compositions and methods of use thereof
US20100303770A1 (en) * 2006-12-28 2010-12-02 John Maslowski Methods for culturing dermal cells for treatment of skin injuries such as burns
US7980000B2 (en) * 2006-12-29 2011-07-19 Applied Materials, Inc. Vapor dryer having hydrophilic end effector
EP2118267B1 (en) * 2007-01-17 2017-03-15 Noveome Biotherapeutics, Inc. Novel methods for modulating inflammatory and/or immune responses
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
EP2915537A3 (en) * 2007-02-12 2015-10-28 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
KR20090109127A (en) * 2007-02-12 2009-10-19 안트로제네시스 코포레이션 Hepatocytes and chondrocytes from adherent placental stem cells? and cd34?, cd45? placental stem cell-enriched cell populations
AU2008218763B2 (en) * 2007-02-20 2013-10-24 Biomimetic Therapeutics, Llc. Prevention and treatment for osteonecrosis and osteoradionecrosis of the jaw using PDGF and a bone matrix
JP2010530214A (en) * 2007-03-01 2010-09-09 クライオ−セル インターナショナル インコーポレイテッド Acquisition, isolation and cryopreservation of endometrial / menstrual cells
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
EP2626416A3 (en) * 2007-04-07 2013-12-18 The Whitehead Institute for Biomedical Research Reprogramming of somatic cells
WO2009008928A2 (en) * 2007-04-13 2009-01-15 Stemnion, Inc. Methods for treating nervous system injury and disease
EP2137300B1 (en) 2007-04-26 2011-10-26 Ramot at Tel-Aviv University Ltd. Pluripotent autologous stem cells from oral or gastrointestinal mucosa
EP2607477B1 (en) 2007-05-03 2020-09-23 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US8574567B2 (en) 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
US8114668B2 (en) * 2007-05-14 2012-02-14 Cardiac Pacemakers, Inc. Composition for cold storage of stem cells
US20100111914A1 (en) * 2007-05-21 2010-05-06 Yuanyuan Zhang Stem cells from urine and methods for using the same
WO2008153685A2 (en) 2007-05-21 2008-12-18 Wake Forest University Health Sciences Progenitor cells from urine and methods for using the same
TWM322542U (en) * 2007-05-23 2007-11-21 Universal Scient Ind Co Ltd Testing machine
US20090053182A1 (en) * 2007-05-25 2009-02-26 Medistem Laboratories, Inc. Endometrial stem cells and methods of making and using same
ATE487441T1 (en) * 2007-06-01 2010-11-15 Allergan Inc DEVICE FOR GENERATING TENSILE-INDUCED GROWTH OF BIOLOGICAL TISSUE
US9693486B1 (en) * 2007-06-14 2017-06-27 Switch, Ltd. Air handling unit with a canopy thereover for use with a data center and method of using the same
CA2688504A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US20090004253A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Composite device for the repair or regeneration of tissue
US20090004271A1 (en) 2007-06-29 2009-01-01 Brown Laura J Morselized foam for wound treatment
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US9095562B2 (en) * 2007-07-05 2015-08-04 Regenerative Sciences, Inc. Methods and compositions for optimized expansion and implantation of mesenchymal stem cells
DE102007034679A1 (en) * 2007-07-25 2009-01-29 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Material compositions containing adult stem cells derived from exocrine glandular tissue, especially for use in regenerative medicine, e.g. to restore injured or damaged myocardial tissue
US9744043B2 (en) 2007-07-16 2017-08-29 Lifenet Health Crafting of cartilage
CA2693827A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of multi-lineage progenitor cells to chondrocytes
CA2694700C (en) 2007-07-27 2015-04-14 Humacyte, Inc. Compositions and methods for soft tissue augmentation
RU2473685C2 (en) 2007-07-31 2013-01-27 Лайфскен, Инк. Differentiation of human embryo stem cells
US20110070282A1 (en) * 2007-08-08 2011-03-24 Pervasis Therapeutics, Inc. Materials and Methods for Treating Skeletal System Damage and Promoting Skeletal System Repair and Regeneration
US20090062907A1 (en) * 2007-08-31 2009-03-05 Quijano Rodolfo C Self-expanding valve for the venous system
US20090068153A1 (en) * 2007-09-06 2009-03-12 Vitelli Francesca P Cell composition for tissue regeneration
EP3189731B1 (en) 2007-09-07 2020-01-29 MiMedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
JP2010539228A (en) * 2007-09-19 2010-12-16 プルリステム リミテッド Adherent cells derived from adipose tissue or placental tissue and their use in therapy
WO2009042768A1 (en) * 2007-09-25 2009-04-02 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Triggerably dissolvable hollow fibers for controlled delivery
MX2010003217A (en) * 2007-09-26 2010-07-30 Celgene Cellular Therapeutics Angiogenic cells from human placental perfusate.
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
BRPI0818183A2 (en) * 2007-10-05 2019-07-16 Ethicon Inc Renal tissue repair and regeneration using cells derived from human umbilical cord tissue
WO2009052209A2 (en) * 2007-10-16 2009-04-23 University Of Kansas Isolation of stem cells and effective control of contamination
US20100297234A1 (en) * 2007-10-19 2010-11-25 Ilene Sugino Method of using an extracellular matrix to enhance cell transplant survival and differentiation
US20120219737A1 (en) 2007-10-19 2012-08-30 University Of Medicine And Dentistry Of New Jersey Production of extracellular matrix, conditioned media and uses thereof
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
US20110086080A1 (en) * 2007-11-14 2011-04-14 Osteosphere, Llc Ex-vivo production of human demineralized bone matrix
WO2009070592A2 (en) * 2007-11-27 2009-06-04 Lifescan, Inc. Differentiation of human embryonic stem cells
WO2009085969A2 (en) * 2007-12-19 2009-07-09 Regenerative Sciences, Llc Compositions and methods to promote implantation and engrafment of stem cells
US20090163990A1 (en) * 2007-12-19 2009-06-25 Chunlin Yang Decellularized omentum matrix and uses thereof
WO2009085216A2 (en) 2007-12-20 2009-07-09 Squicor Compositions and methods for detecting or elimninating senescent cells to diagnose or treat disease
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
AU2008343203B2 (en) * 2007-12-27 2014-07-10 DePuy Synthes Products, LLC Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
US20100279413A1 (en) * 2008-01-14 2010-11-04 Zymes, Llc Applications of ubiquinones and ubiquinols
US8088163B1 (en) 2008-02-06 2012-01-03 Kleiner Jeffrey B Tools and methods for spinal fusion
JP5864106B2 (en) 2008-02-07 2016-02-17 バイオミメティック セラピューティクス, エルエルシー Compositions and methods for callus extension
EP3095470A1 (en) * 2008-02-07 2016-11-23 Shahar Cohen Compartmental extract compositions for tissue engineering
US20090209456A1 (en) * 2008-02-19 2009-08-20 Iliana Sweis Compositions and methods for improving facial and body aesthetics
BRPI0908033A2 (en) 2008-02-21 2015-08-04 Centocor Ortho Biotech Inc Surface modified plate method and compositions for cell adhesion, culture and detachment
US8318485B2 (en) * 2008-02-25 2012-11-27 Natalie Gavrilova Stem cell therapy for the treatment of diabetic retinopathy and diabetic optic neuropathy
US20090220995A1 (en) 2008-02-28 2009-09-03 Sachs David H Multiple administrations of umbilicus derived cells
US20090232782A1 (en) * 2008-03-14 2009-09-17 Yu-Show Fu Method for treating brain ischemic injury through transplantation of human umbilical mesenchymal stem cells
WO2009114785A2 (en) * 2008-03-14 2009-09-17 Regenerative Sciences, Inc. Compositions and methods for cartilage repair
US9005599B2 (en) 2008-04-21 2015-04-14 Tissue Regeneration Therapeutics Inc. Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US8623648B2 (en) * 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US9480549B2 (en) 2008-04-25 2016-11-01 Allosource Multi-layer tissue patches
US9358320B2 (en) 2008-04-25 2016-06-07 Allosource Multi-layer tissue patches
CN102076366B (en) * 2008-04-30 2014-06-11 伊西康公司 Tissue engineered blood vessel
US20110143429A1 (en) * 2008-04-30 2011-06-16 Iksoo Chun Tissue engineered blood vessels
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
US8748181B2 (en) * 2008-05-23 2014-06-10 President And Fellows Of Harvard College Methods of generating patterned soft substrates and uses thereof
US20090297485A1 (en) * 2008-05-28 2009-12-03 Allan Mishra Compositions and methods for treating psychiatric and neurodegenerative disorders
WO2009152384A1 (en) * 2008-06-11 2009-12-17 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
CA3060170A1 (en) 2008-06-13 2009-12-17 Whitehead Institute For Biomedical Research Programming and reprogramming of cells
CN103655040A (en) * 2008-06-26 2014-03-26 凯希特许有限公司 Stimulation of cartilage formation using reduced pressure treatment
JP5294041B2 (en) * 2008-06-27 2013-09-18 独立行政法人産業技術総合研究所 Pancreatic cell regenerative transplant kit for pancreatic disease or diabetes
JP2011526784A (en) * 2008-06-30 2011-10-20 セントコア・オーソ・バイオテツク・インコーポレーテツド Differentiation of pluripotent stem cells
CN102159703B (en) 2008-06-30 2015-11-25 森托科尔奥索生物科技公司 The differentiation of multipotential stem cell
US20100028307A1 (en) * 2008-07-31 2010-02-04 O'neil John J Pluripotent stem cell differentiation
WO2010021714A2 (en) 2008-08-20 2010-02-25 Anthrogenesis Corporation Improved cell composition and methods of making the same
JP5950577B2 (en) * 2008-08-20 2016-07-13 アンスロジェネシス コーポレーション Treatment of stroke using isolated placental cells
RU2011110736A (en) 2008-08-22 2012-09-27 Антродженезис Корпорейшн (Us) METHODS AND COMPOSITIONS FOR TREATING BONE DEFECTS USING PLACENTAL CELLULAR POPULATIONS
US8524496B2 (en) 2008-09-02 2013-09-03 Pluristem Ltd. Adherent cells from placenta tissue and use thereof in therapy
JP5816553B2 (en) 2008-09-09 2015-11-18 バイオミメティック セラピューティクス, エルエルシー Platelet-derived growth factor compositions and methods for treating tendon or ligament injury
US20100069827A1 (en) * 2008-09-12 2010-03-18 Barry Neil Silberg Pre-Surgical Prophylactic Administration of Antibiotics and Therapeutic Agents
US9446227B2 (en) 2008-09-12 2016-09-20 Sonescence, Inc. Ultrasonic dispersion of compositions in tissue
RU2011118341A (en) 2008-10-06 2012-11-20 3-Д Матрикс, Лтд. (Jp) AGENT FOR OCCLUSION OF TISSUES
WO2010048418A1 (en) * 2008-10-22 2010-04-29 The Trustees Of Columbia University In The City Of New York Cartilage regeneration without cell transplantation
KR101712085B1 (en) * 2008-10-31 2017-03-03 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells to the pancreatic endocrine lineage
KR20110097785A (en) * 2008-10-31 2011-08-31 신세스 게엠바하 Method and device for activating stem cells
CN107435038B (en) * 2008-10-31 2021-07-09 詹森生物科技公司 Differentiation of human embryonic stem cells into the pancreatic endocrine lineage
BRPI0921494A2 (en) 2008-11-03 2018-10-30 Prad Reasearch And Development Ltd method of planning a underground forming sampling operation, method of controlling a underground forming sampling operation, method of controlling a drilling operation for an underground formation, and method of sampling during the drilling operation.
MX2011005229A (en) 2008-11-19 2011-06-16 Anthrogenesis Corp Amnion derived adherent cells.
AU2009316580B2 (en) 2008-11-20 2016-04-14 Janssen Biotech, Inc. Pluripotent stem cell culture on micro-carriers
CA2744227C (en) * 2008-11-20 2018-10-02 Centocor Ortho Biotech Inc. Methods and compositions for cell attachment and cultivation on planar substrates
CN102215788A (en) * 2008-11-20 2011-10-12 生命细胞公司 Method for treatment and prevention of parastomal hernias
EP2375907B1 (en) * 2008-11-21 2019-02-27 Celularity, Inc. Treatment of diseases, disorders or conditions of the lung using placental cells
US8366748B2 (en) 2008-12-05 2013-02-05 Kleiner Jeffrey Apparatus and method of spinal implant and fusion
JP2012510874A (en) 2008-12-05 2012-05-17 リジェネレイティブ サイエンシーズ, エルエルシー Methods and compositions for promoting repair of avascular tissue
JPWO2010064702A1 (en) * 2008-12-05 2012-05-10 国立大学法人 東京大学 Biomarkers for predicting cancer prognosis
US20100168022A1 (en) * 2008-12-11 2010-07-01 Centeno Christopher J Use of In-Vitro Culture to Design or Test Personalized Treatment Regimens
US20130302283A1 (en) 2012-05-14 2013-11-14 Advanced Technologies And Regenerative Medicine, Llc hUTC MODULATION OF PRO-INFLAMMATORY MEDIATORS 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
AU2009327382B2 (en) * 2008-12-19 2014-07-17 DePuy Synthes Products, LLC Umbilical cord tissue derived cells for treating neuropathic pain and spasticity
AU2009327383B2 (en) * 2008-12-19 2014-08-28 DePuy Synthes Products, LLC Regeneration and repair of neural tissue following injury
EP2379088B1 (en) * 2008-12-19 2018-02-28 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
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
AU2009333066B2 (en) * 2008-12-30 2016-02-18 Solventum Intellectual Properties Company Reduced pressure augmentation of microfracture procedures for cartilage repair
KR101632756B1 (en) * 2009-01-23 2016-06-22 고쿠리츠다이가쿠호우진 도쿄다이가쿠 Method for assessing cultured cell
US9247943B1 (en) 2009-02-06 2016-02-02 Kleiner Intellectual Property, Llc Devices and methods for preparing an intervertebral workspace
US20120142603A1 (en) * 2009-02-12 2012-06-07 University Of Southern California Bioadhesive patch for sutureless closure of soft tissue
KR101422690B1 (en) * 2009-02-27 2014-07-23 (주)차바이오앤디오스텍 Composition for skin regeneration by using medium or secretion of embryonic stem cell derived endothelial progenitor cells and use thereof
JP5908394B2 (en) * 2009-03-26 2016-04-26 デピュイ・シンセス・プロダクツ・インコーポレイテッド Human umbilical cord tissue cells as a therapy for Alzheimer's disease
EP2411505A4 (en) * 2009-03-26 2013-01-30 Univ California Mesenchymal stem cells producing inhibitory rna for disease modification
WO2010118059A1 (en) * 2009-04-06 2010-10-14 Capricor, Inc. Systems and methods for cardiac tissue repair
MX357440B (en) * 2009-05-13 2018-07-10 Medipost Co Ltd Tsp-1, tsp-2, il-17br and hb-egf associated with stem cell activities and applications thereof.
AU2010249805B2 (en) * 2009-05-20 2015-06-11 Humacyte, Inc. Elastin for soft tissue augmentation
WO2010138782A1 (en) * 2009-05-28 2010-12-02 University Of Central Florida Research In vitro production of oligodendrocytes from human umbilical cord stem cells
US20150335400A1 (en) * 2009-06-17 2015-11-26 The Trustees Of Columbia University In The City Of New York Tooth scaffolds
MX340952B (en) * 2009-07-20 2016-07-29 Janssen Biotech Inc Differentiation of human embryonic stem cells.
RU2540016C2 (en) * 2009-07-20 2015-01-27 Янссен Байотек, Инк. Differentiating human embryonic stem cells
WO2011011300A2 (en) 2009-07-20 2011-01-27 Centocor Ortho Biotech Inc. Differentiation of human embryonic stem cells
CN102625829B (en) * 2009-07-21 2015-05-06 Abt控股公司 Use of stem cells to reduce leukocyte extravasation
ES2360434B1 (en) 2009-07-21 2012-04-12 Universitat Internacional De Catalunya PLURIPOTENTIAL MOTHER CELLS OBTAINED FROM THE DENTAL PULP.
WO2011022070A2 (en) * 2009-08-20 2011-02-24 The Regents Of The University Of California Perivascular stem cell composition for bone
US20110054929A1 (en) * 2009-09-01 2011-03-03 Cell Solutions Colorado Llc Stem Cell Marketplace
US8207651B2 (en) 2009-09-16 2012-06-26 Tyco Healthcare Group Lp Low energy or minimum disturbance method for measuring frequency response functions of ultrasonic surgical devices in determining optimum operating point
USD750249S1 (en) 2014-10-20 2016-02-23 Spinal Surgical Strategies, Llc Expandable fusion cage
US20170238984A1 (en) 2009-09-18 2017-08-24 Spinal Surgical Strategies, Llc Bone graft delivery device with positioning handle
US10245159B1 (en) 2009-09-18 2019-04-02 Spinal Surgical Strategies, Llc Bone graft delivery system and method for using same
US9173694B2 (en) 2009-09-18 2015-11-03 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US8906028B2 (en) 2009-09-18 2014-12-09 Spinal Surgical Strategies, Llc Bone graft delivery device and method of using the same
US8685031B2 (en) 2009-09-18 2014-04-01 Spinal Surgical Strategies, Llc Bone graft delivery system
US9629729B2 (en) 2009-09-18 2017-04-25 Spinal Surgical Strategies, Llc Biological delivery system with adaptable fusion cage interface
USD723682S1 (en) 2013-05-03 2015-03-03 Spinal Surgical Strategies, Llc Bone graft delivery tool
US10973656B2 (en) 2009-09-18 2021-04-13 Spinal Surgical Strategies, Inc. Bone graft delivery system and method for using same
US9060877B2 (en) 2009-09-18 2015-06-23 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US9186193B2 (en) 2009-09-18 2015-11-17 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
WO2011038133A2 (en) * 2009-09-23 2011-03-31 Davinci Biosciences Llc Umbilical cord lining stem cells and methods and material for isolating and culturing same
EP2488209B1 (en) 2009-10-16 2016-09-28 Rutgers, the State University of New Jersey Method for treating chronic nerve tissue injury using a cell therapy strategy
BR112012010136A2 (en) * 2009-10-29 2015-09-15 Janssen Biotech Inc pluripotent stem cells
KR20120093993A (en) 2009-10-30 2012-08-23 아벨라 파마슈티칼스, 인코포레이티드 Dimethyl sulfoxide (dmso) and methylsulfonylmethane (msm) formulations to treat osteoarthritis
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
WO2011060079A1 (en) * 2009-11-10 2011-05-19 The Trustees Of Columbia University In The City Of New York Compositions and methods for wound treatment
US20130129686A1 (en) * 2009-11-19 2013-05-23 Regents Of The University Reducing Inflammation Using Cell Therapy
RU2586506C2 (en) * 2009-12-23 2016-06-10 Янссен Байотек, Инк. Differentiation of human embryonic stem cells
SG10201408552YA (en) 2009-12-23 2015-02-27 Janssen Biotech Inc Differentiation of human embryonic stem cells
EA201290577A1 (en) 2010-01-26 2013-06-28 Стем Селз Спин С.А. CELL HOMOGENATES FROM STEM CELLS OBTAINED FROM GROWING REIT HORNS, METHOD FOR ITS OBTAINING AND APPLICATION
US9121007B2 (en) * 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
EP3351622B1 (en) * 2010-02-18 2022-01-19 Osiris Therapeutics, Inc. Methods of manufacture of immunocompatible chorionic membrane products
JP5701908B2 (en) * 2010-02-19 2015-04-15 ライフセル コーポレーションLifeCell Corporation Abdominal wall treatment tool
MX2012009687A (en) 2010-02-22 2012-11-29 Biomimetic Therapeutics Inc Platelet-derived growth factor compositions and methods for the treatment of tendinopathies.
RU2607380C2 (en) 2010-03-01 2017-01-10 Янссен Байотек, Инк. Methods of purifying cells, derived from pluripotent stem cells
CN102933703A (en) 2010-04-07 2013-02-13 人类起源公司 Angiogenesis using placental stem cells
KR20130092394A (en) 2010-04-08 2013-08-20 안트로제네시스 코포레이션 Treatment of sarcoidosis using placental stem cells
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US8529883B2 (en) 2010-05-07 2013-09-10 Fibrocell Technologies, Inc. Dosage unit formulations of autologous dermal fibroblasts
SG185511A1 (en) 2010-05-12 2012-12-28 Centocor Ortho Biotech Inc Differentiation of human embryonic stem cells
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
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
WO2011153236A1 (en) * 2010-06-03 2011-12-08 The Board Of Trustees Of The Leland Stanford Junior University Purified compositions of cardiovascular progenitor cells
CN103249404A (en) 2010-07-02 2013-08-14 北卡罗来纳-查佩尔山大学 Biomatrix scaffolds
US8808687B2 (en) 2010-07-12 2014-08-19 Mark Humayun Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
CN103097520B (en) 2010-07-13 2017-12-05 人类起源公司 The method for producing NK
US8825388B2 (en) 2010-07-13 2014-09-02 Qualcomm Incorporated Indoor likelihood heatmap
JP2012031127A (en) * 2010-08-03 2012-02-16 Nagoya Univ Composition including umbilical cord matrix stromal cell
SG187667A1 (en) * 2010-08-03 2013-03-28 Agency Science Tech & Res Microfabricated scaffold structures
IL207586A0 (en) 2010-08-12 2010-12-30 Omrix Biopharmaceuticals Ltd A fibrin based therapeutic preparation and use thereof
WO2012021885A1 (en) * 2010-08-13 2012-02-16 The Trustees Of Columbia University In The City Of New York Three-dimensional tissue engineering devices and uses thereof
CA2809272A1 (en) * 2010-08-31 2012-03-08 Cook General Biotechnology Llc Systemic, allogenic stem cell therapies for treatment of diseases in animals
KR101836855B1 (en) 2010-08-31 2018-04-19 얀센 바이오테크 인코포레이티드 Differentiation of pluripotent stem cells
EP3372672A1 (en) 2010-08-31 2018-09-12 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
KR101836850B1 (en) 2010-08-31 2018-03-09 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
US9677042B2 (en) 2010-10-08 2017-06-13 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US8546338B2 (en) 2010-12-08 2013-10-01 Johnson & Johnson Consumer Companies, Inc. Self-assembling hydrogels based on dicephalic peptide amphiphiles
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
JP5388233B2 (en) * 2011-01-19 2014-01-15 富士ソフト株式会社 Method for evaluating the cartilage characteristics of regenerated cartilage
US8945536B2 (en) * 2011-01-26 2015-02-03 The Chinese University Of Hong Kong Stem cell sheet for tissue repair
US9044430B2 (en) 2011-03-18 2015-06-02 Microvascular Tissues, Inc. Allogeneic microvascular tissue for soft tissue treatments
KR101835917B1 (en) 2011-03-22 2018-03-07 플루리스템 리미티드 Methods for treating radiation or chemical injury
US9758395B2 (en) 2011-04-28 2017-09-12 Aquero Company, Llc Lysine-based polymer coagulants for use in clarification of process waters
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
WO2012149468A2 (en) 2011-04-29 2012-11-01 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
AU2012262311B2 (en) * 2011-05-31 2016-04-28 Lifecell Corporation Adipose tissue matrices
PT2714059T (en) 2011-06-01 2019-02-04 Celularity Inc Treatment of pain using placental stem cells
EP2717888B1 (en) 2011-06-10 2020-09-09 Tissuetech, Inc. Methods of processing fetal support tissues
US20140189897A1 (en) 2011-06-21 2014-07-03 Mayo Foundation For Medical Education And Research Transgenic animals capable of being induced to delete senescent cells
AU2012275335B2 (en) 2011-06-29 2017-04-20 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
US20130005829A1 (en) 2011-06-30 2013-01-03 Advanced Technologies And Regenerative Medicine, Llc. Segmented, epsilon-Caprolactone-Rich, Poly(epsilon-Caprolactone-co-p-Dioxanone) Copolymers for Medical Applications and Devices Therefrom
WO2013010045A1 (en) 2011-07-12 2013-01-17 Biotime Inc. Novel methods and formulations for orthopedic cell therapy
KR101851925B1 (en) 2011-08-10 2018-04-26 디퍼이 신테스 프로덕츠, 인코포레이티드 Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
KR20140119023A (en) 2011-12-13 2014-10-08 버크 인스티튜트 포 리서치 온 에이징 Methods for improving medical therapies
US9162011B2 (en) 2011-12-19 2015-10-20 Allosource Flowable matrix compositions and methods
AU2012355463C1 (en) 2011-12-20 2016-09-22 Lifecell Corporation Sheet tissue products
US20130157365A1 (en) * 2011-12-20 2013-06-20 Advanced Technologies And Regenerative Medicine, Llc Induced pluripotent stem cells from human umbilical cord tissue-derived cells
ES2860464T3 (en) 2011-12-20 2021-10-05 Lifecell Corp Fluidizable Tissue Products
JP6441080B2 (en) 2011-12-22 2018-12-19 ヤンセン バイオテツク,インコーポレーテツド Differentiation of human embryonic stem cells into single hormone insulin-positive cells
WO2013096686A1 (en) 2011-12-23 2013-06-27 Advanced Technologies And Regenerative Medicine, Llc Detection of human umbilical cord tissue-derived cells
ES2890126T3 (en) 2011-12-30 2022-01-17 Amit Patel Methods and compositions for the clinical obtaining of an allogeneic cell and therapeutic uses
CA2861048C (en) 2012-01-24 2021-01-12 Lifecell Corporation Elongated tissue matrices
EP2816894B1 (en) * 2012-02-23 2018-01-03 Anthrogenesis Corporation Identification of antitumor compounds using placenta
US8940294B2 (en) 2012-03-02 2015-01-27 Tissuetech, Inc. Methods of isolating and culturing stem cells
US9434920B2 (en) 2012-03-07 2016-09-06 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US20150064137A1 (en) 2012-04-17 2015-03-05 Kythera Biopharmaceuticals, Inc. Use of engineered viruses to specifically kill senescent cells
ES2749178T3 (en) 2012-04-24 2020-03-19 Lifecell Corp Functionalized tissue matrices
EP2861238A4 (en) 2012-06-05 2016-03-16 Capricor Inc Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
CN108103006A (en) 2012-06-08 2018-06-01 詹森生物科技公司 Differentiation of the human embryo stem cell to pancreatic endocrine cell
EP3466964A1 (en) 2012-07-06 2019-04-10 3-D Matrix Ltd. Fill-finish process for peptide solutions
JP6282270B2 (en) * 2012-07-09 2018-02-21 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Neutral protease derived from recombinant Paenibacillus polymixer
WO2014011402A1 (en) 2012-07-13 2014-01-16 Lifecell Corporation Methods for improved treatment of adipose tissue
US8960674B2 (en) 2012-07-27 2015-02-24 Bally Gaming, Inc. Batch card shuffling apparatuses including multi-card storage compartments, and related methods
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9901081B2 (en) 2012-08-23 2018-02-27 Buck Institute For Research On Aging Transgenic mouse for determining the role of senescent cells in cancer
US9901080B2 (en) 2012-08-23 2018-02-27 Buck Institute For Research On Aging Transgenic mouse having a transgene that converts a prodrug into a cytotoxic compound in senescent cells
EP3476410B1 (en) 2012-09-19 2021-08-18 MicroVascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US10596202B2 (en) 2012-09-19 2020-03-24 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US9872937B2 (en) 2012-09-19 2018-01-23 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US11819522B2 (en) 2012-09-19 2023-11-21 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
BR112015006393A2 (en) 2012-09-26 2017-07-04 Lifecell Corp method for producing a fabric product, fabric product produced by a process, and treatment method
KR101728808B1 (en) * 2012-09-28 2017-04-20 한국생명공학연구원 Pharmaceutical composition for preventing or treating muscle weakness diseases comprising Acecainide or derivatives thereof
ITTO20120859A1 (en) * 2012-10-02 2014-04-03 Univ Degli Studi Torino NEW THERAPEUTIC APPLICATION OF A MEANS CONDITIONED BY PLACENTARY STEM CELLS
US20150284689A1 (en) * 2012-10-26 2015-10-08 The Regents Of The University Of California Strategy for engineering various 3d tissues, organoids and vasculature
WO2014089124A1 (en) 2012-12-03 2014-06-12 Cenexys, Inc. Immunogenic compositions for inducing an immune response for elimination of senescent cells
US20140170748A1 (en) 2012-12-14 2014-06-19 DePuy Synthes Products, LLC Nutrient Enriched Media for hUTC Growth
AU2013370228B2 (en) 2012-12-31 2018-10-18 Janssen Biotech, Inc. Suspension and clustering of human pluripotent cells for differentiation into pancreatic endocrine cells
MX2015008578A (en) 2012-12-31 2015-09-07 Janssen Biotech Inc Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators.
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
AU2013368221B2 (en) 2012-12-31 2018-11-01 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
EP2756754B1 (en) 2013-01-17 2017-01-04 Vita 34 Ag Method for the treatment of umbilical cord tissue, in particular associated with the preservation of the tissue
US20140212390A1 (en) * 2013-01-30 2014-07-31 NuTech Medical, Inc. Placental Membrane Preparation and Methods of Making and Using Same
CN115137753A (en) 2013-02-05 2022-10-04 细胞结构公司 Natural killer cells from placenta
US9867939B2 (en) 2013-03-12 2018-01-16 Allergan, Inc. Adipose tissue combinations, devices, and uses thereof
WO2014164900A1 (en) 2013-03-13 2014-10-09 Allosource Fascia fibrous compositions and methods for their use and manufacture
EP2970883B8 (en) * 2013-03-14 2021-06-23 Celularity Inc. Enhanced placental stem cells and uses thereof
CA2899713C (en) 2013-03-15 2022-07-19 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
RU2539750C2 (en) * 2013-04-09 2015-01-27 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Московский государственный университет имени М.В. Ломоносова" (МГУ) Method of evaluating immunosuppressive properties of human mesenchymal stromal cells
US20140350516A1 (en) 2013-05-23 2014-11-27 Allergan, Inc. Mechanical syringe accessory
KR102363552B1 (en) 2013-05-30 2022-02-15 그라함 에이치. 크리시 Topical neurological stimulation
US11229789B2 (en) 2013-05-30 2022-01-25 Neurostim Oab, Inc. Neuro activator with controller
US10058572B2 (en) * 2013-08-15 2018-08-28 The Regents Of The University Of California Placenta-derived multipotent stem cells
US9248384B2 (en) 2013-10-02 2016-02-02 Allergan, Inc. Fat processing system
WO2015073918A1 (en) 2013-11-16 2015-05-21 Terumo Bct, Inc. Expanding cells in a bioreactor
WO2015085232A1 (en) 2013-12-06 2015-06-11 Allosource Method of drying sheets of tissue
GB2536174B (en) * 2013-12-17 2020-12-16 Dtherapeutics Llc Devices, systems and methods for tissue engineering of luminal grafts
US10328058B2 (en) 2014-01-28 2019-06-25 Mayo Foundation For Medical Education And Research Treating atherosclerosis by removing senescent foam cell macrophages from atherosclerotic plaques
WO2015116735A1 (en) 2014-01-28 2015-08-06 Mayo Foundation For Medical Education And Research Methods and combinations for killing senescent cells and for treating senescence-associated diseases and disorders
AU2015211021B2 (en) 2014-01-28 2020-07-02 Buck Institute For Research On Aging Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders
WO2015126694A1 (en) 2014-02-12 2015-08-27 Ethicon Endo-Surgery, Inc. Method and apparatus for suprachoroidal administration of therapeutic agent
US10369237B2 (en) 2014-03-10 2019-08-06 3-D Matrix, Ltd. Sterilization and filtration of peptide compositions
KR20160127827A (en) 2014-03-10 2016-11-04 가부시끼가이샤 쓰리디 매트릭스 Self-assembling peptide compositions
EP3613841B1 (en) 2014-03-25 2022-04-20 Terumo BCT, Inc. Passive replacement of media
CA2948126A1 (en) * 2014-05-07 2015-11-12 Osiris Therapeutics, Inc. Therapeutic placental compositions, methods of making and methods of use
EP3140417B1 (en) * 2014-05-09 2021-04-21 Reelabs Private Limited Foetal polymix of mesenchymal stem cells under hypoxic conditions for the treatment of clinical disorders
US10029048B2 (en) 2014-05-13 2018-07-24 Allergan, Inc. High force injection devices
EP3954759A1 (en) 2014-05-16 2022-02-16 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
TW201603818A (en) 2014-06-03 2016-02-01 組織科技股份有限公司 Compositions and methods
US9949874B2 (en) 2014-06-06 2018-04-24 Janssen Biotech, Inc. Therapeutic agent delivery device with convergent lumen
US9925088B2 (en) 2014-06-06 2018-03-27 Janssen Biotech, Inc. Sub-retinal tangential needle catheter guide and introducer
US20160000550A1 (en) * 2014-07-05 2016-01-07 Deborah Nagle Methods for treating diseases of the colon
CN106661552B (en) * 2014-07-11 2020-07-07 玛土撤拉有限公司 Cardiac cell culture material
US10064752B2 (en) 2014-09-11 2018-09-04 Orbit Biomedical Limited Motorized suprachoroidal injection of therapeutic agent
US10322028B2 (en) 2014-09-11 2019-06-18 Orbit Biomedical Limited Method and apparatus for sensing position between layers of an eye
US10219936B2 (en) 2014-09-11 2019-03-05 Orbit Biomedical Limited Therapeutic agent delivery device with advanceable cannula and needle
US10258502B2 (en) 2014-09-18 2019-04-16 Orbit Biomedical Limited Therapeutic agent delivery device
US9504905B2 (en) 2014-09-19 2016-11-29 Bally Gaming, Inc. Card shuffling device and calibration method
KR101613478B1 (en) * 2014-09-22 2016-04-19 (주)안트로젠 Composition comprising mesenchymal stem cell-hydrogel and preparation method thereof
JP6830059B2 (en) 2014-09-26 2021-02-17 テルモ ビーシーティー、インコーポレーテッド Scheduled cell feeding
AU2015327812B2 (en) 2014-10-03 2021-04-15 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
AU2015332556B2 (en) 2014-10-14 2021-04-29 Samuel Lynch Compositions for treating wounds
WO2016068616A1 (en) * 2014-10-29 2016-05-06 차의과학대학교 산학협력단 Placenta-derived cells excreting c3 or c1r complement and composition containing same
US10709811B2 (en) * 2014-10-31 2020-07-14 The Administrators Of The Tulane Educational Fund Surgical grafts for replacing the nipple and areola or damaged epidermis
CN107106613B (en) 2014-11-07 2021-07-06 胞外体干细胞株式会社 Composition comprising stem cell-derived exosomes for adipogenic differentiation induction, adipose tissue regeneration, skin whitening or wrinkle improvement
AU2015358330A1 (en) * 2014-12-05 2017-06-08 Janssen Biotech, Inc. Treatment of ocular conditions using progenitor cells
JP6563492B2 (en) * 2014-12-05 2019-08-21 メリディジェン・バイオテック・カンパニー・リミテッドMeridigen Biotech Co., Ltd. How to differentiate mesenchymal stem cells
US20170080033A1 (en) * 2014-12-16 2017-03-23 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
WO2016099949A2 (en) * 2014-12-16 2016-06-23 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
US20160215268A1 (en) 2014-12-19 2016-07-28 Janssen Biotech, Inc. Suspension Culturing of Pluripotent Stem Cells
US10342830B2 (en) 2015-01-05 2019-07-09 Gary M. Petrucci Methods and materials for treating lung disorders
WO2016126122A2 (en) * 2015-02-04 2016-08-11 한양대학교 에리카산학협력단 Composition for chondrocyte differentiation induction or cartilage tissue regeneration, containing exosomes extracted from stem cells differentiating into chondrocytes
US11077301B2 (en) 2015-02-21 2021-08-03 NeurostimOAB, Inc. Topical nerve stimulator and sensor for bladder control
WO2016138025A2 (en) 2015-02-23 2016-09-01 Tissuetech, Inc. Apparatuses and methods for treating ophthalmic diseases and disorders
EP3268063A4 (en) 2015-03-10 2018-10-31 Allergan Pharmaceuticals Holdings (Ireland) Unlimited Company Multiple needle injector
US10772917B2 (en) 2015-03-11 2020-09-15 Ccs Ventures Limited Pancreatic endocrine progenitor cell therapies for the treatment of obesity and type 2 diabetes (T2D)
WO2016160918A1 (en) * 2015-03-31 2016-10-06 The University Of North Carolina At Chapel Hill Delivery vehicles for stem cells and uses thereof
US10286009B2 (en) * 2015-05-16 2019-05-14 Asterias Biotherapeutics, Inc. Pluripotent stem cell-derived oligodendrocyte progenitor cells for the treatment of spinal cord injury
TWI720984B (en) 2015-05-20 2021-03-11 美商帝聖工業公司 Compositions and methods for preventing the proliferation and epithelial-mesenchymal transition of epithelial cells
US10531957B2 (en) 2015-05-21 2020-01-14 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10335435B2 (en) 2015-05-22 2019-07-02 Marco Merida Method for endoscopically delivering stem cells to the brain using an intranasal, injectable approach
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US10384207B2 (en) 2015-07-21 2019-08-20 Neuro Probe Incorporated Assay apparatus and methods
WO2017019832A1 (en) 2015-07-29 2017-02-02 Medivation Technologies, Inc. Methods and compositions using repair cells and cationic dyes
WO2017019822A1 (en) * 2015-07-29 2017-02-02 Medivation Technologies, Inc. Pellet composition containing repair cells
US11707488B2 (en) 2015-08-28 2023-07-25 Rohto Pharmaceutical Co., Ltd. ROR1-positive mesenchymal stem cells and method for preparing same, pharmaceutical composition containing ROR1-positive mesenchymal stem cells and method for preparing same, and method for preventing or treating disease using ROR1-positive mesenchymal stem cells
USD797290S1 (en) 2015-10-19 2017-09-12 Spinal Surgical Strategies, Llc Bone graft delivery tool
EP3377225A4 (en) 2015-11-18 2019-07-03 President and Fellows of Harvard College Cartridge-based system for long term culture of cell clusters
WO2017095991A1 (en) * 2015-12-04 2017-06-08 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
KR20170076484A (en) * 2015-12-24 2017-07-04 삼성전자주식회사 Method of isolating senescent cell with high expression of Protocadherin gene
US10814038B2 (en) 2016-01-06 2020-10-27 3-D Matrix, Ltd. Combination compositions
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
CA3010792A1 (en) 2016-01-14 2017-07-20 DePuy Synthes Products, Inc. Composition and methods for cryopreservation of hutc
TW201733600A (en) 2016-01-29 2017-10-01 帝聖工業公司 Fetal support tissue products and methods of use
US10993969B2 (en) 2016-02-05 2021-05-04 Gary M. Petrucci Methods and materials for treating nerve injuries and neurological disorders
US11458224B2 (en) * 2016-03-04 2022-10-04 University of Pittsburgh—of the Commonwealth System of Higher Education Ovarian-derived hydrogels for biomedical and biotechnology applications
US10478553B2 (en) 2016-03-09 2019-11-19 Orbit Biomedical Limited Apparatus for subretinal administration of therapeutic agent via a curved needle
GB201604304D0 (en) 2016-03-14 2016-04-27 Tigenix S A U Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in crohn's disease
EP3223181B1 (en) * 2016-03-24 2019-12-18 Sofradim Production System and method of generating a model and simulating an effect on a surgical repair site
US11920155B2 (en) 2016-03-30 2024-03-05 Asterias Biotherapeutics, Inc. Oligodendrocyte progenitor cell compositions
EP3439716B1 (en) 2016-04-08 2023-11-01 Allergan, Inc. Aspiration and injection device
MA45479A (en) 2016-04-14 2019-02-20 Janssen Biotech Inc DIFFERENTIATION OF PLURIPOTENT STEM CELLS IN ENDODERMAL CELLS OF MIDDLE INTESTINE
WO2017188403A1 (en) 2016-04-27 2017-11-02 ロート製薬株式会社 Mesenchymal stem cell expressing at least one cell surface marker selected from the group consisting of cd201, cd46, cd56, cd147, and cd165 and method for preparing same, and pharmaceutical composition containing the mesenchymal stem cell and method for preparing same
WO2017210652A1 (en) 2016-06-03 2017-12-07 Cedars-Sinai Medical Center Cdc-derived exosomes for treatment of ventricular tachyarrythmias
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US10806629B2 (en) 2016-06-17 2020-10-20 Gyroscope Therapeutics Limited Injection device for subretinal delivery of therapeutic agent
US11000410B2 (en) 2016-06-17 2021-05-11 Gyroscope Therapeutics Limited Guide apparatus for tangential entry into suprachoroidal space
US10646374B2 (en) 2016-06-17 2020-05-12 Orbit Biomedical Limited Apparatus and method to form entry bleb for subretinal delivery of therapeutic agent
MA45502A (en) 2016-06-21 2019-04-24 Janssen Biotech Inc GENERATION OF FUNCTIONAL BETA CELLS DERIVED FROM HUMAN PLURIPOTENT STEM CELLS WITH GLUCOSE-DEPENDENT MITOCHONDRIAL RESPIRATION AND TWO-PHASE INSULIN SECRETION RESPONSE
SG11201811100RA (en) 2016-07-05 2019-01-30 Janssen Biotech Inc Treatment of retinal vascular disease using progenitor cells
US10639398B2 (en) 2016-07-05 2020-05-05 Lifecell Corporation Tissue matrices incorporating multiple tissue types
JP2019528314A (en) * 2016-08-19 2019-10-10 リーゲンティス コーポレイション Extracellular matrix for tissue reconstruction of mucosal tissue
EP3405204A4 (en) * 2016-08-26 2020-03-18 Restem Llc Composition and methods of using umbilical cord lining stem cells
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
MX2019005172A (en) * 2016-11-02 2019-09-26 Axogen Corp Amnion tissue grafts and methods of preparing and using same.
RU2644306C1 (en) * 2016-11-22 2018-02-08 Общество с ограниченной ответственностью "ДЖИ-Групп" Method of recovery of defects of covering tissues
WO2018125851A1 (en) * 2016-12-26 2018-07-05 Michael Moeller Systems and methods to isolate and expand stem cells from urine
US11273072B2 (en) 2017-01-13 2022-03-15 Gyroscope Therapeutics Limited Suprachoroidal injection device
US10772986B2 (en) 2017-01-26 2020-09-15 Allosource Fascia fibrous compositions and methods for their use and manufacture
WO2018164228A1 (en) 2017-03-08 2018-09-13 ロート製薬株式会社 Ror1-positive mesenchymal stem cell-containing pharmaceutical composition for preventing or treating disease associated with fibrosis, method for preparing same, and method for preventing or treating disease associated with fibrosis using ror1-positive mesenchymal stem cells
US11076984B2 (en) 2017-03-13 2021-08-03 Gyroscope Therapeutics Limited Method of performing subretinal drainage and agent delivery
US11278597B2 (en) 2017-03-17 2022-03-22 Rutgers, The State University Of New Jersey Compositions and methods for wound healing
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP4317422A2 (en) 2017-04-13 2024-02-07 Senti Biosciences, Inc. Combinatorial cancer immunotherapy
WO2018195210A1 (en) 2017-04-19 2018-10-25 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US10478531B2 (en) 2017-06-22 2019-11-19 Gary M. Petrucci Methods and materials for treating blood vessels
US11685951B2 (en) 2017-07-18 2023-06-27 The Research Foundation For The State University Of New York Biomarkers for intracranial aneurysm
US10251917B1 (en) 2017-09-19 2019-04-09 Gary M. Petrucci Methods and materials for treating tumors
US10821205B2 (en) 2017-10-18 2020-11-03 Lifecell Corporation Adipose tissue products and methods of production
US11123375B2 (en) 2017-10-18 2021-09-21 Lifecell Corporation Methods of treating tissue voids following removal of implantable infusion ports using adipose tissue products
CA3075106A1 (en) 2017-10-19 2019-04-25 Lifecell Corporation Flowable acellular tissue matrix products and methods of production
US11246994B2 (en) 2017-10-19 2022-02-15 Lifecell Corporation Methods for introduction of flowable acellular tissue matrix products into a hand
US11629318B2 (en) 2017-10-20 2023-04-18 President And Fellows Of Harvard College Methods for producing mature adipocytes and methods of use thereof
US20210038650A1 (en) * 2017-10-23 2021-02-11 Cell Medicine, Inc. Mesenchymal stem cell therapy of leigh syndrome
TW201932126A (en) * 2017-11-03 2019-08-16 美商健生生物科技公司 Method of inhibiting angiogenesis
CN111601636A (en) 2017-11-07 2020-08-28 Oab神经电疗科技公司 Non-invasive neural activator with adaptive circuit
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11285177B2 (en) 2018-01-03 2022-03-29 Globus Medical, Inc. Allografts containing viable cells and methods thereof
EP3738598A4 (en) 2018-01-12 2021-06-09 Osaka University Stratified squamous epithelial cell normal differentiation and maturation promoting agent, epithelial disease therapeutic agent, and stratified squamous epithelial cell normal differentiation and maturation promoting method
US20190218521A1 (en) * 2018-01-18 2019-07-18 Lorenzo Bracco Culture medium of viruses for human vaccines have to consist of human cells from placenta and/or from umbilical cord of a fetus of the blood type 0 Rh- and of the mother of the blood type 0 Rh- (both, fetus and mother, must be of the blood type 0 Rh-)
WO2019152483A1 (en) * 2018-01-30 2019-08-08 University Of Georgia Research Foundation, Inc. Methods for vascular construction and products therefrom
CN108384752A (en) * 2018-02-13 2018-08-10 中国人民解放军第四五五医院 Application at Cartilage culture base and its in micro- splitting technique in knee cartilage differentiation
JP2021526556A (en) * 2018-05-30 2021-10-07 ダイレクト バイオロジクス エルエルシー Auxiliary agents containing mesenchymal stem cell (MSC) preparations, which are frozen or powdered growth factors and extracellular vesicles, and how to use them.
CN108961233A (en) * 2018-06-28 2018-12-07 华侨大学 A kind of composite polycrystal-diamond surface defect classifying identification method
US11419898B2 (en) 2018-10-17 2022-08-23 Senti Biosciences, Inc. Combinatorial cancer immunotherapy
EP3866813A4 (en) 2018-10-17 2022-08-03 Senti Biosciences, Inc. Combinatorial cancer immunotherapy
AU2020210757A1 (en) 2019-01-23 2021-08-05 Asterias Biotherapeutics, Inc. Dorsally-derived oligodendrocyte progenitor cells from human pluripotent stem cells
US11759355B1 (en) 2019-02-26 2023-09-19 Gyroscope Therapeutics Limited Method of delivering leading blebs and agent to subretinal space
KR102169924B1 (en) * 2019-03-26 2020-10-26 연세대학교 산학협력단 Composition for inducing chondrogenesis and use thereof
KR102167257B1 (en) * 2019-05-28 2020-10-19 부산대학교 산학협력단 Method of promoting differentiation of stem cells into mature cardiomyocytes using tomatidine
WO2020243497A1 (en) 2019-05-30 2020-12-03 Lifecell Corporation Biologic breast implant
US11458311B2 (en) 2019-06-26 2022-10-04 Neurostim Technologies Llc Non-invasive nerve activator patch with adaptive circuit
WO2021039943A1 (en) * 2019-08-29 2021-03-04 味の素株式会社 Method for producing mesenchymal stem cells from living body-derived cell sample containing mesenchymal stem cells
WO2021050907A1 (en) * 2019-09-13 2021-03-18 The University Of North Carolina At Chapel Hill Method of making human mouse xenografts
JP2022550911A (en) * 2019-10-07 2022-12-05 ユニバーシティ オブ ユタ リサーチ ファウンデーション Chondrogenic human mesenchymal stem cell (MSC) sheet
KR102623662B1 (en) 2019-10-11 2024-01-12 자이로스코프 테라퓨틱스 리미티드 Dosing clip assembly for syringe
KR20220115802A (en) 2019-12-16 2022-08-18 뉴로스팀 테크놀로지스 엘엘씨 Non-invasive neural activator with boost charge transfer function
CN111718410B (en) * 2020-06-05 2022-10-11 江南大学 Method for preparing yolk immunoglobulin
JP6967308B1 (en) 2020-06-30 2021-11-17 国立大学法人高知大学 Cranial nerve disorder therapeutic agent containing tissue cell culture supernatant derived from fetal appendages
WO2022046954A1 (en) * 2020-08-25 2022-03-03 Celularity Inc. Appl cells improved placental-derived adherent cells and methods of their use
US20230390111A1 (en) 2020-12-22 2023-12-07 Gyroscope Therapeutics Limited Ocular cannula guide
JP2024502646A (en) * 2021-01-12 2024-01-22 セラトズ セラピュティクス インコーポレイテッド Screening method for nerve regeneration-promoting cells with nerve regeneration activity
CN115068438B (en) * 2022-04-28 2023-09-22 浙江大学医学院附属邵逸夫医院 Method for preparing cell membrane nano vesicle with osteoclast precursor homology targeting and application thereof
CN115044542B (en) * 2022-06-30 2023-09-26 上海市东方医院(同济大学附属东方医院) Application of SJ000291942 in inducing mesenchymal stem cells to osteogenic differentiation
WO2024024708A1 (en) * 2022-07-26 2024-02-01 国立大学法人大阪大学 Composition for cartilage repair and method for manufacturing same

Family Cites Families (326)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US229971A (en) 1880-07-13 Ttornets
US2324800A (en) 1941-08-14 1943-07-20 Pfizer Charles & Co Purification of riboflavin
US2654735A (en) 1949-07-29 1953-10-06 Us Vitamin Corp Process for the production of derivatives of 9-polyhydroxyalkylisoalloxazines and products obtained
US2864848A (en) * 1954-07-19 1958-12-16 Ca Nat Research Council Method of producing l-alpha-glycerylphosphorylcholine
US2912332A (en) 1958-02-27 1959-11-10 Swift & Co Stabilized thiamine composition and method of enriching food products
US3665061A (en) 1969-07-16 1972-05-23 United States Banknote Corp Process for producing collagen sponges
JPS5651747B2 (en) * 1973-05-31 1981-12-08
CH635748A5 (en) 1977-08-16 1983-04-29 Cellorgan Laboratoires Sa PROCESS FOR OBTAINING PLACENTAL CELLS OF SHEEP.
US4216144A (en) 1977-10-20 1980-08-05 Ashmead H H Soluble iron proteinates
JPS6040439B2 (en) * 1978-03-29 1985-09-11 大正製薬株式会社 hydrocortisone derivatives
US4352883A (en) 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4393240A (en) 1981-07-06 1983-07-12 Stille John K Optically active phosphines
US4544516A (en) 1982-07-28 1985-10-01 Battelle Development Corporation Collagen orientation
US4465776A (en) * 1982-09-27 1984-08-14 Research Corporation Monoclonal antibodies to vitamin B6 and immunoassay method
US4657866A (en) * 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
US4487865A (en) 1983-12-15 1984-12-11 Biomatrix, Inc. Polymeric articles modified with hyaluronate
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
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
US4925667A (en) * 1986-05-27 1990-05-15 Qmax Technology Group, Inc. Substrate with particulate cosmetic
JPS642643A (en) 1987-06-26 1989-01-06 Bio Material Yunibaasu:Kk Artificial skin
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
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
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
US5162405A (en) * 1987-12-24 1992-11-10 Elf Atochem North America, Inc. Single-functional and mixtures of multi-functional oligomeric performance additive compositions and their uses
GB8803697D0 (en) 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
US4963439A (en) 1988-04-19 1990-10-16 Ube Industries, Ltd. Continuous fiber-reinforced Al-Co alloy matrix composite
US20030032178A1 (en) * 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
US5618670A (en) * 1988-08-26 1997-04-08 The United States Of America As Represented By The Department Of Health & Human Services Detection method for c-raf-1 genes
US4925677A (en) 1988-08-31 1990-05-15 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
JPH06104061B2 (en) 1989-02-10 1994-12-21 花王株式会社 Cell culture support material
US5284766A (en) 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
US4994602A (en) * 1989-02-27 1991-02-19 Takasago International Corporation Process for preparing optically active 6-t-butyoxy-3,5-dihydroxyhexanoic esters
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
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
US5574205A (en) 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
US5840580A (en) 1990-05-01 1998-11-24 Becton Dickinson And Company Phenotypic characterization of the hematopoietic stem cell
US5145770A (en) * 1990-06-04 1992-09-08 Biosurface Technology, Inc. Cryopreservation of cultured epithelial sheets
US5612205A (en) 1990-08-29 1997-03-18 Genpharm International, Incorporated Homologous recombination in mammalian cells
US5336616A (en) * 1990-09-12 1994-08-09 Lifecell Corporation Method for processing and preserving collagen-based tissues for transplantation
US5342761A (en) 1990-10-01 1994-08-30 Research Development Foundation Oncofetal gene, gene product and uses therefor
US5506134A (en) 1990-10-22 1996-04-09 Corvas International, Inc. Hypridoma and monoclonal antibody which inhibits blood coagulation tissue factor/factor VIIa complex
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
US5140100A (en) * 1990-12-28 1992-08-18 Cedars-Sinai Medical Center Protein that inhibits production of human choriogonadotropin
US5286632A (en) * 1991-01-09 1994-02-15 Jones Douglas H Method for in vivo recombination and mutagenesis
NL9100038A (en) * 1991-01-11 1992-08-03 Stamicarbon ENZYME-CATALYZED PREPARATION OF OPTICALLY ACTIVE CARBONIC ACIDS.
SK120193A3 (en) 1991-05-02 1994-07-06 Yeda Res & Dev Pharmaceutical composition for the prevention and/or treatment of pathological processes
US6399369B1 (en) * 1991-07-08 2002-06-04 Neurospheres Holdings Ltd. Multipotent neural stem cell cDNA libraries
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
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
WO1993006726A1 (en) * 1991-09-30 1993-04-15 Mackenzie Walser Methods for treatment of free-radical-mediated tissue injury
US5308763A (en) * 1991-10-01 1994-05-03 The Johns Hopkins University Method of making primary culture of olfactory neurons
US5914265A (en) * 1992-04-30 1999-06-22 Baylor College Of Medicine Keratin K1 expression vectors and methods of use
WO1994000484A1 (en) 1992-06-22 1994-01-06 Young Henry E Scar inhibitory factor and use thereof
US5320962A (en) * 1992-07-22 1994-06-14 Duke University DNA encoding the human A1 adenosine receptor
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US5356807A (en) * 1992-09-08 1994-10-18 Cornell Research Foundation Cultured cell line of adult diploid cells from human brain and meningeal tissue
US20040224409A1 (en) 1992-09-25 2004-11-11 Laurent Pradier Recombinant adenoviruses coding for brain-derived neurotrophic factor (BDNF)
WO1994010331A1 (en) 1992-10-29 1994-05-11 The Australian National University Angiogenesis inhibitory antibodies
US5955343A (en) 1992-12-28 1999-09-21 Massachusetts Institute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5670483A (en) 1992-12-28 1997-09-23 Massachusetts Insititute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5707643A (en) * 1993-02-26 1998-01-13 Santen Pharmaceutical Co., Ltd. Biodegradable scleral plug
US5494899A (en) * 1993-04-07 1996-02-27 Oklahoma Medical Research Foundation Selective regulation of B lymphocyte precursors by hormones
WO1994025584A1 (en) 1993-04-28 1994-11-10 Johns Hopkins University School Of Medicine Chronic endothelial cell culture under flow
IL110589A0 (en) 1993-08-10 1994-11-11 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF- beta family
JP3680114B2 (en) 1993-09-17 2005-08-10 敏一 中村 Cranial nerve disorder treatment
US6686198B1 (en) 1993-10-14 2004-02-03 President And Fellows Of Harvard College Method of inducing and maintaining neuronal cells
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US5456835A (en) * 1993-11-08 1995-10-10 Hemasure, Inc. Device and process for removing free hemoglobin from blood
DE4406073A1 (en) * 1994-02-24 1995-08-31 Univ Ludwigs Albert Process for the production of human, clonogenic fibroblasts, process for gene transfection of fibroblasts and fibroblasts obtained in this way
US5698518A (en) * 1994-03-30 1997-12-16 Oklahoma Medical Research Foundation Method for regulating inflammation and tumor growth with calmodulin, calmodulin analogues or calmodulin antagonists
US5466233A (en) 1994-04-25 1995-11-14 Escalon Ophthalmics, Inc. Tack for intraocular drug delivery and method for inserting and removing same
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
US6703017B1 (en) 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US5834308A (en) 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
DK0952792T3 (en) 1994-06-06 2003-12-08 Osiris Therapeutics Inc Biomatrix for tissue regeneration
IL114397A0 (en) 1994-07-01 1995-10-31 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF-beta-family
US5935849A (en) * 1994-07-20 1999-08-10 Cytotherapeutics, Inc. Methods and compositions of growth control for cells encapsulated within bioartificial organs
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US5660982A (en) * 1994-10-04 1997-08-26 Tryggvason; Karl Laminin chains: diagnostic uses
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
US5725493A (en) 1994-12-12 1998-03-10 Avery; Robert Logan Intravitreal medicine delivery
US5684032A (en) 1994-12-13 1997-11-04 Smithkline Beecham Corporation Compounds
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5906934A (en) * 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5718922A (en) 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
US5869079A (en) 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
US5693332C1 (en) 1995-08-11 2001-01-09 Univ California Human keratinocytes supported on a hydrophilic membrane and methods of using same to effect wound closure
US5641750A (en) * 1995-11-29 1997-06-24 Amgen Inc. Methods for treating photoreceptors using glial cell line-derived neurotrophic factor (GDNF) protein product
US6200606B1 (en) * 1996-01-16 2001-03-13 Depuy Orthopaedics, Inc. Isolation of precursor cells from hematopoietic and nonhematopoietic tissues and their use in vivo bone and cartilage regeneration
US5842477A (en) 1996-02-21 1998-12-01 Advanced Tissue Sciences, Inc. Method for repairing cartilage
DK0888452T3 (en) 1996-03-15 2004-06-14 Munin Corp Human CYR61, an extracellular matrix signaling molecule
US6223188B1 (en) * 1996-04-10 2001-04-24 Sun Microsystems, Inc. Presentation of link information as an aid to hypermedia navigation
US6497875B1 (en) 1996-04-26 2002-12-24 Case Western Reserve University Multilayer skin or dermal equivalent having a layer containing mesenchymal stem cells
US6358737B1 (en) 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US6787355B1 (en) * 1996-08-26 2004-09-07 Mcgill University Multipotent neural stem cells from peripheral tissues and uses thereof
US5919702A (en) 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
US6541028B1 (en) * 1997-01-17 2003-04-01 Celadon Science, Llc Methods for promoting healing of corneal resurfacing wounds
AU6144698A (en) 1997-02-05 1998-08-25 Case Western Reserve University Stimulatory effects of bfgf and bmp-2 on osteogenic differentiation of mesenchymal stem cells
EP0989855A4 (en) 1997-05-13 2002-06-12 Osiris Therapeutics Inc Osteoarthritis cartilage regeneration using human mesenchymal stem cells
WO1998052560A1 (en) * 1997-05-21 1998-11-26 Sloan-Kettering Institute For Cancer Research Method for increasing the concentration of ascorbic acid in brain tissues of a subject
JP4132089B2 (en) 1997-05-30 2008-08-13 オステオバイオロジックス,インコーポレイテッド Fiber reinforced porous biodegradable implantation device
ES2251773T5 (en) 1997-07-14 2009-05-12 Osiris Therapeutics, Inc. REGENERATION OF THE CARDIAC MUSCLE USING MESENQUIMATOSE PRECURSOR CELLS.
US5902598A (en) 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
ATE478135T1 (en) 1997-12-02 2010-09-15 Artecel Sciences Inc DIFFERENTIATION IN OSTEOBLASTS OF ADIIO TISSUE-STROMAL CELLS AND THEIR USES
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
EP1062321B1 (en) 1998-03-13 2004-12-29 Osiris Therapeutics, Inc. Uses for humane non-autologous mesenchymal stem cells
DE69925268T2 (en) 1998-03-16 2006-03-23 Cytovia, Inc., San Diego Dipeptide caspase inhibitors and their use
US6179872B1 (en) * 1998-03-17 2001-01-30 Tissue Engineering Biopolymer matt for use in tissue repair and reconstruction
US6171610B1 (en) * 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
ES2246085T3 (en) 1998-05-07 2006-02-01 University Of South Florida OSEA MEDULA CELLS AS A SOURCE OF NEURONS USEFUL TO REPAIR THE SPINAL MEDULA AND THE BRAIN.
ES2292245T3 (en) * 1998-05-29 2008-03-01 Osiris Therapeutics, Inc. MESENCHIMATIC MOTHER CELLS CD45 + AND / OR FIBROBLASTO + HUMANAS.
US6323188B1 (en) 1998-07-01 2001-11-27 Donald L. Weissman Treatment and prevention of cardiovascular diseases, heart attack, and stroke, primary and subsequent, with help of aspirin and certain vitamins
DE19833340A1 (en) 1998-07-24 2000-02-10 Karlsruhe Forschzent Worm-shaped working mechanism
US20040037818A1 (en) * 1998-07-30 2004-02-26 Brand Stephen J. Treatment for diabetes
EP1105460B1 (en) 1998-08-10 2009-10-07 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Differentiation of non-insulin producing cells into insulin producing cells by glp-1 or exendin-4 and uses thereof
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
US6284245B1 (en) 1998-08-25 2001-09-04 Diacrin, Inc. Neural retinal cells and retinal pigment epithelium cells and their use in treatment of retinal disorders
US6444205B2 (en) 1998-09-30 2002-09-03 Diacrin, Inc. Transplantation of neural cells for the treatment of chronic pain or spasticity
US6610540B1 (en) 1998-11-18 2003-08-26 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US6241369B1 (en) * 1998-11-20 2001-06-05 Cooper Technologies Company Quick mount fixture
US6144054A (en) * 1998-12-04 2000-11-07 International Business Machines Corporation DRAM cell having an annular signal transfer region
DE19856428C1 (en) * 1998-12-08 2000-05-04 Heraeus Noblelight Gmbh Discharge lamp, having spiral, inner electrode arranged at inner wall of interior tube
DE69919029T2 (en) 1998-12-24 2005-09-08 Biosafe S.A. DEVICE FOR BLOOD SEPARATION, ESPECIALLY FOR CONCENTRATING HEMATOPOIETIC STEM CELLS
JP4932069B2 (en) 1999-01-25 2012-05-16 株式会社セルシード Acrylamide derivatives and polymers containing the derivatives
JP2002535982A (en) 1999-02-04 2002-10-29 マックギル ユニヴァーシティー Platform for cell differentiation
EP1175487A2 (en) 1999-02-10 2002-01-30 Curis, Inc. Pancreatic progenitor cells, methods and uses related thereto
US6592623B1 (en) 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
KR100979664B1 (en) 1999-03-10 2010-09-02 유니버시티 오브 피츠버그 오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Adipose-derived stem cells and lattices
US20030007954A1 (en) 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US6384105B1 (en) 1999-04-16 2002-05-07 William Marsh Rice University Poly(Propylene Fumarate) cross linked with Poly(Ethylene Glycol)
US7371400B2 (en) 2001-01-02 2008-05-13 The General Hospital Corporation Multilayer device for tissue engineering
US6261600B1 (en) * 1999-04-30 2001-07-17 Drugtech Corporation Folic acid supplement
US6287340B1 (en) 1999-05-14 2001-09-11 Trustees Of Tufts College Bioengineered anterior cruciate ligament
US6372494B1 (en) * 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
WO2000073421A2 (en) 1999-06-02 2000-12-07 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6329904B1 (en) * 1999-06-11 2001-12-11 Safety Through Cellular, Inc. Apparatus and method for providing weather and other alerts
AU5886000A (en) 1999-06-25 2001-01-31 Northwestern University Compositions, kits, and methods for modulating survival and differentiation of multi-potential hematopoietic progenitor 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
EP2348104A1 (en) 1999-08-05 2011-07-27 Mcl Llc Multipotent adult stem cells and methods for isolation
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US6555374B1 (en) 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
CA2386536A1 (en) 1999-09-14 2001-03-22 The Children's Medical Center Corporation Methods for treating muscular dystrophy with bone marrow cells
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
US20030129745A1 (en) 1999-10-28 2003-07-10 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
EP1099754A1 (en) 1999-11-10 2001-05-16 Universiteit Leiden Mesenchymal stem cells and/or progenitor cells, their isolation and use
US20020164307A1 (en) 1999-12-06 2002-11-07 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20030082155A1 (en) * 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
IL149933A0 (en) 1999-12-06 2002-11-10 Gen Hospital Corp Pancreatic stem cells and their use in transplantation
US7785882B2 (en) 2000-01-18 2010-08-31 Cornell Research Foundation, Inc. Neuronal progenitor cells from hippocampal tissue and a method for isolating and purifying them
US7544509B2 (en) 2000-01-24 2009-06-09 Mcgill University Method for preparing stem cell preparations
US6610535B1 (en) 2000-02-10 2003-08-26 Es Cell International Pte Ltd. Progenitor cells and methods and uses related thereto
US7514259B2 (en) 2000-02-11 2009-04-07 Schepens Eye Research Institute Isolation and transplantation of retinal stem cells
WO2001059072A1 (en) 2000-02-11 2001-08-16 Philadelphia Health And Education Corporation Differentiation of bone marrow cells into neuronal cells and uses therefor
US7160724B2 (en) * 2000-03-09 2007-01-09 University Of South Florida Human cord blood as a source of neural tissue for repair of the brain and spinal cord
WO2001065934A2 (en) * 2000-03-09 2001-09-13 Lee Walters Applications of immune system tolerance to treatment of various diseases
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
US6436704B1 (en) 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
US6673606B1 (en) * 2000-04-12 2004-01-06 The Children's Hospital Of Philadelphia Therapeutic uses for mesenchymal stromal cells
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
AU6459801A (en) 2000-05-12 2001-11-26 Mark T Keating Compositions and methods for cell dedifferentiation and tissue regeneration
US8273570B2 (en) 2000-05-16 2012-09-25 Riken Process of inducing differentiation of embryonic cell to cell expressing neural surface marker using OP9 or PA6 cells
US7049072B2 (en) 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
US6759039B2 (en) * 2000-06-30 2004-07-06 Amcyte, Inc. Culturing pancreatic stem cells having a specified, intermediate stage of development
EP1312669B1 (en) 2000-07-21 2011-09-14 CellSeed Inc. Cultured epidermal cell sheet, laminated cultured skin sheet and process for producing the same
DE60143892D1 (en) 2000-07-21 2011-03-03 Cellseed Ind HEART MUSCLES-SIMILAR CELL LAYER, THREE-DIMENSIONAL CONSTRUCT, HEART MUSCLE-SIMILAR TISSUE, AND METHOD OF MANUFACTURE
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
DE10038814A1 (en) * 2000-08-09 2002-02-21 Abb Research Ltd High voltage direct current transformer
AU2001293586A1 (en) 2000-09-29 2002-04-08 Vincent Tropepe Primitive neural stem cells and method for differentiation of stem cells to neural cells
US6639470B1 (en) 2000-10-06 2003-10-28 Skyworks Solutions, Inc. Constant current biasing circuit for linear power amplifiers
US7560280B2 (en) 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
WO2002036749A2 (en) 2000-11-06 2002-05-10 The Salk Institute For Biological Studies Postmortem stem cells
AU2001297880B2 (en) 2000-11-30 2007-05-31 Stemron Inc. Isolated homozygous stem cells differentiated cells derived therefrom and materials and methods for making and using same
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
AU2020902A (en) 2000-12-06 2002-06-18 Robert J Hariri Method of collecting placental stem cells background of the invention
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US6599323B2 (en) 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
CA2435826A1 (en) 2001-01-24 2002-08-01 The Government Of The United States Of America Differentiation of stem cells to pancreatic endocrine cells
WO2002061053A1 (en) 2001-01-31 2002-08-08 The General Hospital Corporation Renal stem cells and uses thereof
US7449180B2 (en) 2001-02-06 2008-11-11 John Kisiday Macroscopic scaffold containing amphiphilic peptides encapsulating cells
JP5057629B2 (en) 2001-02-06 2012-10-24 マサチューセッツ インスティテュート オブ テクノロジー Encapsulation of tissue cells and their use in peptide scaffolds
ES2549161T3 (en) 2001-02-14 2015-10-23 Anthrogenesis Corporation Postpartum mammalian placenta, its use and placental stem cells thereof
IL157350A0 (en) * 2001-02-14 2004-02-19 Anthrogenesis Corp Post-partum mammalian placenta, its use and placental stem cells therefrom
EP1367899A4 (en) 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
JP2005503120A (en) * 2001-03-27 2005-02-03 マサチューセッツ インスティテュート オブ テクノロジー Methods and products for FGF dimerization
US7838292B1 (en) 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
CA2442177A1 (en) 2001-03-29 2002-10-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
US20050054102A1 (en) 2001-04-19 2005-03-10 Anna Wobus Method for differentiating stem cells into insulin-producing cells
KR100762261B1 (en) 2001-04-27 2007-10-04 (주)바이오니아 Process for preparation of full-length cDNA and anchor and primer used for the same
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
US20030022369A1 (en) 2001-05-18 2003-01-30 Helen Fillmore Differentiation of specialized dermal and epidermal cells into neuronal cells
EP1401282A4 (en) * 2001-05-25 2005-03-30 Cythera Inc Stem cell differentiation
EP1406573A4 (en) * 2001-06-07 2005-03-30 Skinmedica Inc Conditioned cell culture media and uses thereof
RU2183966C1 (en) 2001-07-06 2002-06-27 Общество с ограниченной ответственностью "Сибларекс" Composition of flavonoid complex "siblareks" for biologically active supplements of medicinal and chemical-pharmaceutical articles and method of preparing bioflavonoid complex "siblareks" for biologically active supplements of medicinal and chemical-pharmaceutical articles
US6402263B1 (en) 2001-07-24 2002-06-11 Robert Bosch Corporation Dual actuation master cylinder
ATE443758T1 (en) * 2001-08-08 2009-10-15 Levesque Biosciences Inc COMPOSITIONS AND METHODS FOR ISOLATION, PROPAGATION AND DIFFERENTIATION OF NON-EMBRYONAL HUMAN STEM CELLS AND USES THEREOF
US20030211603A1 (en) 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
WO2003018767A2 (en) * 2001-08-27 2003-03-06 Advanced Cell Technology, Inc. Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20030104997A1 (en) * 2001-09-05 2003-06-05 Black Ira B. Multi-lineage directed induction of bone marrow stromal cell differentiation
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
US20050064587A1 (en) 2001-09-07 2005-03-24 Lawrence Rosenberg Pancreatic small cells and uses thereof
US9969980B2 (en) 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
EP1298201A1 (en) 2001-09-27 2003-04-02 Cardion AG Process for the production of cells exhibiting an islet-beta-cell-like state
US7072332B2 (en) * 2001-09-27 2006-07-04 Samsung Electronics Co., Ltd. Soft switch using distributed firewalls for load sharing voice-over-IP traffic in an IP network
GB2396623B (en) 2001-09-28 2006-04-05 Es Cell Int Pte Ltd Methods of derivation and propagation of undifferentiated human embryonic stem (hes) cells on feeder-free matrices and human feeder layers
WO2003033697A1 (en) 2001-10-18 2003-04-24 Ixion Biotechnology, Inc. Conversion of liver stem and progenitor cells to pancreatic functional cells
US7129034B2 (en) * 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
EP1453954A4 (en) * 2001-11-09 2004-12-15 Artecel Sciences Inc Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
US7491690B2 (en) 2001-11-14 2009-02-17 Northwestern University Self-assembly and mineralization of peptide-amphiphile nanofibers
EP1442115B9 (en) * 2001-11-15 2009-12-16 Children's Medical Center Corporation Methods of isolation, expansion and differentiation of fetal stem cells from chorionic villus, amniotic fluid, and placenta and therapeutic uses thereof
JP3728750B2 (en) * 2001-11-22 2005-12-21 ニプロ株式会社 Cultured skin and method for producing the same
EP1449542B1 (en) 2001-11-28 2012-06-13 AnGes MG, Inc. Hgf as genetic remedy for parkinson's disease
US6712850B2 (en) 2001-11-30 2004-03-30 Ethicon, Inc. Porous tissue scaffolds for the repair and regeneration of dermal tissue
WO2003048336A2 (en) 2001-12-04 2003-06-12 Organogenesis Inc. Cultured cells from pancreatic islets
AU2002341725A1 (en) 2001-12-06 2003-07-09 The Regents Of The University Of California Method for differentiating islet precursor cells into beta cells
TW200301132A (en) * 2001-12-06 2003-07-01 Sankyo Co Pharmaceutical compositions containing cells derived from human caul
EP1463798A4 (en) * 2001-12-07 2005-01-19 Geron Corp Islet cells from human embryonic stem cells
EP1451299A4 (en) * 2001-12-11 2008-03-05 Cytograft Tissue Engineering I Tissue engineered cellular sheets, methods of making and use thereof
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
US20030113910A1 (en) * 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
AU2002350352A1 (en) * 2001-12-21 2003-07-15 Mount Sinai Hospital Cellular compositions and methods of making and using them
US7101546B2 (en) 2001-12-21 2006-09-05 Amcyte, Inc. In situ maturation of cultured pancreatic stem cells having a specified, intermediate stage of development
CN1671835A (en) 2001-12-28 2005-09-21 塞拉提斯股份公司 A method for the establishment of a pluripotent human blastocyst-derived stem cell line
AU2003202988A1 (en) 2002-01-14 2003-07-30 The Board Of Trustees Of The University Of Illinois Use of modified pyrimidine compounds to promote stem cell migration and proliferation
US20030158089A1 (en) * 2002-01-24 2003-08-21 Xenoport, Inc. Administrative agents via the SMVT transporter
US20030162290A1 (en) * 2002-01-25 2003-08-28 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
WO2003066832A2 (en) 2002-02-07 2003-08-14 The Research Foundation Of The State University Of New York Generation of new insulin cells from progenitor cells present in adult pancreatic islets
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
AU2003215297A1 (en) 2002-02-15 2003-09-09 Cornell Research Foundation, Inc. Enhancing neurotrophin-induced neurogenesis by endogenous neural progenitor cells by concurrent overexpression of brain derived neurotrophic factor and an inhibitor of a pro-gliogenic bone morphogenetic protein
WO2003070749A2 (en) 2002-02-15 2003-08-28 Northwestern University Self-assembly of peptide-amphiphile nanofibers under physiological conditions
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
AU2003217658A1 (en) 2002-02-22 2003-09-09 University Of Florida Cellular trans-differentiation
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20030161818A1 (en) 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7150990B2 (en) 2002-03-06 2006-12-19 Reprocell, Inc. Self-renewing pluripotent hepatic stem cells
JP2003259862A (en) 2002-03-12 2003-09-16 Fuji Photo Film Co Ltd Cell-culturing carrier
AU2003221173A1 (en) 2002-03-27 2003-10-08 Asahi Kasei Kabushiki Kaisha Placenta-origin mesenchymal cells and medicinal use thereof
CA2481385A1 (en) 2002-04-12 2003-10-23 Celgene Corporation 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
JP4136434B2 (en) 2002-04-17 2008-08-20 進 清野 Induction of insulin-producing cells
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
CA2483010A1 (en) 2002-04-19 2003-10-30 University Of Pittsburgh Of The Commonwealth System Of Higher Education Placental derived stem cells and uses thereof
CA2484223A1 (en) 2002-04-25 2003-11-06 Wisconsin Alumni Research Foundation Use of human neural stem cells secreting gdnf for treatment of parkinson's and other neurodegenerative diseases
US20040029269A1 (en) 2002-05-07 2004-02-12 Goldman Steven A Promoter-based isolation, purification, expansion, and transplantation of neuronal progenitor cells, oligodendrocyte progenitor cells, or neural stem cells from a population of embryonic stem cells
US20040014662A1 (en) * 2002-05-08 2004-01-22 Per Lindquist Modulation of neural stem cells and neural progenitor cells
JP2005527241A (en) 2002-05-28 2005-09-15 ベクトン・ディキンソン・アンド・カンパニー Methods for proliferation of human pancreatic acinar cells and transdifferentiation into insulin producing cells in vitro
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
EP1511838A4 (en) 2002-06-07 2007-01-10 Es Cell Int Pte Ltd Methods of regulating differentiation in stem cells
US7410797B2 (en) 2002-06-11 2008-08-12 Ogle Roy C Meningeal-derived stem cells
AU2003279912A1 (en) 2002-06-27 2004-01-19 Northwestern University Peptide rod amphiphiles and self-assembly of same
US7285415B2 (en) 2002-07-11 2007-10-23 The Regents Of The University Of California Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
GB0216286D0 (en) 2002-07-15 2002-08-21 Univ Leeds Network
JP2005532810A (en) 2002-07-16 2005-11-04 イッサム リサーチ ディベロップメント カンパニー オブ ザ ヘブリュー ユニバーシティー オブ エルサレム Methods for transplanting mesenchymal stem cells for tissue repair and tissue formation
US7390659B2 (en) * 2002-07-16 2008-06-24 The Trustees Of Columbia University In The City Of New York Methods for inducing differentiation of embryonic stem cells and uses thereof
EP1531838A2 (en) 2002-07-29 2005-05-25 Asahi Kasei Kabushiki Kaisha Stem cells for treating pancreatic damage
CA2494040A1 (en) 2002-07-29 2004-02-05 Es Cell International Pte Ltd. Multi-step method for the differentiation of insulin positive, glucose
WO2004016747A2 (en) 2002-08-14 2004-02-26 University Of Florida Bone marrow cell differentiation
AU2003265094A1 (en) * 2002-08-26 2004-03-11 Neuronova Ab Method for culturing stem cells
US20040063202A1 (en) * 2002-08-28 2004-04-01 Petersen Bryon E. Neurogenesis from hepatic stem cells
EP1539928A4 (en) 2002-09-06 2006-09-06 Amcyte Inc Cd56 positive human adult pancreatic endocrine progenitor cells
US9969977B2 (en) 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
US6766863B2 (en) * 2002-09-20 2004-07-27 Hypro Corporation Fire fighting foam injection system with auto-start feature
US20040062753A1 (en) 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
JP4262465B2 (en) * 2002-10-24 2009-05-13 富士フイルム株式会社 Cell culture method
WO2004039248A2 (en) 2002-10-31 2004-05-13 The General Hospital Corporation Repairing or replacing tissues or organs
CA2547899C (en) 2002-12-05 2014-04-15 Case Western Reserve University Cell-based therapies for ischemia
JP4571387B2 (en) 2003-02-07 2010-10-27 宣男 櫻川 Human amnion-derived side population cells and uses thereof
PT1594957E (en) * 2003-02-11 2010-11-30 John E Davies Progenitor cells from wharton`s jelly of human umbilical cord
AU2004210853A1 (en) 2003-02-11 2004-08-26 Northwestern University Methods and materials for nanocrystalline surface coatings and attachment of peptide amphiphile nanofibers thereon
US7521481B2 (en) 2003-02-27 2009-04-21 Mclaurin Joanne Methods of preventing, treating and diagnosing disorders of protein aggregation
US20040224401A1 (en) 2003-03-28 2004-11-11 Ludwig Tenneille E. Physiochemical culture conditions for embryonic stem cells
US7960166B2 (en) 2003-05-21 2011-06-14 The General Hospital Corporation Microfabricated compositions and processes for engineering tissues containing multiple cell types
US7875273B2 (en) 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US7413734B2 (en) * 2003-06-27 2008-08-19 Ethicon, Incorporated Treatment of retinitis pigmentosa with human umbilical cord cells
EP1660644A1 (en) 2003-08-29 2006-05-31 Regents Of The University Of Minnesota Kidney derived stem cells and methods for their isolation, differentiation and use
US20050089513A1 (en) 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
AU2004308511B2 (en) * 2003-12-23 2010-09-16 Fmc Biopolymer As Use of alginate matrices to control cell growth
TWI276685B (en) 2003-12-30 2007-03-21 Ind Tech Res Inst Conditional medium for culturing Schwann cells
US7534606B2 (en) 2004-01-12 2009-05-19 National Health Research Institutes Placental stem cell and methods thereof
DE102004043256B4 (en) 2004-09-07 2013-09-19 Rheinische Friedrich-Wilhelms-Universität Bonn Scalable process for culturing undifferentiated stem cells in suspension
US8039258B2 (en) * 2004-09-28 2011-10-18 Ethicon, Inc. Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
JP2008518597A (en) 2004-10-29 2008-06-05 セントカー・インコーポレーテツド Chemically defined medium composition
US7655678B2 (en) * 2004-11-30 2010-02-02 Council of Scientfic & Industrial Research Pharmaceutical composition for the management of tumors
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
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
CA2589041C (en) * 2004-12-23 2019-08-20 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
EP1838842A2 (en) 2004-12-23 2007-10-03 Ethicon, Incorporated Treatment of osteochondral diseases using postpartum-derived cells and products thereof
EP1831355A2 (en) 2004-12-23 2007-09-12 Ethicon, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US7741311B2 (en) 2005-01-03 2010-06-22 Shaker Mousa Composition and method for treating occlusive vascular diseases, nerve regeneration, and wound healing
WO2006079854A1 (en) 2005-01-28 2006-08-03 Novathera Ltd Methods for embryonic stem cell culture
CN101589137B (en) 2005-03-31 2013-06-05 斯丹姆涅恩有限公司 Amnion-derived cell compositions, methods of making and uses thereof
US7923007B2 (en) 2005-08-08 2011-04-12 Academia Sinica Brain tissue damage therapies
PL1971681T3 (en) 2005-12-16 2018-01-31 Depuy Synthes Products Inc Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
AU2006327073B2 (en) 2005-12-19 2012-08-30 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
AU2006330409B2 (en) * 2005-12-28 2012-07-19 Ethicon, Incorporated Treatment of peripheral vascular disease using postpartum-derived cells
EP2471907B1 (en) 2005-12-29 2018-09-26 Celularity, Inc. Placental stem cell populations
JP5106416B2 (en) 2006-01-06 2012-12-26 アジレント・テクノロジーズ・インク Reaction buffer composition for nucleic acid replication comprising packed DNA polymerase
ES2549528T3 (en) 2006-03-23 2015-10-29 Pluristem Ltd. Methods of cell expansion and uses of cells and conditioning media produced by them for therapy
WO2007146105A2 (en) * 2006-06-05 2007-12-21 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
SI2078073T1 (en) * 2006-10-12 2013-11-29 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
EP2089511B1 (en) * 2006-11-13 2014-09-17 DePuy Synthes Products, LLC In vitro expansion of postpartum-derived cells using microcarriers
EP2915537A3 (en) 2007-02-12 2015-10-28 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US20080305148A1 (en) 2007-03-19 2008-12-11 National Yang Ming University Treatment of spinal injuries using human umbilical mesenchymal stem cells
BRPI0818183A2 (en) 2007-10-05 2019-07-16 Ethicon Inc Renal tissue repair and regeneration using cells derived from human umbilical cord tissue
US20090123620A1 (en) * 2007-11-14 2009-05-14 Hiti Thomas R Automated Application of an Antimycotic Composition to Sliced Foodstuffs and an Antimycotic Application Apparatus
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
WO2009086215A2 (en) 2007-12-21 2009-07-09 Wyeth Pathway analysis of cell culture phenotypes and uses thereof
AU2008343203B2 (en) 2007-12-27 2014-07-10 DePuy Synthes Products, LLC Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
TWI387135B (en) * 2008-03-28 2013-02-21 Ind Tech Res Inst Light emitting device and manufacturing method thereof
EP2379088B1 (en) 2008-12-19 2018-02-28 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
AU2009327383B2 (en) 2008-12-19 2014-08-28 DePuy Synthes Products, LLC Regeneration and repair of neural tissue following injury
AU2009327382B2 (en) 2008-12-19 2014-07-17 DePuy Synthes Products, LLC Umbilical cord tissue derived cells for treating neuropathic pain and spasticity
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
JP5908394B2 (en) 2009-03-26 2016-04-26 デピュイ・シンセス・プロダクツ・インコーポレイテッド Human umbilical cord tissue cells as a therapy for Alzheimer's disease
EP2666200A1 (en) * 2011-01-19 2013-11-27 E.I. Du Pont De Nemours And Company Lithium battery separator with shutdown function

Also Published As

Publication number Publication date
US10039793B2 (en) 2018-08-07
ES2600555T3 (en) 2017-02-09
PL2322599T3 (en) 2015-10-30
EP1641915B1 (en) 2016-07-27
EP1641913A2 (en) 2006-04-05
WO2005001079A2 (en) 2005-01-06
EP2322599B1 (en) 2015-04-15
US20170166864A1 (en) 2017-06-15
US20170166863A1 (en) 2017-06-15
EP1641918B1 (en) 2016-01-06
WO2005001077A2 (en) 2005-01-06
EP2322599A3 (en) 2011-07-27
AU2004281371B2 (en) 2012-05-10
US10195233B2 (en) 2019-02-05
US9504719B2 (en) 2016-11-29
CA2530533A1 (en) 2005-01-13
PL1641917T3 (en) 2016-11-30
JP4948165B2 (en) 2012-06-06
JP4948164B2 (en) 2012-06-06
EP2336298A3 (en) 2011-08-03
WO2005001076A2 (en) 2005-01-06
EP2338982B1 (en) 2015-08-26
EP2341131A3 (en) 2011-08-03
ES2550267T3 (en) 2015-11-05
AU2004252571A1 (en) 2005-01-06
WO2005001078A3 (en) 2005-04-21
WO2005001079A3 (en) 2005-04-28
US10758576B2 (en) 2020-09-01
EP2338981B1 (en) 2015-04-22
US9579351B2 (en) 2017-02-28
US20140045263A1 (en) 2014-02-13
US7413734B2 (en) 2008-08-19
PL2399990T3 (en) 2015-12-31
EP1649013B1 (en) 2016-01-20
AU2004252566A1 (en) 2005-01-06
PL2336298T3 (en) 2016-08-31
US8658152B2 (en) 2014-02-25
EP1641914A2 (en) 2006-04-05
US20050019865A1 (en) 2005-01-27
US20170326183A1 (en) 2017-11-16
JP4950661B2 (en) 2012-06-13
AU2004252568B2 (en) 2011-06-30
AU2004281371A1 (en) 2005-04-28
PL1641913T3 (en) 2016-06-30
CA2530421A1 (en) 2005-01-06
JP2007528706A (en) 2007-10-18
ES2542069T3 (en) 2015-07-30
US20060234376A1 (en) 2006-10-19
JP4950660B2 (en) 2012-06-13
JP4950659B2 (en) 2012-06-13
US9717763B2 (en) 2017-08-01
EP2338981A2 (en) 2011-06-29
CA2530732C (en) 2015-03-31
PL1641916T3 (en) 2016-08-31
US7510873B2 (en) 2009-03-31
US20180338999A1 (en) 2018-11-29
WO2005001080A3 (en) 2005-03-24
US8703121B2 (en) 2014-04-22
JP2007521009A (en) 2007-08-02
US20050058630A1 (en) 2005-03-17
CA2530421C (en) 2015-04-21
US11191789B2 (en) 2021-12-07
ES2597837T3 (en) 2017-01-23
AU2004254616B2 (en) 2011-09-01
EP1641917A2 (en) 2006-04-05
EP2341131A2 (en) 2011-07-06
JP5148873B2 (en) 2013-02-20
PL2341131T3 (en) 2016-01-29
EP1641917B1 (en) 2016-04-27
US7524489B2 (en) 2009-04-28
US10383898B2 (en) 2019-08-20
US20060153818A1 (en) 2006-07-13
EP1641914B1 (en) 2016-07-20
CA2530732A1 (en) 2005-01-06
AU2004252571B2 (en) 2011-09-15
US8277796B2 (en) 2012-10-02
EP2338980A3 (en) 2011-08-10
JP2007521793A (en) 2007-08-09
ES2565582T3 (en) 2016-04-05
AU2004254616C1 (en) 2012-03-01
US20060154367A1 (en) 2006-07-13
US20050054098A1 (en) 2005-03-10
US9498501B2 (en) 2016-11-22
CA2530255C (en) 2015-12-08
JP4948166B2 (en) 2012-06-06
ES2552226T3 (en) 2015-11-26
WO2005001078A2 (en) 2005-01-06
US10500234B2 (en) 2019-12-10
AU2004252571C1 (en) 2012-03-01
AU2004252567B2 (en) 2011-10-06
EP2338982A3 (en) 2011-08-03
EP2336298B1 (en) 2016-02-17
US11179422B2 (en) 2021-11-23
US20060153816A1 (en) 2006-07-13
US20100260843A1 (en) 2010-10-14
CA2530412C (en) 2016-02-09
US11000554B2 (en) 2021-05-11
US20100210013A1 (en) 2010-08-19
EP2338980A2 (en) 2011-06-29
EP2338982A2 (en) 2011-06-29
WO2005038012A3 (en) 2005-09-15
PL2338982T3 (en) 2016-01-29
WO2005001077A3 (en) 2005-03-03
PL1641914T3 (en) 2017-01-31
WO2005001076A3 (en) 2005-03-31
US20070036767A1 (en) 2007-02-15
US20050037491A1 (en) 2005-02-17
US20170143770A1 (en) 2017-05-25
US20170239299A1 (en) 2017-08-24
CA2530416C (en) 2015-08-25
AU2004252570A1 (en) 2005-01-06
JP2007528705A (en) 2007-10-18
ES2554343T3 (en) 2015-12-18
WO2005038012A2 (en) 2005-04-28
EP1641918A2 (en) 2006-04-05
JP2007521008A (en) 2007-08-02
JP2007528702A (en) 2007-10-18
WO2005001080A2 (en) 2005-01-06
US20190192577A1 (en) 2019-06-27
US20050058631A1 (en) 2005-03-17
ES2569780T3 (en) 2016-05-12
US20070014771A1 (en) 2007-01-18
US10220059B2 (en) 2019-03-05
ES2582342T3 (en) 2016-09-12
WO2005003334A2 (en) 2005-01-13
ES2564045T3 (en) 2016-03-17
WO2005003334A3 (en) 2005-04-07
CA2530412A1 (en) 2005-04-28
EP1641913B1 (en) 2016-01-06
PL1649013T3 (en) 2016-07-29
PL2338980T3 (en) 2016-04-29
EP1641916A2 (en) 2006-04-05
AU2004252566B2 (en) 2011-09-15
JP2007528703A (en) 2007-10-18
US20060153817A1 (en) 2006-07-13
CA2530422A1 (en) 2005-01-06
EP2336298A2 (en) 2011-06-22
CA2530255A1 (en) 2005-01-06
AU2004252567A1 (en) 2005-01-06
AU2004252570B2 (en) 2011-09-15
US20060154366A1 (en) 2006-07-13
ES2541604T3 (en) 2015-07-22
AU2004252568A1 (en) 2005-01-06
US20050058629A1 (en) 2005-03-17
US20060188983A1 (en) 2006-08-24
PL1641918T3 (en) 2016-06-30
AU2004281371C1 (en) 2012-10-11
EP1641916B1 (en) 2016-02-17
US20130022585A1 (en) 2013-01-24
EP2338980B1 (en) 2015-04-22
ES2542070T3 (en) 2015-07-30
EP2341131B1 (en) 2015-08-05
US7560276B2 (en) 2009-07-14
EP2399990A1 (en) 2011-12-28
EP2338981A3 (en) 2011-08-03
PL1641915T3 (en) 2017-01-31
AU2004254616A1 (en) 2005-01-13
PL2338981T3 (en) 2016-01-29
US20070009494A1 (en) 2007-01-11
US20050032209A1 (en) 2005-02-10
AU2004252570C1 (en) 2012-03-01
ES2564044T3 (en) 2016-03-17
ES2568463T3 (en) 2016-04-29
EP2322599A2 (en) 2011-05-18
CA2530416A1 (en) 2005-01-06
US8318483B2 (en) 2012-11-27
EP1641915A2 (en) 2006-04-05
EP1649013A2 (en) 2006-04-26
EP2399990B1 (en) 2015-07-22
CA2530533C (en) 2015-02-10

Similar Documents

Publication Publication Date Title
CA2530422C (en) Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
EP1831356B1 (en) Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20060171930A1 (en) Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20220627