CA2530422A1 - 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
CA2530422A1
CA2530422A1 CA002530422A CA2530422A CA2530422A1 CA 2530422 A1 CA2530422 A1 CA 2530422A1 CA 002530422 A CA002530422 A CA 002530422A CA 2530422 A CA2530422 A CA 2530422A CA 2530422 A1 CA2530422 A1 CA 2530422A1
Authority
CA
Canada
Prior art keywords
cells
cell
derived
postpartum
protein
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.)
Granted
Application number
CA002530422A
Other languages
French (fr)
Other versions
CA2530422C (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
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
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

POSTPARTUM-DERIVED CELLS FOR USE IN TREATMENT OF DISEASE OF THE
HEART AND CIRCULATORY SYSTEM
CROSS-REFERENCE TO RELATED APPLICATIONS
This claims benefit of U.S. Provisional Application Serial No. 60/483,264, filed June 27, 2003, the entire contents of which are incorporated by reference herein. Qther related applications include the following commonly-owned, co-pending applications, the entire contents of each of which are incorporated by reference herein: U.S.
Application No. [ETH-5073 NP1], filed June 25, 2004; U.S. Application No. [ETH-5073 NP2], filed June 25, 2004;
U.S. Application No. [ETH-5073 NP3], filed June 25, 2004; U.S. Application No.
[ETH-5073 NP4], filed June 25, 2004; U.S. Application No. [ETH-5073 NPS], filed June 25, 2004; U.S.
Application No. [ETH-5073 NP8], filed June 25, 2004; and U.S. Provisional Application No.
60/555,908, filed March 24, 2004.
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 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, polyornithine, vitronectin, or fibronectin;
production of at least one of tissue factor, vimentin, 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, CDl 17, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry;
expression of at least one of interleulcin 8; reticulon 1; chemokine (C-X-C
motif) ligand 1 (melonoma growth stimulating activity, alpha); chemolcine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemol~ine (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 dehydrogenase 1 family, member A2; and renin; oxidized low density lipoprotein (lectin-like) receptor 1; Horno Sapiens, clone IMAGE:4179671, mRNA, partial cds; protein kinase 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 ileac 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 mRNA; cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (Drosophila); ciystallin, 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 l, anti-proliferative; cholesterol 25-hydroxylase; runt-related transcription factor 3; hypothetical protein FLJ23191; interleulcin 11 receptor, alpha;
procollagen C-endopeptidase enhancer; frizzled homolog 7 (DYOSOphila);
hypothetical gene BC008967; collagen, type VIII, alpha l; tenascin C (hexabrachion); Iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo Sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytolcine 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 (D~osophila); 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 E1B l9kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
insulin-like growth factor binding protein 2, 361cDa;
secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP 1 a, R.ANTES, and TIMP 1; and lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIPlb, I309, 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 UMB 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 lcaryotype, which is maintained as the cells are passaged.

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 Garner 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 endothelimn, 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.
Purl~inje 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.

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 of 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 lilce. Thereapeutic compositions can also comprise additional biological compounds or pharamaceuticals 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 sapieras, clone IMAGE:4179671, mRNA, partial cds; protein kinase C, zeta;
hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; Homo sapieras 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 ileac crest bone marrow cell, for at least one of 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 DI~FZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (D~osophila); crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DI~FZP586B2420 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 (D~osophila);
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;
DI~FZP586L151 protein; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (D~osophila); I~IAA1034 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); Horno 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 l9kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit VIIa 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, TP O, MIP 1 a, R.ANTES, and TIIVVIP 1; and do not secrete at least one of TGF-beta2, ANG2, PDGFbb, MIPlb, I309, 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 aclininistering 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.
_7_ 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 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 worlc, 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 ih 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 inj ected 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 achninistering 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 _g_ 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 ih vitro or iya 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 axe provided herein. Cell lysates, soluble cell fractions and membrane-enriched cell fractions axe 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, lcits for isolation of the cells, kits for coculturing the cells, bits for _g_ utilizing the cells and culture in vitYO 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.
These and other aspects of the invention are described in more detail below.
DETAILED DESCRIPTION
Defirzitioras Various terms used throughout the specification and claims are defined as set forth below.
Stem 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 vitYO 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) plu>~ipotent; (3) znultipotezzt; (4) oligopotent; and (5) unipotezzt.
Totipotent cells are able to give rise to all embryonic and extraembryonic cell types. Plus°ipotent 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 emb~onic stern 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. Apostpaz-tuzn stern cell is a multipotent or pluripotent cell that 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 extraembryomic membranes (chorion, amnion, yollc 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 ("committed") 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 of progenitor cell may be used. 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. Tlus type of progenitor cell is generally not able to self renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a raon-renewirag progenitor cell or as an iraternZediate progenitor or precursor cell.
As used herein, the phrase differentiates into a mesoder~fraal, ectodermal or eradodernral lineage refers to a cell that becomes committed to a specific mesoderrnal, 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 endodermal 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 andlor 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 prifraary cell culture is a culture of cells, tissues, or organs taken directly from an organisms) 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 subcultivations 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 rnay be referred to as a PIO
culture. The primary culture, i.e., the first culture following the isolation of cells from tissue, is designated P0. 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 tine 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 ti°ophic 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 ~eplica~ive senescence or cellular senescence) refers to a property attributable to finite cell cultuxes; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayfliclz'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 ntrogen 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.

As used herein, the term 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 term standard growth conditions, as used herein refers to culturing of cells at 37°C, in a standard atmosphere comprising 5% C02,. 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;
C:D for collagenase and dispase treatment C:D:Hfor collagenase, dispase, and hyaluronidase treatment CKl8 for cytolceratin 18;
CXC ligand 3 for chemol~ine receptor ligand 3;

DAPI for 4' - 6-Diamidino-2-phenylindole-2HCl DMEM for Dulbecco's Minimal Essential Medium;
DMEM.~Ig (or DMEM.~Lg, DMEM.~LG) for DMEM with low glucose;
EDTA for ethylene diamine tetraacetic acid;
EGF for epidermal growth factor;
FACS 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;
HNP 1 alpha for hepatocyte-specific transcription factor;
HUYEC 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. I309 binds to endothelial cells, stimulates chemotaxis and invasion of these cells, and enhances HWEC differentiation into capillary-like structures in an in vits°o Matrigel assay. Furthermore, I309 is an inducer of angiogenesis in vivo in both the rabbit cornea and the chiclc chorioallantoic membrane assay (CAM).
IL-6 for interleul~in-6;
IL-8 for interleukin-8;
Kl9 for keratin 19;
K8 for keratin 8;
KGF for lceratinocyte growth factor;
MCP-1 for monocyte chemotactic protein 1;
MDC for macrophage-derived chemoleine;
MIPI alpha for macrophage inflammatory protein 1 alpha;
MIPI beta 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;

PBMC for Peripheral blood mononuclear cell;
PBS for phosphate buffered saline;
PDGFbb for platelet derived growth factor;
PDGFrlalpha for platelet derived growth factor receptor alpha;
PD-L2 for programmed - death ligand 2;
PO (or po) for "per os" (by mouth);
PPDC for postpartum-derived cell;
Rantes (or RANTES) for regulated on activation, normal T cell expressed and secreted;
~laGDF 5 for recombinant human growth and differentiation factor 5;
SC for subcutaneously;
SDF 1 alpha for stromal-derived factor 1 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;
TIMPl for tissue inhibitor of matrix metalloproteinase 1;
TPO for thrombopoietin;
TuJl for BIII Tubulin;
UDC for umbilicus-derived cell;
T~EGF for vascular endothelial growth factor;
vWF for von Willebrand factor;
alplaaFP for alpha-fetoprotein;
Descriptio~z Various articles and references are cited herein and throughout, each of which is hereby incorporated in its entirety by such reference.
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 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 banl~ed 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, vitTOnectin, 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, vimentin, 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 is also preferred. The cells are also characterized in their laclc of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-Id2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; more preferable cells laclc production of all of these surface marlcers. Also preferred are cells which express at least one of W terleukin 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. 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); Wilins tumor 1;
aldehyde dehydrogenase 1 family, member A2; and resin; oxidized low density lipoprotein (lectin-like) receptor 1; Homo sapiefas, clone IMAGE:4179671, mRNA, partial cds; protein l~inase 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 mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of short stature homeobox 2;
heat shoclc 27kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1);
elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapieras mRNA; cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (D~osophila); 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; interleul~in 11 receptor, alpha;
procollagen C-endopeptidase eWancer; frizzled homolog 7 (D~osophila);
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); Iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiesZS cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-lilce factor 1; potassium interrnediate/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 l;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Ho~ao sapieras mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KTA_A_0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Hor~2o sapiens mRNA; cDNA DI~FZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus E1B l9kDa 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, chemol~ines, 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 TIMP1; preferred cells may alternatively be characterized in their lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIPlb, I309, 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 spilled 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 lacl~ing production of at least five, six, seven or eight or more of these markers are also preferred. More preferred are 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 interleul~in 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.
W 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 sapieras, clone IMAGE:4179671, mRNA, partial cds; protein l~inase C, zeta; hypothetical protein DKFZp564F013;
downregulated in ovarian cancer 1; Homo Sapiens mRNA; and cDNA DKFZp547K1113 (from clone DKF'Zp547K1113). 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 axe cells, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, have reduced expression for at least one of the genes corresponding to: 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); Horno Sapiens mRNA;
cDNA
DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeobox 2 (growth arrest-specific homeobox); sine oculis homeobox homolog 1 (D~osophila); crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein;
similar to neuralin l; 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; interleul~in 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 (Dr~osophila); 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-lilce 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 l9kDa interacting protein 3-like; AE binding protein l;
cytochrome c oxidase submit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity 1;
insulin-like growth factor binding protein 2, 361~Da. More preferred are cells that have, relative to human fibroblasts, mesenchymal stem cells, or ileac crest bone marrow cells, reduced expression of at least 5, 10, 1-5 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 ileac 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, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIPla, RANTES, and TIMP1. 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, MIPIb, I309, MDC, 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.

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.
deoxyribonucleases and the neutral protease, disease) 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 collagenase and disease, or collagenase, disease, and hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral protease disease are used in the dissociating step. Presently preferred axe 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 isolated in the presence of one or more activities of collagenase, hyaluronidase and disease.
More preferred are those cells isolated in the presence of a collagenase from Clost~~idium laistolyticum, and either of the protease activities, disease and thermolysin.
Still more preferred are cells isolated with collagenase and disease enzyme activities. Also preferred are such cells isolated in the presence of a hyaluronidase activity, in addition to collagenase and disease 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, LIBERASE Blendzymes available from Roche Diagnostics. The skilled artisan will appreciate the methods for optimizing enzyne 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 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 further wherein the cells express each of the markers CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C, wherein the cells produce the irmnunologically-detectable proteins vcrhich 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 determined 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% postp artum=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.

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, ifz 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 rej ection. 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 malting 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 tamers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions. Cell lysates may be used iya vitro or ifz vivo, alone or, for example, with sylgeneic or autologous live cells. The lysates, if introduced i~z 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 vasculogenic lines. Cell lysates from differentiated cells, or cells more committed than the PPDCs are also desirable. For example, lysates from cells with characteristics of cardiomyoblasts, cardiomyocytes, 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, hemangiogeW c, 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, FGF, TAK, GATA, Csx, NIA, 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 of 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 nyosin, 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 differentiated cells acquiring two or more of the indicia of cardiomyogenic, 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 lilce 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 l, 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 ifa 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 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 iWibitors, 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 factors, , 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 PPaclc (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 lmown 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 axe 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 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.

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, inj ection, surgical attachment, transplantation with other tissue, inj ection, 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 ifa vivo, where the seeded cells may proliferate on or in the frameworlc or help establish replacement tissue iya 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 irr. 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, lceratinocytes, and other soft tissue type progenitors, including stem cells. When grown in this three-dimensional system, the proliferating cells 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 vita°o, at which point, the matrix is degraded. A review of matrix design is provided by Hutmacher, J.
Biomat. Sci.
Polymef° 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 10~ to 5 x 10~ 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 microenviromnent found i~c vivo, such that the extent to which the cells are grown prior to implantation in vivo or use ih 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 iya 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 b a used for aspects of the invention include mats (woven, knitted, and more preferably nonwoven) porous or semiporous 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, formed 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 iya situ polymerizable gels, and hydrogels, for example composed of self assembling peptides. These materials are frequently used as supports for growth of tissue. Ifz 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, Biomatef°ials 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, PLA, 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 frameworlc 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 frameworlc 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 cellulax 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 treatments include but are not limited to natural materials such as basement membrane proteins such as laminin and Type IV collagen, synthetic materials such as ePTFE, and segmented polyurethaneurea silicones, such as PURSPAN (The Polymer Technology Group, Inc., Berkeley, 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 iya 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 desiref. 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 framev~ork 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 frauneworks 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 i~ 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, both of which are incorporated herein by reference in its entirety.
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 frameworlcs 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 franework 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, migratiori, and extracellular matrix deposition. Examples of these materials and treatments include but are not limited to natural materials such as basement membrane proteins such as laminin and Type IV
collagen, synthetic materials such as ePTFE, and segmented polyurethaneurea silicones, such as PURSPAN (The Polymer Technology Group, Inc., Berkeley, 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 implaritable devices. For example the cells, cell lysates and cell fractions can be coadminstered with, for example stems, artificial valves, ventricular assist devices, Gugliehni 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 inplantable devices, to minimize problems when they are used ih 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 rnotif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemolcine (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 mesenchymal stem cell, or an ileac 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); ~Iorfao 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 l; 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 (Df°osopl~ila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); ixoquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Ho~rao 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 (DYOSOplzila); 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-lilce repeat domains); Homo sapiehs mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; I~IAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiefzs mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1;
BCL2/adenovirus E1B l9kDa interacting protein 3-like; AE binding protein l;
cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); neuroblastoma, suppression of tumorigenicity l;
insulin-like growth factor binding protein 2, 36kDa.
PrefeiTed therapeutic cell compositions also comprise cells which secrete at least one of MCP-1, I-L-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIPla, R.ANTES;
and TIMP 1; and do not secrete at least one of TGF-beta2, ANG2, PDGFbb, MIPlb, I309, 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 (mural valve prolapse), bradycardia, Buerger's Disease (thromboangiitis obliterans), cardiomegaly, cardiomyopathy, carditis, carotid artery disease, coarctation of the aorta, congenital heart diseases (congental 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, tuuncus 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 rhemnatic 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, pulinonary embolism, chronic pulmonary heart disease), kyphoscoliotic heart disease, pericarditis, myocarditis, endocarditis, endomyocardial fibrosis, endocardial fibroelastosis, atrioventricular bloclc, 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), ej ection fraction (EF), left ventricular ej ection 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 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 life may be shovm 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 achninistering 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, chemol~ines, cytolcines and the lilce, 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 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 asry 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 bits for use in the treatment of myocardial infarction.
The bits provide the therapeutic cell composition which can be prep ared in a pharmaceutically acceptable form, 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, card><omyoblasts, cardiomyocytes, or progenitors of myoblasts, myocytes, cardiomyoblasts, or cardiomyocytes are used.

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 admsnistered as a matrix-cell complex. In certain embodiments, the matrix is a scaffold, preferably bioabsorbable, comprising at least the postpartum-derived cells.
Fits 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 bits include a therapeutic cell composition, with or without a matrix, and with or without a coculture present. The bits also optionally include a means of administering the cells, for example by inj ection, and a pharmaceutically-acceptable carrier for the cells, if required. The bits include instructions for use of the cells. Fits prepared for field hospital use, such as for military use may include full-procedure supplies including tissue scaffolds, surgical sutures, and the life, 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 irmnunological, biochemical and genetic properties of the cells. The cells so frozen 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 nay be preferable to use cells freshly prepared, the remainder can be cryopreserved and bnked 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.

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 pants 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 form), antibiotic compounds, hormones, and the like. Fits for growth can for example include all of the components of the Growth Medium as used herein, including serum, for example fetal bovine senam. 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. Fits 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 bai~lc 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 alos 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 lcits 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 lcits 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 ira vitYO methods as described herein may contain one or more of (1) UDCs or fractions, components or products of UDCs, (2) reagents for practicing the in vita°o method, (3) other cells or cell populations, as appropriate, for example for cocultures and (4) instructions for conducting the in vity~o 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 fiaction 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 life.
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-term placental and umbilical cord tissues. A highly preferred way of isolating cells from these tissues is by using a combination of digestive enzymes. Particularly preferred are collagenase, hyaluronidase and disease. 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 fiom placental and umbilical cord tissues were isolated.
Postpartum umbilicus and placenta were obtained upon births of either full- 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 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 cmz 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 enzymes. In some experiments an enzyne mixture of collagenase and disease was used ("C:D;"
collagenase (Sigma, St Louis, MO), 500 Units/milliliter; and disease (Invitrogen), 50 Unitslmilliliter in DMEM:-Low glucose medium). hl other experiments a mixture of collagenase, disease and hyaluronidase ("C:D:H") was used (collagenase, 500 Units/milliliter;
disease, 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, N~ 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 (FBS; defined bovine serum;
Lot#AND1 X475;
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 volurne 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 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 Celllsolatioya 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/rnilliliter;
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 (Invitrogen), 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 enzymes. In some experiments an enzyme mixture of collagenase and disease ("C:D") was used containing collagenase (Sigma, St Louis, MO) at 500 Units/milliliter and disease (Invitrogen) at 50 Units/milliliter in DMEM:-Low glucose medium. In other experiments, a mixture of collagenase, disease and hyaluronidase (C:D:H) was used (collagenase, 500 Units/milliliter;

dispase, 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 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 supernatant 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 Celllsolatiora Cells were isolated from postpartum tissues in DMEM-Low glucose medium with LIBERASE (2.5 milligrams per milliliter, Blendzyme 3; (Roche Applied Sciences, Indianapolis, IN)) and hyaluroudase (5 Units/milliliter, Sigma). Digestion of the tissue and isolation of the cells was as described for other digestions above, with the LIBERASE/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) dispase; iii) hyaluronidase; iv) collagenase:dispase mixture (C:D); v) collagenase:hyaluronidase mixture (C:H); vi) dispase: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 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 US0229971 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) contaiung 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 Dicl~inson, Bedford, MA).
Isolation of postpartum cells using different efazyme combifzations 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 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 banlced for later investigation.
Results Cell isolation using diffe~e~zt 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 efzzyme combinations and g~omth 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 banged for further investigation.
Isolation of cells from residual blood in the co~~ds Nucleated cells attached and grew rapidly. These cells were analyzed by flow cytometry and were similar to cells obtained by enzyme digestion.
Isolatiofz. of cells from Coed Blood The preparations contained red blood cells acid platelets. No nucleated cells attached and divided during the first 3 weeps. The medium was changed 3 weelcs 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), dispase (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.

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 s1. HO, Tony, W. ;I~OPEN, Gene, C. ;RIGHTER, William, F. ;RUTI~OWSKI, 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 + (>10 h) +

Hyaluronidase X X

Collagenase:Dispase ++ (< 3 h) ++

Collagenase:Hyaluronidase ++ (< 3 h) +

Dispase:Hyaluronidase + (>10 h) +

Collagenase:Dispase:Hyaluronidase+++ (< 3 h) +++

Key: + = good, ++ = very good, +++ = excellent,,x = no success Table 1-2: Isolation and culture expansion of postpartum cells under varying conditions:
ConditionMedium 15% FBS BME Gelatin 20% OZ 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 (2,0 ng/rni lliliter) 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/rni lliliter) 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 quantities 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 demonstrate that postpartum cell lines as developeel 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 (Hayfliclc L. The longevity of cultured human cells. J. Am. Gef°iat~.
Soc. 22(1):1-12, 1974;
Hayflick L. The strategy of senescence. Ge~ontologist 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.
CompaYison of expansion poteyatial ofpostpaYtum 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 B1; 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 cellslcm2 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.

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)/ln2] and doubling time (time in culture (h)/population doubling) were calculated. For the purposes of determirung 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 bazz7~s at low density The expansion potential of cells banked at passage 10 was also tested. A
different set of conditions was used. Normal dermal slcin 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 hypan 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)/ln2) 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 postpaz~tuzn cells at low de>zsity frofzz 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/cna and passaged as described above until senescence. Cells were grown under standard atmospheric conditions at 37°C. Growth Medium was changed twice per weelc. Cells were passaged as they reached about 85%
confluence. At each passage, cells were trypsinized and counted by trypan blue staining. The cell yield, population doubling (ln (cell final/cell initial)/ln2) 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 flaslcs.
Expazzsiozz of Clorzal Neozzatal Placeyztal 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 kaiyotype-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.
Expa>zsion of cells iza low oxygen cultuzre cozzditiozzs It has been demonstrated that low OZ 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.
US20040005704). lii 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% OZ) culture conditions.
Other growth cozzditiozzs 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.

Results Comparison of expansion potential of postpar~tuna cells vs. otlzen stem cell and noyz-stern cell populations Both umbilical-derived and placenta-derived cells expanded for greater than 40 passages generating cell yields of > 1E17 cells in 60 days. hl 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.SE12 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 > lEl l 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/cma) 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 flasles 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, 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.
Karyotypimg confines that clones so-derived are neotal or maternal in nature.
Expansion of cells ih low oxygen culture conditions Cells expanded well under the reduced oxygen conditions, however, culturing urider 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/cmz). Postpartum-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion p ~tential 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, postpa~.-tum-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:

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. JAm Gey~iatr Soc. 1974 Jan;22(1):1-12.
2) Hayfliclc L. The strategy of senescence. Ge~oratologist. 1974 Feb;l4(1):37-45.
3) Patent US20040058412 4) Patent US20040048372
6) Csete, Marie; (Ann Arbor, MI) ; Doyle, John; (South Pasadena, CA) ; Wold, Barbaxa 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 PopulationTotal Cell Doublings Yield MSc 24 d 8 4.72 E7 Adipose-derived57 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 PopulationTotal Cell Doublings Yield ' Fibroblast 80 d 43.68 2.59 E11 (P10) Umbilical 80 d 53.6 1.25 E14 (P10) Placental 60 d 32.96 6.09 E12 (P10) Growth of Postpartum cells in Medium containing D-Valine Summary Culture media containing D-valise instead of the L-valise isoform reportedly selectively inhibit the growth of fibroblast-like cells in culture. To determine whether postpartum-derived cells can grow in medium containing D-valise, cells derived from placenta and umbilical cord were grown in medium containing D-valise for 4 weeks. The cells did not proliferate and eventually died. Medium containing D-valise is not suitable for selectively growing postparturn-derived cells. L-valise is required for postpartum-derived cell proliferation and long-term viability.
Introduction It has been reported that medium containing D-valise instead of the normal L-valise 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-valise.
Methods & Materials Placenta-derived cells (P3), fibroblasts (P9) and umbilical-derived cells (PS) 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-valise (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 weelcs.
Discussion and Conclusion 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 Iht 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 ahd 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 flaslc (Coming, Corning, NY) and expanded to 80%
confluence. A T25 flaslc 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 lcaryotype 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 vcrere counted and analyzed when a heterogeneous karyotype number (four) was identified.

Results All cell samples sent for chromosome analysis were interpreted by the cytogenetlcs laboratory staff as exhibiting a normal appearance. Three of the sixteen cell lines analy2ed exhibited a heterogeneous phenotype (XX and XY) indicating the presence of cells derived from both neonatal and maternal origins ~Tabie 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.
issue passagea ap ase ce a ap ase ce m er aryo ypa s coun a s ana yze o aryo yp acenta 2 m i ica m i ica acen a m i ica acen a-acen a-acen a-acen a-acen a-acen a-acen a-acen a-acenta-acen a-ICey: N- Neonatal aspect; V- vinous 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 kaiyotype.

Evaluation of Postpartum-Deriyed 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 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, CD11'7, 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.
Cultuy-e Tlessels Cells were cultured in plasma-treated T75, T150, and T225 tissue culture flasks (Coming, 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 Staiuiug 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 1x10' 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

ana centritugea to remove unnouna annbody. Cells were resuspended in 500 microliters PBS
and analyzed by flow cytometry.
Flow Cytometry Afzalvsis Flow cytometry analysis was performed with a FACScalibur instrument (Becton Diclcinson, San Jose, CA).
Aratibodies to Cell Surface Markers The following antibodies to cell surface markers were used.
Antibody Manufacture Catalog Number CD10 BD Pharmingen (San Diego,555375 CA) 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 CD 117 BD Pharmingen 340529 CD141 BD Pharmingen 559781 PDGFr-alpha BD Pharmingen 556002 HLA-A, B, C BD Pharmingen 555553 HLA-DR, DP, DQBD 555558 Pharmingen IgG-FITC Sigma (St. Louis, M~) F-6522 IgG-PE Sigma P-4685 Placefata and Zlmbilicus ComparisofZ
Placenta cells were compared to umbilicus at passage 8.
Passage to Passage Compariso~z Placenta and umbilicus were analyzed at passages 8, 15, and 20.
Donor to Dofzor 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 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 Cornparisoya Four treatments used for isolation and preparation of cells were compared.
Cells isolated from placenta by treatment with 1) collagenase; 2) collagenase/dispase; 3) collagenase/hyaluronidase; and 4) collagenase/hyaluroudase/dispase were compared.
Placental Layers Compa~isa~
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 Compa~-isora- Placenta Placenta-derived cells at passages 8, 15, and 20 analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha and HLA-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.

Passage to Passage Compaf°ison- Umbilicus Umbilical cells at passage 8, 15, and 20 analyzed by flow cytometiy 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, CD 117, CD 141, 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.
Donoj° to Doyao~~ CompaYison- Placenta Placenta-derived cells isolated from separate donors analyzed by flow cytornetry 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.
Doho~ to Donor Compa~isoh- 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.
Tlae Effect of Surface Coating with Gelatin oh Placenta-derived Cells Placenta-derived cells expanded on either gelatin-coated or uncoated flasl~s 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

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 Coatifag with Gelatin on Umbilicus-derived Cells Umbilical cells expanded on gelatin and uncoated flasl~s analyzed by flow cytometry a_ll 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 Digestiofa Procedure Used for Ps°eparatioh. and Isolation of the Cells Effect the Cell Surface Maf°ker 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 CD31, 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.
Placefatal Layer Comparison Cells isolated from the maternal, vinous, 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 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, DQ. 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 wluch has positive expression of the markers.

Analysis of Postpartum Tissue-Derived Cells using Oli~onucleotide 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 afzd Methods Isolation and Culture of Cells Postpa~~tmn tissue-derived cells. Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, PA) from nornal full term deliveries with patient consent. The tissues were received arid 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.
Fib~~oblasts. Human dernal fibroblasts were purchased from Cambrex Incoiporated (Wallcersville, 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 s erum (Hyclone) and penicillin/streptomycin (Invitrogen). The cells were gromn on standard tissue-treated plastic.
Human Mesenchynaal Stern 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% C02.

Human Ileac Cnest Bone Mart°ow 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, et al. (W003/025149). The marrow was mixed with lysis buffer (155 millimolar NH4C1, 10 millimolar I~HC03, and 0.1 millimolar EDTA, pH 7.2) at a ratio of 1 part bone manow to 20 parts lysis buffer. The cell suspension was voi-texed, 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 W
able 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 OZ or at 5% OZ. 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 mRNA and GENECHIP Analysis. Actively growing cultures of cells were removed from the flasks with a cell scraper in cold phosphate buffered saline WPBS). 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 mRNA was extracted and transcribed into cDNA. cDNA was then transcribed into cRNA and biotin-labeled.
The biotin-labeled cRNA was hybridized with Affymetrix GENECHIP HG-U133A oligonucleotide arrays (Affylnetrix, Santa Clara CA). The hybridizations and data collection were perforned according to the manufacturer's specifications. The hybridization 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). Licenses for the analysis software are available through the Office of Technology Licensing, Stanford University, and more information is available on the World Wide Web at Professor Tibshirani's web site in the Dept of Statistics, Stanford University (www-stat.stanford.edul tibslSAM~.
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.

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 SubstrateMedia Umbilical (022803)2 Gelatin DMEM, 15% FBS, ~iME

Umbilical (042103)3 Gelatin DMEM, 15% FBS, (3ME

Umbilical (071003)4 Gelatin DMEM, 15% FBS, [3ME

Placenta (042203) 12 Gelatin DMEM, 15% FBS, [3ME

Placenta (042903) 4 Gelatin DMEM, 15% FBS, (3ME

Placenta (071003) 3 Gelatin DMEM, 15% FBS, (3ME

ICBM (070203) (5% 3 Plastic MEM 10% FBS
02) ICBM (062703) (std5 Plastic MEM 10% FBS
02) ICBM (062703 )(5% 5 Plastic MEM 10% FBS
02) 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 Paif~ 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 MS C-Placenta 42. 84 MSC-Umbilical 46.86 ICBM-placenta 48.41 Tables 6-3, 6-4, and 6-5 show the expression of genes increased in placenta-derived cells (Table 6-3), increased in mnbilical 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 Probe Set Gene Name NCBI Accession ID Numb er 209732 C-type (calcium dependent, carbohydrate-recognitionAF070642 at domain) - lectin, superfamily member 2 (activation-induced) 206067 Wilms tumor 1 NM 024426 s at 207016 aldehyde dehydrogenase 1 family, member AB015228 s at A2 206367 Renin NM 000537 at 210004 oxidized low density lipoprotein (lectin-like)AF035776 at receptor 1 214993 Homo sapiens, clone IMAGE:4179671, mRNA, AF070642 at partial cds 202178 protein kinase C, zeta NM 002744 at 209780 hypothetical protein DKFZp564F013 AL136883 at 204135 downregulated in ovarian cancer 1 NM 014890 at 213542 Homo sapiens mRNA; cDNA DKFZp547K1113 AI246730 at (from clone - 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 Probe Set Gene Name NCBI Accession ID Number 202859 Interleukin 8 NM 000584 x at 211506 Interleukin 8 AF043337 s at 210222 reticulon 1 BC000314 s at chemokine (C-X-C motif) ligand 1 (melanoma 204470 growth NM 001511 at stimulating activity 206336 chemokine (C-X-C motif) ligand 6 (granulocyteNM 002993 at chemotactic -- protein 2) 207850 Chemokine (C-X-C motif) ligand 3 NM 002090 at 203485 reticulon 1 NM 021136 at 202644 tumor necrosis factor, alpha-induced proteinNM 006290 s at 3 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 NCBI Accession ID Gene name Number 210135 short stature homeobox 2 AF022654.1 s at 205824 heat shock 27kDa protein 2 N M 001541.1 at 209687 chemokine (C-X-C motif) ligand 12 (stromal019495.1 at cell-derived factor 1) .

203666 chemokine (C-X-C motif) ligand 12 (stromalN M 000609.1 at cell-derived factor 1 ) 212670 elastin (supravalvular aortic stenosis, AA479278 at Williams-Beuren syndrome) 213381 Homo sapiens mRNA; cDNA DKFZp586M2022 Ng1149 at (from clone - DKFZp586M2022) 206201 mesenchyme homeobox 2 (growth arrest-specificN M 005924.1 s at homeobox) 205817 Sine oculis homeobox homolog 1 (Drosophila)N M 005982.1 at 209283 crystallin, alpha B AF007162.1 at 212793 dishevelled associated activator of morphogenesisBF513244 at 2 213488 DKFZP586B2420 protein AL050143.1 at 209763 similar to neuralin 1 AL049176 at 205200 Tetranectin (plasminogen binding protein)N M 003278.1 at 205743 src homology three (SH3) and cysteine N M 003149.1 at rich domain 200921 B-cell translocation gene 1, anti-proliferativeN M 001731.1 s at 206932 cholesterol 25-hydroxylase N M 003956.1 at 204198 runt-related transcription factor 3 AA541630 s at 219747 hypothetical protein FLJ23191 N M 024574.1 at 204773 Interleukin 11 receptor, alpha N M 004512.1 at 202465 Procollagen C-endopeptidase enhancer N M 002593.2 at 203706 Frizzled homolog 7 (Drosophila) N M 003507.1 s at 212736 hypothetical gene BC008967 BE299456 at 214587 Collagen, type VIII, alpha 1 BE877796 at 201645 Tenascin C (hexabrachion) N M 002160.1 at 210239 Iroquois homeobox protein 5 090304.1 at 203903 Hephaestin N M 014799.1 s at 205816 integrin, beta 8 N M 002214.1 at 203069 synaptic vesicle glycoprotein 2 N M 014849.1 at 213909 Horno sapiens cDNA FLJ12280 fis, clone AU147799 at MAMMA1001744 206315 cytokine receptor-like factor 1 NM 004750.1 at 204401 Potassium intermediate/small conductance NM 002250.1 at calcium-activated channel, -- subfamily N, member 4 216331 integrin, alpha 7 AK022548.1 at 209663 integrin, alpha 7 AF072132.1 s at 213125 DKFZP586L151 protein AW007573 at 202133 transcriptional co-activator with PDZ-bindingAA081084 at motif (TAZ) 206511 Sine oculis homeobox homolog 2 (Drosophila)NM 016932.1 s at 213435 KIAA1034 protein AB028957.1 at 206115 early growth response 3 NM 004430.1.
at 213707 distal-less homeobox 5 NM 005221.3 s at 218181 hypothetical protein FLJ20373 NM 017792.1 s-at 209160 aldo-keto reductase family 1, member C3 Ag018580.1 at (3-alpha hydroxysteroid - dehydrogenase, type II) 213905 Biglycan AA845258 x at 201261 Biglycan BC002416.1 x at 202132 transcriptional co-activator with PDZ-bindingAA081084 at motif (TAZ) 214701 fibronectin 1 AJ276395.1 s at 213791 Proenkephalin NM 006211.1 at 205422 Integrin, beta-like 1 (with EGF-like repeatNM 004791.1 s at domains) 214927 Homo Sapiens mRNA full length insert cDNA AL359052.1 at clone EUROIMAGE

206070 EphA3 AF213459.1 s at 212805 KIAA0367 protein AB002365.1 at 219789 natriu retic peptide receptor C/guanylate AI628360 at cyclase C (atrionatriuretic - peptide receptor C) 219054 hypothetical protein FLJ14054 NM 024563.1 at Homo sapiens mRNA; cDNA DKFZp564B222 (from 213429 clone AW025579 at DKFZp564B222) 204929 vesicle-associated membrane protein 5 (myobrevin)NM 006634.1 s at 201843 EGF-containing fibulin-like extracellular NM 004105.2 s at matrix protein 1 221478~at BCL2/adenovirus E1 B 19kDa interacting AL132665.1 protein 3-like 201792 AE binding protein 1 NM 001129.2 at 204570 cytochrome c oxidase subunit Vlla polypeptideNM 001864.1 at 1 (muscle) 201621 neuroblastoma, suppression of tumorigenicityNM 005380.1 at 1 202718 Insulin-like growth factor binding proteinNM 000597.1 at 2, 36kDa 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 sapie~s cDNA: FLJ23224 fi's, clone ADSU02206 dynein, cytoplasmic, intermediate polypeptide 1 anlcyrin 3, node of Ranvier (ankyrin G) inhibin, beta A (activin A, activin AB alpha polypeptide) ectonucleotide pyrophosphatase/phosphodiesterase 4 (putative function) I~IAA1053 protein microtubule-associated protein lA
zinc finger protein 41 HSPC019 protein Homo sapieyZS cDNA: FLJ23564 fis, clone LNG10773 HonZO sapieyzs mRNA; cDNA DI~FZp564A072 (from clone DI~FZp564A072) LIM protein (similar to rat protein kinase Gbinding enigma) inhibitor of l~appa light polypeptide gene enhancer in B-cells, kinase complex-associated protein hypothetical protein FLJ22004 Human (clone CTG-A4) mRNA sequence ESTs, Moderately similar to cytolcine receptor-like factor 2; cytokine receptor CRL2 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 FLJ22362 ~UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 3 (GaINAc-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 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/lymphoma 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.
T'he study also included two different lines of dermal fibroblasts, three lines of mesenchymal 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 confirned 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 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 l, interleukin-8, renin, reticulon, chemol~ine receptor ligand 3 (CXC ligand 3) and granulocyte chemotactic protein 2 (GCP-2).
Four of these genes (oxidized LDL receptor l, renin, reticulon a~Zd 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 Mediiun.
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 of cells derived from other sources (using an Affymetrix GENECHIP). Six "signature"
genes yvere identified: oxidized LDL receptor 1, interleul~in-8, renin, reticulon, chemolcine receptor 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 determine 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 Placenta-derived cells (three isolates, including one isolate predominately rieonatal as identified by lcaryotyping 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 cultuj°e fog ELISA assay.
Postpartum cells derived from placenta and umbilical cord, as well as human fibroblasts derived from human neonatal foreslcin 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 l5ml centrifuge tubes. After centrifugation at 150 x g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 rnl 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 houxs. 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/EDT1~
(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-treated water and stored at -80°C. RNA was reversed transcribed using random hexamers with the TaqMan 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 Assays-on-DemandTM 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 TaqMan 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 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).

Cof~ventioyaal PCR
Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Massachusetts, USA) to confirm the results fiom real-time PCR. PCR was performed using 2 microliters of cDNA solution, 1 ~ AmpliTaq Gold 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 resin (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 ~,M except for GAPDH which was 0.5 ~.M. GAPDH primers were the same as real-time PCR, except that the manufacturer's TaqMan probe 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:l)' A: 5'-AGAATGGAAAACTGGAATAGG -3' (SEQ ID N0:2) Renin S: 5'-TCTTCGATGCTTCGGATTCC -3' (SEQ ID N0:3) A: 5'-GAATTCTCGGAATCTCTGTTG -3' (SEQ ID NO:4) Reticulon S: 5'- TTACAAGCAGTGCAGAAAACC-3' (SEQ ID NO:S) A: 5'- AGTAAACATTGAAACCACAGCC-3' (SEQ ID NO:6) Interleukin-8 S: 5'- TCTGCAGCTCTGTGTGAAGG-3' (SEQ ID N0:7) A: 5'-CTTCAAAAACTTCTCCACAACC- 3' (SEQ ID N0:8) Chemokine (CXC) ligand 3 S: 5'- CCCACGCCACGCTCTCC-3' (SEQ ID N0:9) A: 5'-TCCTGTCAGTTGGTGCTCC -3' (SEQ ID NO:10) Imnam2ofluo~esceyace Postpartum cells were fixed with cold 4% (w/v) parafonnaldehyde (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.
Immunocytochemistry 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;

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 III; 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 Rl ; 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) Triton (Triton X-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 Rl), Triton X-100 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.
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 bloclc 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 Prob es) or donkey anti-goat IgG-FITC (1:150, Santa Cruz Biotech). Cultures were then washed and 10 ~,M 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).
Pf~eparation of cells for FRCS analysis Adherent cells 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 1x10 per 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 cy-tometry.
The following antibodies were used: oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROa (555042;
BD Phanningen, Bedford, MA), Mouse IgGl 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 Dicl~inson San Jose, CA).
Results Results of real-time PCR for selected "signature" genes performed 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 r analyzed by the ~OCT 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 cytolcine, 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.

When supernatants of cells grown in serum-free medium were examined for the presence of TL-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 hmnan dermal 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 (normal17.27_+8.63 Oz) Placenta Isolate 3 264.92+9.88 (low0z, W/O
BME) Results of the ELISA
assay for interleukin-8 (IL-8) performed on placenta-and umbilical cord-derived cells as well as human skin fibroblasts.
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 immnocytochemical 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 cytol~eratin 18.
Cells derived from the human umbilical cord at passage 0 were probed for the expression of selected proteins by irmnunocytochemical 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 l and reticulon. Complete results of that experiment are still pending.
Discussion and Conclusion Thus far concordance between gene expression levels measured by microarray 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 FAGS 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 microarray experiments. As additional protein experiments are complete, the relationships between mRNA and protein expression will be more comprehensively understood.

Tmmunohistochemical 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) parafonnaldehyde overnight at 4°C.
Tmmunohistochemistry 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 Rl; 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 bloclcs were then sectioned (10 ~m thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
Table 8-1: Summary of Primary Antibodies Used Antibody ConcentrationVendor Vimentin 1:500 Sigma, St. Louis, MO

Desmin (rb) 1:150 Sigma Desmin (m) 1:300 Chemicon, Temecula, CA

alpha smooth muscle 1:400 Sigma actin (SMA) Cytokeratin 18 (CK18)1:400 Sigma von Willebrand factor1:200 Sigma (vWF) CD34 III 1:100 DalcoCytonation, 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. T_m_m__unohistochemistry was performed similar to previous studies (e.g., Messina, et al. (2003) Exper. Neurol. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocl~ing solution containing 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 Rl), 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 Rl, NOGO-A), 3% (v/v) donl~ey 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 amti-rabbit IgG-Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG-FITC (1:150; Santa Cruz Biotech). Cultures were washed, and 10 ~,M 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 invented epi-fluorescent microscope ~Olynpus, 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. CKl8 and desmin yvere 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, cytol~eratin 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. Synap. 212:215-26), monocyte chemotactic protein 1 (MCP-1) (Salcedo et al. (2000) Blood 96;34-40), W
terleukin-8 (IL-8) ( Li et al. (2003) J. Irnfnuraol. 170:3369-76), keratinocyte growth factor (KGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) (Hughes et al.
(2004) Ann.
Tho~ac. 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 1 alpha (SDF-1 alpha)), neurotrophic/neuroprotective activity (brain-derived neurotrophic factor (BDNF) (Cheng et al. (2003) Dev.
Biol. 258;319-33), interleulcin-6 (IL-6), granulocyte chemotactic protein-2 (GCP-2), transforming growth factor beta2 (TGFbeta2)), or chemokine activity (macrophage inflammatory protein 1 alpha (MIP 1 a), macrophage inflammatory protein lbeta (MIPlb), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), I309, thymus and activation-regulated chemolcine (TARO), Eotaxin, macrophage-derived chemokina (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), penicilliustreptomycin (Gibco)) for 8 hours. Conditioned serum-free media was collected at the 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-1 alpha , GCP-2 , IL-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 (MIPla, MIPlb, MCP-1, Rantes, I309, TARC, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, TIMP1, ANG2, PDGF-bb, TPO, HB-EGF were measured using Searchhight 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 witlun 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 IL-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% Oa and by fibroblasts. GCP-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 IL-6 VEGF SDF-la GCP-2 IL-8 TGF-Fibroblast 17+1 61+3 29_+219_+1 21_+1 ND ND

Placenta (042303) 60+3 41+2 ND ND ND ND ND

Umbilical (022803) 1150_+744234_+289ND ND 160_+112058+145ND

Placenta (071003) 125_+1610+1 ND ND ND ND ND

Umbilical (071003) 2794+841356+43ND ND 2184+982369+23ND

_8~_ Placenta (101503) 21+10 67+3 ND ND 44+9 17+9 ND
BME

Placenta (101503) 77+16 339_+21ND 1149_+13754+2 265+10 ND
5% OZ, W/O

BME

ney: m~: mot Letectea.
Searchlight Multiplexed ELISA assay. TIIVIPl, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIPlb, MCP1, R.ANTES, I309, TARC, MDC, and IL-8 were secreted from umbilical cord-derived PPDCs (Tables 9-2 and 9-3). TIMP1, TPO, KGF, HGF, HBEGF, BDNF, MIPla, MCP-1, R.ANTES, 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. Searclil,ight Multiplexed ELISA assay results TIMP1 ANG2 PDGFbbTPO I~GF HGF FGF VEGF HBEGF BDNF

hFB 19306.3ND ND 230.5 5.0 ND ND 27.9 1.3 ND

P 24299.5ND ND 546.6 8.8 16.4 ND ND 3.81.3ND

U1 57718.4ND ND 1240.05.8 559.3148.7 ND 9.3 165.7 P3 14176.8ND ND 568.7 5.2 10.2 ND ND 1.9 33.6 U3 21850.0ND ND 1134.59.0 195.630.8 ND 5.4 388.6 Key: hr't3 (human ttbroblasts), Y1 (placenta-derived YYLW (u4LSU3)), u1 (umbtncat cord-aertvea rriW ; (uGG~us)), P3 (placenta-derived PPDC (071003)), U3 (umbilical cord-derived PPDC
(071003)). ND: Not Detected.
Table 9-3. Searchlight Multiplexed ELISA assay results MIPla MIPlb MCP1 RANTES I309 TARC EotaxinMDC IL8 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 U1 ND 8.0 1694.2ND 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 2018.741.5 11.6 21.4 ND 4.8 10515.9 trey: nits (numan tibrobiasts), ri (placenta-aertvea rrLU; (U4G3u3)), a i ~ummncai cora-aenvea rrm; ~u~~au3~~, 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.

Ifa vitro Immunology Postpartum cell lines were evaluated ifZ vitf~o for their immunological characteristics in an effort to predict the immunological response, if any, these cells would elicit upon ih vivo 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 & Lichtman, 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).Iounnal oflmmunological Methods 224, 185-196), and PD-L2 (Abbas & Lichtman, CELLULAR AND MOLECULAR
IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171; Brown, et. al.
(2003) The .Iou~nal 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 ute~o. 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).
Mate~~ials 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 TrypsinEDTA (Gibco, Carlsbad, MO). Cells were harvested, centrifuged, and re-suspended in 3% (vlv) FBS in PBS at a cell concentration of 1x10 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 centriftiged 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, 557227 CA) CD86 BD Pharmingen (San Diego, 555665 CA) B7-H2 BD Pharmingen (San Diego, 552502 CA) HLA-G Abcam (Cambridgeshire, UI~)ab 7904-100 CD178 Santa Cruz (San Cruz, CA) sc-19681 PD-L2 BD Pharmingen (San Diego, 557846 CA) Mouse IgG2a Sigma (St. Louis, MO) F-6522 Mouse IgGlkappaSigma (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, [3H]thymidine was added to each sample and cultured for 18 hours.
Following harvest of the cells, radiolabeled DNA was extracted, and [3H]-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 1 O-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) ~______~__ - d~f~o~ife~a'tlon s~ -- .__.~.._~ _..
_! °~ -1' aC"~e lb I F"iate1 ~ ... .. I °°
L.. -...._... ......_ ... 1.......~. .r. ... ............................ ....
~- -. ............. "....~.___.................._...._..
...,_ ..... a.......... ....~.........~_.. __...............r..
...........__.._.. ......... ......... ........
_.................,.............._..._. w.._a..__.............................
I
na ytlca a lure ep Icates ._ ...umber ...._... .............-..._........_.. ..1 .
_..._MeabSD_....._..__._.....CU._.,.....
......... . . _. -System_....-..~ __... ~ , -_._ .... ...-_ -. .....~.....
_ ~ ......
.

FYOliferation bsseline of receiver 79 119 138 112.030.1226.9 Control of autostimulation -~'"2~2~2 17 229 9 2~
(Nitorttycm Ctreated autologous 3 54-' cells) IM03-7769 ..... ..._ ...
. ~~ .._-:. . ..224'14.7 155:97...
N~RaIlogenic~donorlN>D3-7768'(Nitorrycin'Ctreatedj-' 22352_. "..2o~21_.._.
.__ ' ~ 71 ._..6...8........

"" fV~Rw ith cell line (Mtomycin X59 ~0'JO ~~,T~~,6~,.2 X6.5-,T, C treated cell type B) onor ( 200 I (~ ) ...................
,.,....._............................._.._......_...........................__.
................_..........,............................,...._...:.............
..................................................__....-......7......................._.........._........__...........................
.
...................,.. ............."........................ ._. ~
_.
SI (cell line) ~. ._.- _ ._.._ . ..~...

FYOliferation baseline of receiver 6 26 0. . 3 U 3 2 LU ~4 0 /

. . ., .....52. ..... . .....739..6. ._ Nitortrycin'Clreated'autologous _._....091.......... . ...8.0 _-....1.
cells) 602 ...,... ..,3~...e6 Control'of autostirrulatlon" 4 _.. 7. _......
IM03-7770 "' 1 ( -----~Rallogenic donor IMg3-7768 ""~SC~4~T1~ 4-426F~36~ ' (Nitonycin Ctreated) 4~, ......_. .._.. 533. .__._ ......530.31128.24-'~"
MLRw'ith cell line (Nitorrycin 2582 '- 2376.-.... ~fi1.6-.._ Ctreated cell type Bj--n Srce ~ _.
ne ~ "'-""~""-FYOliferation baseline of receiver 128 124.03 l~/ i/ .1 Gi /

Control of autostirrulation -29S X38 8 3'f~ "1881~, (Nitorrycin Ctreated autologous b cells) IM03-7771 _......._...............~_.........-....._.._._........................._.....__....-.-.-_-_.._._....._.______.............__...._ . ...;........ ... _......._ NLR allogenic donor INlo3-7768 . ........ 30077.7 5054.53_ (Nitomycin C treated) .... ....... ........
24497 34348 16.8 . . 6Ø........ 5.22.4.....__.....29.70.
'MLRwilh cell line (Nitomycin~Ctreated 643......_ ..
.......4..8.........
cell~type B)" _".,.a_...._ onor ~ ' _........._..............................._....................................
............_............_......_.....................
_....Sl..tceli Ilnej.......~.................._.........._ ....._ .,.. .... , . ._......................._.....__...........-._ ......m . "
.....................-..... ._..........................
. .._.,................,........................... . <...~.- ~ ......

._....._....... .._..~ , , _......
....._..._~ ._-....._.......
. _. __... . ...__.._._........

FYOliferalion baseline of receiver or s o ii Go 3/
8 i 1 o i s . .

8 .. 20_....... _..155.3...54:36"..._....32..4_......
"Control'ofautostimulation'(NitortrycinCtreatedautologouscells) .213.......
~
IM03-7772 -- -yRallogen''ic'iionor 13 1 188'22 4118:75 "21 INb'3'=7768 (Nitoiiyci~ Cireatedj 14222 20076" 0 . 9 ' 22'168 __.._.._ ____._...
-_. W _. __..___._, .__.
...
_............._.._;.
_.._...
~.
.._ .

____.____..__.._.:..___._._,........~_._.... a a m ~ :............__...._...._,......_..YP_..__.~. -, _ ., . a NILRw tth cell Ine Nato cin _ C treated cell t a B .....
_ a ' a a e S ce hf ne) ----~- ._ ...__..._..-_._....._.....
_....._..._._.-......._.......... ...._ - _...__ ___._._. a..._..._._ _ .
......_.................._..,.................._..................._...........
.............. ..._._. ..... - ._._._ ....._._.._.... _.... . . . - ., - 68 Proliferation baseline 44 of receiver ~
(allogenic donor) ~ontrel of ~'6~6 ~0~ ~ ~ 3 ~ 39.-b autostimulation (Nitomycin 66.y'I
treated autologous cells) Cell line type B Proliferation ' 3 baseline of receiver A., ~ ~ . - ~~
- J

Control of autostimulation 487 799 29.9537.
(Nitomycin treated autologous 822075 cells]

Plate ID: Plate 2 Analyticala lure ttep Ica es number System 1 2 3 Mean SD CV

Proliferation baseline 908 181 330 473.0384.0281.2 of receiver IM03-7773Contrel of autostimulation 269 405 572 415.3151.7636.5 (Mitomycln C treated autologous cells) MLR allogenic donor 29151 28691 28719.0418.701.5 IMO3-7768 (Mitomycin 28315 C treated) MLR with cell line (Mitomycin 567 732 905 734.7169.0223.0 C treated cell type B) ~I (aonor) m SI (cell 2 line) Proliferation baseline 893 1376 185 818.0599.0373.2 of receiver Control of autostlmulation 261 381 568 403.3154.7138.4 (Mitomycln C treated autologous cells) IM03-7774MLR allogenic donor 53101 42839 48074.35134.1810.7 IM03-77ss (Mitomycin 48283 C treated) MLR with cell line (Mitomycin 515 789 294 532.7247.9746.6 C treated cell type B) onor 59 SI (cell 1 line) Proliferation baseline 1272 300 544 705.3505.6971.7 of receiver IM03-7775Control of autosttmulation 232 199 484 305.0155.8951.1 (Mitomycln C treated autologous cells) MLR allogenic donor 23554 10523 21014.09479.7445.1 IMg3-7768 (Mitomycin 28965 C treated) MLR with cell line (Mitomycin 768 924 563 751.7181,0524.1 C treated cell type B) 51 (oonor) SI (cell 1 line) Proliferation baseline IbJU 1JI lU4b UU4.3IUl.LLltS.G
ofrecelver Control of autostimulation 420 218 394 344.0109.8931.9 (Mitomycln C treated autologous cells) IM03-7776MLR allogenic donor 28893 32493 32044.02952.229.2 IMO3-7768 (Mitomycin 34746 C created) MLR with cell line (Mitomycin a a a a a a C treated cell type B) ~I (aonor) J:l SI (cell a line) 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 279 3 1-3) Plate 2 (receivers 46.25 1.3 4-~

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 (plate 1) to 9 (plate 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 na ytlcaa ture ep Icates number System 1 2 3 Mean SD CV

Proliferation baseline 074 406 391 623.7390.0762.5 of receiver Control of autostlmulatlon 672 510 1402 861.3475.1955.2 (Mitomycin C treated autologous cells) IM04-2478MLR allogenic donor 43777 48391 43466.35087.1211.7 IMO4-2477 (Mitomycin 38231 C treated) MLR with cell line (Mitomycin 2914 5622 4881.71721.3635.3 C treated cell type 6109 A) onor SI (cell 8 line) Proliferalionbaselineofreceiver OJU ours ozi oen.imn.aoe.o Control of autostimulation 701 567 1111 793.0283.4335.7 (Mltomyctn C treated autologous cells) IM04-2479MLR allogenic donor 25593 24732 24344.01481.616.1 IM04-2477 (Mitomycin 22707 C treated) MLR with cell line (Mitomycln 5086 3932 3505.01832.2152.3 C treated cell type 1497 A) 'I (oonor) SI (cell line) 7 Proliferation baseline 1192 854 1330 1125.3244.9021.8 of receiver Contrcl of autostimulation 2963 993 2197 2051.0993.0848.4 (Mitomycin C treated autologous cells) IM04-2480MLR allogenic donor 25416 29721 26298.03078.2711.7 IM04-2477 (Mltomycin 23757 C treated) MLR with cell line (Mltomycin 2596 5076 3699.31262.3934.1 C treated cell type 3426 A) 51 (aonor) SI (cell line) 3 Proliferation baseline tiao 4on goo ~or.uwe.44cn.o of receiver Control of autoslimulatlon 738 1252 464 818.0400.0448.9 (Mltomycin C treated autologous cells) IM04-2481MLR allogenic donor 13177 24885 17835.36209.5234.8 IM04-2477 (Mitomycin 15444 C treated) MLR with cell line (Mitomycin 4495 3671 4280.0534.9512.5 C treated cell type 4674 A) 51 (aonor) SI (cell 8 line) -Plate ID: Plate 2 Analyticala ture ep Icates number System 1 2 3 Mean SD CV

Proliferation baseline 432 533 274 413.0130.5431.6 of receiver Control of autostimulation 1459 633 598 896.7487.3154.3 (Mitomycln C treated autotogous cells) IM04-2482MLR allogenic donor IM04-2477 24286 30823 28818.33933.8213.7 (Mitomycin C treated) 31346 MLR with cell line (Mitomycin 2762 1502 6723 3662.32724.4674.4 C treated cell type A) ~t (aonor) rv SI (cell line) 9 -IM04-2477Proliferation baseline 312 419 349 360.054.3415.1 of receiver (allogenicControl of autosiimulatlon 567 604 374 515.0123.5024.0 donor) (Mitomyctn treated autologous cells) ~

Proliferation baseline 101 3 3 9 3 3936.0 2 of receiver . .4 CEII Control of autostimulation 1924 4570 2153 2882.31466.0450.9 Ilne (Mitomycin treated autologous type cells) A

Table 10-5. Average stimulation index of umbilical cells and an allogeneic donor in a mixed lymphocyte reaction with f ve individual allogeneic receivers.
Average Stimulation Index Recipient Umbilicus Plate 1 (receivers 1-4)42.75 6.5 Plate 2 (receiver 5) 70 9 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. W 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, CD80, 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 naive CD4+ T
cells and the presence of PD-L2, an immuno-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. Neplzrol. 10:299-306). To determine the potential for PPDCs to iutiate clotting, umbilical cord and placenta-derived PPDCs were evaluated for tissue factor expression and their ability to initiate plasma clotting.
Metlzods & Materials Human Tissue factor. SIMPLAST1N, 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 O.1M
calcium chloride, and 100 microliters of normal human plasma. The plate was immediately placed in a temperature-controlled microplate reader and absorbance measured at 405 nanometers at 40 second intervals for 30 minutes.
J-~2 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 and umbilical cord were cultured in Growth Medium in gelatin-coated T75 flasks (Corning, Corning, NY).
Placenta-derived cells at passage 5 and mnbilical 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 (3 0 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 1x10 per milliliter. 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-susp ended in 100 microliters of 3% FBS and secondary antibody added as per the manufacturer's instructions.
Cells were incubated in the darlc 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'/2 to max; Table 11-1).
Clotting was observed with both early (P5) and late (P18) cells. The T %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 the 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 T 1/a 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 PS

50,000 757 Umb PS

50,000 833 Umb P 18 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 (Salcariassen et al. (2001) Thronab. Res. 104:149-74; Engstad et al.
(2002) Int.
Immunophaf°macol. 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 axe injected intravenously.

Figure 11-1. Plasma clotting with J82 cells and umbilical cells without (A) and with (B) CI~TO
859, anti-human tissue factor, 20 ~,g/milliliters. Antibody treatment showed that coagulatiori 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 arrhythsnias is highly desirable. Rodent mesenchymal stem cells treated with 5-azacytidine have been shown to express marlcers of cardiomyocytes (Fukuda et al. (2002) C. R. Biol.
325:1027-38). The same has not been shown for adult hmnan 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 et al. (1997) J. Mol. Cell Ca~diol. 29:1525-39), DMSO
(Xu et al.
(2002) Cir~c. Res. 91:501-8); and chelerythrine chloride (International PCT
Publication No.
W003/025149), which effects the translocation of PI~C from the cytosol to plasma membrane and is an inhibitor of PI~C activity. In the present study, PPDCs (P10) were treated with 5-azacytidine either alone or in combination with DMSO or chelerythrine chloride and marlcers of cardiomyocytes measured by real-time PCR.
Methods c~ Matef°ials Cells. Cryopreserved umbilical cord-derived cells (P10) and placenta-derived cells (1'24) 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 ~.M 5-azacytidine (Sigma, St. Louis, MO) alone or with 5 uM chelerythrine chloride (Sigma], 1% (v/v) dimethylsulfoxide (DMSO) (Sigma), or 1 ~,M 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, transfernn, 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 lit, Qiagen, Valencia, CA) and stored at -80°C. Cell 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 TaqMan 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-DemandTM gene expression products cardiac myosin (Hs00165276 ml), sl~eletal myosin (Hs00428 600), GATA 4 (Hs00171403 ml), GAPDH (Applied Biosystems, Foster City, CA), and TaqMan UW
versal 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 muscle (Ambion Austin TX) were used as a control.
Results Analysis of RNA from cardiac muscle showed expression of cardiac myo sin and GATA
4. Slceletal 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 growri for a furtherl4 days expressed GATA 4, but no slceletal myosin or cardiac myosin. Umbilical cord-derived cells (P12) treated for 48h 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 hepa_tocytes.

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 failure (CHF). Coronary artery occlusion, or the blocl~ing 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 inhacardiac 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 Department 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 envirornnental 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.

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 Standaxd 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 distribution such that individual body weights did not exceed X20% of mean weight.
The animals were anesthetized with sodium pentobarbital (40 milligrams/kilogram) and buprenorphine(O.OSmilligrams/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 slcin 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 the interior cavity. The pericardium was carefully opened via gentle teasing with cotton swab s dampened in sterile saline solution. A damp cotton swab was used to gently push the apex of the heart into the opening where a length of 6-0 sills 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 silly 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 suture 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 (Nalgene, Rochester, NYC.
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 final concentration of 10~ viable cells /100 microliters. The cell suspension was loaded into 1 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 rnm 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 slcin to seal the puncture. The surgical drape was removed and fluids Vii. e., lactated Ringers solution, 25 milliliters/kilogram subcutaneously [SC] or intraperitoneally [IP]) were given.
Table 13-2 °Treatment regimens Dosage Dose Time of Gr No Conc. Route / Necropsy of Dose . . Test ArticleLevel (cells/milliliRegimen Treatment Day No Males . (cells/aniinal)ters) Administration 1 8 Vehicle 0 0 Di t rec Placenta injections) #4 (P10) into 2 8 (A) the ischemic region of the left 15 minutes Umbilical ventricle Day 3 8 (P10) of the after coronary28 (B) heart, (+ 1 consistinga~ery ligationDay) 1 million10 millionof 3 to
10 Placenta intramyocardial #3 (P10) 4 8 (C) injections of 100 ~L
total.

Human 8 f broblasts 1F1853 (P10) (D) Gr. = Group; iV o. = Number; ~:onc. _ (.;oncentrat~on hnmediately 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 2~ 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 c age.
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/lcilogram 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., anmal remains in hunched position), antipathy, lack of eating/drinking, lack of grooming, etc.
Body weight was recorded for each animal prior to initial treatment, weelely 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 KCl infused via the jugular cannula. The heart was then removed from the thoracic cavity. A
limited necropsy was -9~-then perfomned on the heart after which the heart was placed in 10% neutral buffered fonalin.
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 microtome 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 developed 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 Dept on ice. Cells were injected into the infarct with one to three needle entry points and 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 wee similarly utilized. These values were calculated as described by Salm et al. (1978) Circulation 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 axlalysis. The hearts were arrested in diastole and fixed. The results were calculated from am algorithm to estimate the percentage of total heart area that comprises the infarct. The infarct size in the vehicle-treated animals was 22.9% ~ 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% ~

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 functiori 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 aazimals 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.
Metlz~ds & Materials Cell Culture. Umbilical cord-derived cells were grown in Growth Mediurr~ in gelatin-coated flaslcs.
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 irig/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 grow~.i 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 10~ cells in 10% (w/v) sucrose (Sigma, St Louis, MO), 10 millimolar HEPES in Dulbecco's modified medium (DMEM; Gibco) immediately before use. The final concentration of cells in RAD16 hydrogel was 1 x 10~ cells/100 microliters.
TEST MATERIAL (N=4/Rx) 1. Vicryl non-woven + 1 x 10~ umbilical cord-derived cells 2. 35/65 PCL/PGA foam + 1 x 10~ umbilical cord-derived cells 3. RAD 16 self assembling peptide + 1 x 10~ mnbilical 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)lFox Chase SCIDlMale (Ha~lah Sprague Dawley, Ins., Ihdiart.apolis, IhdiaiZa), S weelzs of age. All handling of the SCID mice tools place under a hood.
The mice were individually weighed and anesthetized with an intraperitoneal inj ection of a mixture of 60 milligrams/kilogram KETASET (ketamine hydrochloride, Aveco Co., Ins., 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 craual 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 poclcet and the implant placed (or injected for RAD16) about 1-cm caudal to the incision.

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.
EXAl~~IPLE 15 Endothelial Network Formation: PPDCs Promote Angiogenesis hz Vits~o 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 Trit~o 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 formation (Ito et czl. (1996) Int. J. Cancers 67(1):148-52). The present studies made use of a co-culture system with the postpartum cells seeded onto cultuxe well inserts. The permeable inserts allow for the passive exchange of media components between the endothelial and the postpartum culture media.
Mate~~ial & Methods Cell Culture.
Postpas°tum tissue-derived cells. Hmnan 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~
Transwell0 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 37 °C with 5%
C02.
Human fnesenclayn2al stern cells (hMSC). 1~1VISCs were purchased from Cambrex (Wall~ersville, 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 CostarOO
Transwell~
6.5 mm diameter tissue culture inserts (Corning, Corning, N~ at 15,000 cells per insert. Cells were cultured on the inserts for 48-72 hours in Growth Medium at 37°C
with 5% C02.
Human mnbilical vein endothelial cells (HUVEC). HLJVEC were obtained from Cambrex (Walkersville, MD). Cells were grown in separate cultures in either EBM or EGM

endothelial cell media (Cambrex). Cells were grown on standard tissue cultured plastic at 37°C
with 5% COZ. Cells used in the assay were from about passage 4 to 10.
Human coronary of°teYy eyadothelial cells (HCAEC). HCAEC were purchased from Cambrex Incorporated (Wall~ersville, MD). The cells were maintained in separate cultures in either the EBM or EGM media formulations. Cells were grown on staxidard tissue culture plastic at 37°C with 5% C02. Cells used for experiments were from about passage 4 to 8.
MatrigelTM assays. Culture plates were coated with MatrigelTM 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 (Corning). 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% FBS
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) FBS. 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%C02 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, HLTVEC form cell networks. HUVEC cells form limited cell networl~s in co-culture experiments with hMSC and with 10 nanomolar bFGF. HLTVEC cells without any treatment showed very little or no networlc formation. These results suggest that the postpaitmn 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 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 (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 15-1. 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-BB), 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.

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 picograzns per milliliter of culture media.
TIMP1 ANG2 PDGFBB TP~D KGF HGF FGF VEGF HBEGF
(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 <2.0 <3.2 <5.4 <4.0 <1.2 Summary. Postpartum cells can stimulate both human umbilical vein and coronary artery endothelial cells to form networlcs in an in vitro MatrigelTM assay.
This effect is similar to that with l~nown angiogenic factors in this assay system, suggesting that the postpartum cells are useful for stimulating angiogenesis ira 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 UMB 022803 (P 17) 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 (P 8) 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.

CBAT0016.ST25.txt 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 CARDIAC DISEASE
<130> CBAT-0016 <150> US 60/483,264 <151> 2003-06-27 <160> 10 <170> PatentIn version 3.2 <210> 1 <211> 22 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 1 gagaaatcca aagagcaaat gg 22 <210> 2 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 2 agaatggaaa actggaatag g 21 <210> 3 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 3 tcttcgatgc ttcggattcc 20 <210> 4 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 4 gaattctcgg aatctctgtt g 21 CBAT0016.ST25.txt <210> 5 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 5 ttacaagcag tgcagaaaac c 21 <210> 6 <211> 22 <212> DNA
<213> Artificial Sequence <220> ' <223> Synthetic Construct <400> 6 agtaaacatt gaaaccacag cc 22 <210> 7 <211> 20 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 7 tctgcagctc tgtgtgaagg 20 <210> 8 <211> 22 <212> DNA
<213> Artificial Sequence <220>
<223>- Synthetic Construct <400> 8 cttcaaaaac ttctccacaa cc 22 <210> 9 <211> 17 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 9 cccacgccac gctctcc <210> 10 <211> 19 <2l2> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct <400> 10 CBAT0016.ST25.txt tcctgtcagt tggtgctcc 19

Claims (61)

What is Claimed:
1. An isolated postpartum-derived cell comprising an L-valine-requiring cell derived from.
human postpartum tissue substantially free of blood, said cell capable of self renewal and expansion in culture and having the potential to differentiate into a cell of cardiomyocyte phenotypes; the cell capable of growth in an atmosphere containing oxygen from about 5% to at least about 20%; wherein said cell comprises at least one of the following characteristics:
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, polyornithine, vitronectin, or fibronectin;
production of at least one of tissue factor, vimentin, 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 HLA-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 (melanoma growth stimulating activity, alpha); chemolcine (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 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);
expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac 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 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 cysteW a 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 l; tenascin C (hexabrachion); Iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; Homo sapiens cDNA FLJl 2280 fis, clone MAMMA1001744; cytolcine receptor-like factor 1; potassium intemediate/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; KIA.A0367 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 l9kDa interacting protein 3-like; AE binding protein 1;
cytochrome c oxidase subunit VIIa 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, MIP 1 a, RANTES, and TIMP 1; and lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIPlb, I309, MDC, and ~EGF, as detected by ELISA.
2. The postpartum-derived cell of claim 1 isolated in the presence of one or more enzyme activities comprising metalloprotease activity, mucolytic activity and neutral protease activity.
3. The postpartum-derived cell of claim 2 wherein the enzyme activities are collagenase and dispase.
4. The postpartum-derived cell of claim 3 further comprising hyaluronidase.
5. The postpartum-derived cell of claim 4 comprising a normal karyotype.
6. The postpartum-derived cell of claim 5 that maintains its karyotype as it is passaged.
7. The postpartum-derived cell of claim 6 wherein the cell expresses each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C.
8. The postpartum-derived cell of claim 7 wherein the cell does not express any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
9. The postpartum-derived cell of claim 8 wherein the cell does not spontaneously differentiate along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway when cultured in Growth Medium.
10. A population of cells comprising the postpartum-derived cell of claim 8.
11. The population of claim 10 comprising from about 1% postpartum-derived cells to about 10% postpartum cells.
12. The population of claim 11 comprising about 50% postpartum-derived cells.
13. The population of claim 12 comprising at least 90% postpartum-derived cells.
14. The population of claim 13 comprising substantially only postpartum-derived cells.
15. The population of claim 14 comprising a clonal cell line of postpartum-derived cells.
16. A cell lysate prepared from the population of claim 10.
17. The population of claim 10 incubated in the presence of one or more factors which stimulate stem cell differentiation along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
18. The population of claim 17 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.
19. The population of claim 18 wherein the demethylation agent comprises an inhibitor of DNA
methyltransferase or an inhibitors of a histone deacetylase, or inhibitors of a repressor complex _
20. The population of claim 18 wherein the demethylation agents 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.
21. The population of claim 10 that is seed onto a matrix to form a matrix-cell complex.
22. The population of claim 21 wherein the matrix is a scaffold.
23. The population of claim 22 wherein the scaffold is bioabsorbable.
24. The population of claim 23 wherein the scaffold comprises at least one other cell type.
25. The population of claim 24 wherein the other cell type is a stem cell.
26. A therapeutic cell composition comprising a pharmaceutically-acceptable carrier and post-partum-derived cells derived from human postpartum tissue substantially free of blood, said cells capable of self renewal and expansion in culture and having the potential to differentiate into cells of cardiomyocyte phenotypes; the cells capable of growth in an atmosphere containing oxygen from about 5% to at least about 20%; wherein said cells:
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 comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin, 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.
27. A method of treating a patient with a disease of the heart or circulatory system comprising administering the therapeutic cell composition of claim 26.
28. The method of claim 27 wherein the patient has a cardiomyopathy.
29. The method of claim 28 wherein the cardiomyopathy is idiopathic.
30. The method of claim 29 wherein the cardiomyopathy is ischemic cardiomyopathy or nonischemic cardiomyopathy.
31. The method of claim 30 wherein the cardiomyopathy is a nonischemic cardiomyopathy selected from dilated cardiomyopathy, hypertrophic cardiomyopathy, and restrictive cardiomyopathy.
32. The method of claim 27 wherein the cells are induced along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
33. The method of claim 32 comprising cells induced in vitro.
34. The method of claim 33 comprising cells induced in vitro.
35. The method of claim 27 comprising cells that stimulate adult stem cells present in the heart to divide, or differentiate, or both.
36. The method of claim 27 wherein the administering is by injection.
37. The method of claim 36 wherein the injection is intracardiac injection.
3 8. The method of claim 27 wherein the cells are provided as a matrix-cell complex.
39. The method of claim 38 wherein the matrix is a scaffold.
4Ø The method of claim 39 wherein the scaffold is bioabsorbable.
41 The therapeutic cell composition of claim 26 further comprising the following characteristics:
expression of 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;
expression of 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 kinase C, zeta;
hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; Homo sapiens mRNA; and cDNA
DKfZp547K1113 (from clone DKFZp547K1113);
expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of: short stature homeobox 2; heat shock 27 kDa 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;
disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein;
similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3;
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;

neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein l; early growth response 3; distal-less homeobox 5; .cndot.hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan;
transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin;
integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE
1968422; EphA3; KTAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C
(atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA;
cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19kDa interacting protein 3-like; AE binding protein 1; and cytochrome c oxidase subunit VIIa polypeptide 1 (muscle);
secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, I309, MDC, and VEGF, as detected by ELISA.
42. The therapeutic cell composition of claim 41 comprising about 50%
postpartum-derived cells.
43. The therapeutic cell composition of claim 42 comprising substantially only postpartum-derived cells.
44. The therapeutic cell composition of claim 43 comprising a substantially homogeneous population of postpartum-derived cells.
45. The therapeutic cell composition of claim 44 comprising a clonal cell line of postpartum-derived cells.
46. The therapeutic cell composition of claim 44 wherein the substantially homogeneous population is umbilical-derived cells.
47. The therapeutic cell composition of claim 44 wherein the substantially homogeneous population is substantially free of maternal cells.
48. The therapeutic cell composition of claim 44 wherein the substantially homogeneous population is placental-derived cells.
49. The therapeutic cell composition of claim 44 wherein the substantially homogeneous population is of neonatal origin.
50. The therapeutic cell composition of claim 44 wherein the substantially homogeneous population is of maternal origin.
51. A method for treating a patient with a cardiomyopathy comprising administering a therapeutic postpartum-derived cell composition to a patient with a cardiomyopathy; acid evaluating the patient for improvements in cardiac function.
52. The method of claim 51 wherein the cells are induced along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
53. The method of claim 51 wherein the cells stimulate adult stem cells present in the heart to divide or differentiate, or both.
54. The method of claim 51 wherein improvements include improvements in 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.
55. The method of claim 51 wherein the administering is in vivo by transplanting, implanting, injecting, fusing, delivering via catheter, or providing as a matrix-cell complex.
56. The method of claim 55 wherein the administering is by intracardiac injection.
57. The method of claim 51 wherein the administering is to a patient who is syngeneic with the postpartum-derived cells.
58. The method of claim 51 wherein the administering is to a patient who is allogeneic
59. A coculture of human postpartum cells and another mammalian cell, wherein at least one of the cells is a cardiomyoblast, angioblast, or hemangioblast or induced to differentiate along a pathway leading to a cardiomyoblast, angioblast or hemangioblast.
60. A method of treating a patient with a disease of the heart or circulatory system comprising administering to the patient a therapeutic composition comprising one or more of postpartum-derived cells, a conditioned medium generated by postpartum-derived cells, a cell lysate derived from postpartum-derived cells, a soluble cell fraction from postpartum-derived cells, an extracellular matrix from postpartum-derived cells.
61. The method of claim 60 further comprising coadministering one or more of an antithrombogenic agent, an anti-inflammatory, an immunosuppressive agent, an immunomodulatory agent, and an antiapoptotic agent.
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 true CA2530422A1 (en) 2005-01-06
CA2530422C CA2530422C (en) 2016-01-05

Family

ID=38456561

Family Applications (7)

Application Number Title Priority Date Filing Date
CA2530255A Expired - Fee Related CA2530255C (en) 2003-06-27 2004-06-25 Soft tissue repair and regeneration using postpartum-derived cells
CA2530421A Expired - Fee Related CA2530421C (en) 2003-06-27 2004-06-25 Repair and regeneration of ocular tissue 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
CA2530412A Expired - Fee Related CA2530412C (en) 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells
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

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CA2530255A Expired - Fee Related CA2530255C (en) 2003-06-27 2004-06-25 Soft tissue repair and regeneration using postpartum-derived cells
CA2530421A Expired - Fee Related CA2530421C (en) 2003-06-27 2004-06-25 Repair and regeneration of ocular tissue using postpartum-derived cells

Family Applications After (4)

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

Country Status (8)

Country Link
US (28) US7413734B2 (en)
EP (14) EP1641913B1 (en)
JP (7) JP5148873B2 (en)
AU (7) AU2004252570C1 (en)
CA (7) CA2530255C (en)
ES (14) ES2600555T3 (en)
PL (14) PL2338980T3 (en)
WO (7) WO2005001076A2 (en)

Families Citing this family (536)

* 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
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
KR101132545B1 (en) * 2001-02-14 2012-04-02 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
EP1362095B1 (en) * 2001-02-14 2015-05-27 Anthrogenesis Corporation 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
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20080299090A1 (en) * 2002-02-25 2008-12-04 Kansas State University Research Foundation Use Of Umbilical Cord Matrix Cells
US20080003258A1 (en) * 2002-10-16 2008-01-03 Marcum Frank D Composition and Method for Treating Rheumatoid Arthritis
US7824701B2 (en) 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
US20040078090A1 (en) 2002-10-18 2004-04-22 Francois Binette Biocompatible scaffolds with tissue fragments
WO2004047770A2 (en) 2002-11-26 2004-06-10 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US7807458B2 (en) 2003-01-30 2010-10-05 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Multilineage-inducible cells and uses thereof
CA2515469C (en) 2003-02-11 2013-12-17 John E. Davies Progenitor cells from wharton's jelly of human umbilical cord
KR20050105467A (en) * 2003-02-13 2005-11-04 안트로제네시스 코포레이션 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
CN100377165C (en) * 2003-04-24 2008-03-26 皇家飞利浦电子股份有限公司 Non-invasive left ventricular volume determination
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
US8790637B2 (en) * 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US8491883B2 (en) * 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
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
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
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
AU2004262451B2 (en) * 2003-08-12 2009-11-26 Tigenix N.V. Use of CXCL6 chemokine 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
WO2005055929A2 (en) * 2003-12-02 2005-06-23 Celgene Corporation 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
WO2005071066A1 (en) * 2004-01-23 2005-08-04 Board Of Regents, The University Of Texas System 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
EP2824174B1 (en) 2004-03-22 2018-11-28 Mesoblast International Sàrl Mesenchymal stem cells and uses therefor
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8221780B2 (en) * 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
CN101080486B (en) * 2004-04-23 2012-05-16 佰欧益股份有限公司 Multi-lineage progenitor cells
US7622108B2 (en) 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
US8765119B2 (en) * 2004-05-06 2014-07-01 University Of South Florida Treating amyotrophic lateral sclerosis (ALS)with isolated aldehyde dehydrogenase-positive umbilical cord blood cells
US8802651B2 (en) * 2004-06-30 2014-08-12 Abbott Medical Optics Inc. Hyaluronic acid in the enhancement of lens regeneration
US7794697B2 (en) 2004-06-30 2010-09-14 Abbott Medical Optics Inc. Enhancement of lens regeneration using materials comprising polysiloxane polymers
US20060083732A1 (en) * 2004-06-30 2006-04-20 Arlene Gwon Hyaluronic acid in the enhancement of lens regeneration
MX2007001937A (en) 2004-08-16 2007-08-07 Cellres Corp Pte Ltd Isolation of stem/progenitor cells from amniotic membrane of 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
WO2006101548A2 (en) * 2004-12-21 2006-09-28 Ethicon, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
AU2005322068B2 (en) 2004-12-23 2011-09-01 Ethicon Incorporated Treatment of stroke and other acute neural degenerative disorders using postpartum derived cells
PL1831356T3 (en) * 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. 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
WO2006088867A2 (en) * 2005-02-15 2006-08-24 Medistem Laboratories, Incorporated Method for expansion of stem cells
JP4783909B2 (en) * 2005-03-04 2011-09-28 国立大学法人京都大学 Pluripotent stem cells derived from heart tissue
US20060222634A1 (en) 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
JP5017253B2 (en) * 2005-03-31 2012-09-05 ステムニオン,インコーポレイテッド Amnion-derived cell composition, preparation method and use thereof
US8153430B2 (en) * 2005-03-31 2012-04-10 Stemnion, Inc. Methods related to surgery
US8999706B2 (en) * 2005-04-12 2015-04-07 The Trustees Of The University Of Pennsylvania Methods for preparation of human hair-follicle derived multipotent adult stem cells
WO2006113731A2 (en) * 2005-04-20 2006-10-26 University Of Florida Research Foundation, Inc. Bone marrow-derived neurogenic cells and uses thereof
US20100087546A1 (en) * 2005-04-20 2010-04-08 Biogenic Innovations, Llc Use of dimethyl sulfone (msm) to reduce homocysteine levels
KR20080007380A (en) * 2005-04-25 2008-01-18 메사추세츠 인스티튜트 오브 테크놀로지 Compositions and methods for promoting hemostasis and other physiological activities
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
US20140336514A1 (en) * 2005-08-05 2014-11-13 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
US20150119794A1 (en) * 2005-08-05 2015-04-30 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
AU2006291134C1 (en) 2005-09-12 2013-08-15 Abela Pharmaceuticals, Inc. Systems for removing dimethyl sulfoxide (DMSO) or related compounds, or odors associated with same
US8435224B2 (en) 2005-09-12 2013-05-07 Abela Pharmaceuticals, Inc. Materials for facilitating administration of dimethyl sulfoxide (DMSO) and related compounds
US9427419B2 (en) 2005-09-12 2016-08-30 Abela Pharmaceuticals, Inc. Compositions comprising dimethyl sulfoxide (DMSO)
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
JP2007106760A (en) * 2005-09-16 2007-04-26 Kenji Yoshida Hematopoietic stem cell proliferator
US8182840B2 (en) 2005-09-27 2012-05-22 Tissue Tech, Inc. Amniotic membrane preparations and purified compositions and therapy for scar reversal and inhibition
EP2530145A1 (en) 2005-10-13 2012-12-05 Anthrogenesis Corporation Immunomodulation using placental stem cells
NZ567334A (en) * 2005-10-13 2012-08-31 Anthrogenesis Corp 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
US10117900B2 (en) * 2005-11-09 2018-11-06 Athersys, Inc. MAPC treatment of brain injuries and diseases
US20070178067A1 (en) * 2005-11-09 2007-08-02 John Maier System and method for cytological analysis by raman spectroscopic imaging
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
NZ571113A (en) * 2005-11-17 2012-02-24 Biomimetic Therapeutics Inc Maxillofacial bone augmentation using rhpdgf-bb and a biocompatible matrix
WO2007070870A1 (en) * 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
WO2007073552A1 (en) * 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
CN103173401A (en) 2005-12-22 2013-06-26 简·恩尼斯 Viable cells from frozen umbilical cord tissue
WO2007076522A2 (en) * 2005-12-28 2007-07-05 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
EP1976978A2 (en) 2005-12-29 2008-10-08 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US9598669B2 (en) * 2005-12-29 2017-03-21 Anthrogenesis Corporation Composition for collecting placental stem cells and methods of using the composition
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
EP1981970A4 (en) 2006-01-11 2009-10-21 Technion Res & Dev Foundation 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
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
US8871198B2 (en) * 2006-03-29 2014-10-28 Stemnion, Inc. Methods related to wound healing
US7875451B2 (en) * 2006-01-19 2011-01-25 The University Of Washington Formulation to improve survival of transplanted cells
NZ744465A (en) 2006-01-23 2023-04-28 Abt Holding Co Mapc therapeutics without adjunctive 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
EP1996698A1 (en) * 2006-03-01 2008-12-03 Cartela R&D AB Expansion and differentiation of mesenchymal stem cells
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
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
PT2366775E (en) * 2006-03-23 2015-07-20 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US7727763B2 (en) * 2006-04-17 2010-06-01 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
US8741643B2 (en) * 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
PL2029149T3 (en) 2006-05-05 2017-12-29 John E. Davies Immune privileged and modulatory progenitor cells
EP2023717B1 (en) 2006-05-11 2016-04-13 Anthrogenesis Corporation Methods for collecting 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
WO2007146106A2 (en) * 2006-06-05 2007-12-21 Cryo- Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
US20080044848A1 (en) * 2006-06-09 2008-02-21 Heidaran Mohammad A 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
BRPI0713965A2 (en) * 2006-06-28 2012-11-27 Univ Kansas differentiation of umbilical cord matrix trunk in hepatocyte lineage cells
US9642891B2 (en) * 2006-06-30 2017-05-09 Biomimetic Therapeutics, Llc 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
EP2049043B1 (en) 2006-07-24 2018-10-10 International Stem Cell Corporation Synthetic cornea from retinal stem cells
US7993918B2 (en) * 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US8822030B2 (en) 2006-08-11 2014-09-02 Aqua Resources Corporation Nanoplatelet metal hydroxides and methods of preparing same
WO2008021256A2 (en) * 2006-08-11 2008-02-21 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
PT2078073E (en) 2006-10-12 2013-11-11 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
NZ606814A (en) 2006-10-23 2014-10-31 Anthrogenesis Corp Methods and compositions for treatment of bone defects with placental cell populations
EP3181157B1 (en) * 2006-11-03 2019-08-14 BioMimetic Therapeutics, LLC Compositions and methods for arthrodetic procedures
JP5670053B2 (en) * 2006-11-13 2015-02-18 エシコン・インコーポレイテッドEthicon, Incorporated 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
US20080152630A1 (en) * 2006-12-07 2008-06-26 Irene Ginis 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
ES2623141T3 (en) 2007-01-17 2017-07-10 Noveome Biotherapeutics, Inc. New methods to modulate inflammatory and / or immune responses
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
NZ597779A (en) * 2007-02-12 2013-07-26 Anthrogenesis Corp 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
EP2125000A2 (en) * 2007-02-20 2009-12-02 Biomimetic Therapeutics, Inc. Prevention and treatment for osteonecrosis and osteoradionecrosis of the jaw using pdgf and a bone matrix
TW200902718A (en) * 2007-03-01 2009-01-16 Cryo Cell Int Procurement, 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
CA3071055A1 (en) * 2007-04-07 2008-10-16 Whitehead Institute For Biomedical Research Reprogramming of somatic cells
EP2139497B1 (en) * 2007-04-13 2013-11-06 Stemnion, INC. Methods for treating nervous system injury and disease
WO2008132722A1 (en) 2007-04-26 2008-11-06 Ramot At Tel-Aviv University Ltd. Pluripotent autologous stem cells from oral mucosa and methods of use
US8574567B2 (en) 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
EP2155860B1 (en) * 2007-05-03 2014-08-27 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
US10398804B2 (en) 2007-05-21 2019-09-03 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
EP2201910B1 (en) * 2007-06-01 2013-04-03 Allergan, Inc. Biological tissue growth support trough induced tensile stress
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
WO2008156659A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US20090004271A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Morselized foam for wound treatment
US20090004253A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Composite device for the repair or regeneration of tissue
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
US20090024224A1 (en) 2007-07-16 2009-01-22 Chen Silvia S Implantation of cartilage
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
EP2182971B1 (en) 2007-07-27 2013-12-18 Humacyte, Inc. Compositions comprising human collagen and human elastin and uses thereof
EP2185693B1 (en) 2007-07-31 2019-07-03 Lifescan, Inc. Differentiation of human embryonic stem cells
EP2175898A2 (en) * 2007-08-08 2010-04-21 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
CA2736663C (en) 2007-09-07 2018-01-02 Surgical Biologics, Llc. Placental tissue grafts and improved methods of preparing and using the same
PL2200622T5 (en) 2007-09-19 2016-08-31 Pluristem Ltd Adherent cells from adipose or placenta tissues and use thereof 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
CN101978045A (en) * 2007-09-26 2011-02-16 细胞基因细胞疗法公司 Angiogenic cells from human placental perfusate
EP3524253A1 (en) 2007-09-28 2019-08-14 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
UA99152C2 (en) * 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
WO2009052209A2 (en) * 2007-10-16 2009-04-23 University Of Kansas Isolation of stem cells and effective control of contamination
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
WO2009052459A1 (en) * 2007-10-19 2009-04-23 University Of Medicine And Dentistry Of New Jersey A method of using an extracellular matrix to enhance cell transplant survival and differentiation
KR20210056449A (en) * 2007-11-07 2021-05-18 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
US20110086080A1 (en) * 2007-11-14 2011-04-14 Osteosphere, Llc Ex-vivo production of human demineralized bone matrix
MX2010005805A (en) 2007-11-27 2010-06-09 Lifescan Inc Differentiation of human embryonic stem cells.
US20090163990A1 (en) * 2007-12-19 2009-06-25 Chunlin Yang Decellularized omentum matrix and uses thereof
CN101945994A (en) * 2007-12-19 2011-01-12 再生科学有限责任公司 Compositions and methods to promote implantation and engrafment of stem cells
US20120156134A1 (en) 2007-12-20 2012-06-21 Shayne Squires Compositions and methods for detecting or eliminating 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
WO2009085860A1 (en) * 2007-12-27 2009-07-09 Ethicon, Incorporated 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
US7943573B2 (en) 2008-02-07 2011-05-17 Biomimetic Therapeutics, Inc. Methods for treatment of distraction osteogenesis using PDGF
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
CN105886459A (en) 2008-02-21 2016-08-24 詹森生物科技公司 Methods, surface modified plates and compositions for cell attachment, cultivation 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
EP2257176B1 (en) * 2008-03-14 2013-09-18 Regenerative Sciences, LLC Compositions and methods for cartilage repair
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
CA2721870C (en) 2008-04-21 2020-12-22 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
US9358320B2 (en) 2008-04-25 2016-06-07 Allosource Multi-layer tissue patches
US9480549B2 (en) 2008-04-25 2016-11-01 Allosource Multi-layer tissue patches
EP2268326B1 (en) 2008-04-30 2016-11-23 Ethicon, Inc 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
WO2010011407A2 (en) * 2008-05-23 2010-01-28 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
US9682173B2 (en) * 2008-06-11 2017-06-20 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
JP2011524174A (en) 2008-06-13 2011-09-01 ホワイトヘッド・インスティテュート・フォー・バイオメディカル・リサーチ Cell programming and reprogramming
WO2009158480A2 (en) * 2008-06-26 2009-12-30 Kci Licensing, Inc. Stimulation of cartilage formation using reduced pressure treatment
WO2009157559A1 (en) * 2008-06-27 2009-12-30 独立行政法人産業技術総合研究所 Pancreatic cell regeneration/transplantation kit for pancreatic diseases or diabetes
CA2729734A1 (en) * 2008-06-30 2010-01-07 Centocor Ortho Biotech Inc. Differentiation of pluripotent stem cells
ES2697798T3 (en) 2008-06-30 2019-01-28 Janssen Biotech Inc Differentiation of pluripotent stem cells
US20100028307A1 (en) * 2008-07-31 2010-02-04 O'neil John J Pluripotent stem cell differentiation
EP3539380A3 (en) 2008-08-20 2019-12-18 Celularity, Inc. Improved cell composition and methods of making the same
KR20180108887A (en) * 2008-08-20 2018-10-04 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
CA2734446C (en) 2008-08-22 2017-06-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
SG190659A1 (en) 2008-09-02 2013-06-28 Pluristem Ltd Adherent cells from placenta tissue and use thereof in therapy
BR122020000059B8 (en) 2008-09-09 2021-06-22 Biomimetic Therapeutics Inc composition comprising a biocompatible matrix and a platelet-derived growth factor and kit
US9446227B2 (en) 2008-09-12 2016-09-20 Sonescence, Inc. Ultrasonic dispersion of compositions in tissue
US20100069827A1 (en) * 2008-09-12 2010-03-18 Barry Neil Silberg Pre-Surgical Prophylactic Administration of Antibiotics and Therapeutic Agents
CN102170919A (en) 2008-10-06 2011-08-31 三维肽胶株式会社 Tissue plug
WO2010048418A1 (en) * 2008-10-22 2010-04-29 The Trustees Of Columbia University In The City Of New York Cartilage regeneration without cell transplantation
CA2742268C (en) 2008-10-31 2020-02-18 Centocor Ortho Biotech Inc. Differentiation of human embryonic stem cells to the pancreatic endocrine lineage
EP2350265B1 (en) * 2008-10-31 2019-04-17 Janssen Biotech, Inc. Differentiation of human embryonic stem cells to the pancreatic endocrine lineage
JP5701765B2 (en) * 2008-10-31 2015-04-15 シンセス ゲーエムベーハー Methods and devices for activating stem cells
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.
RU2562154C2 (en) * 2008-11-19 2015-09-10 Антродженезис Корпорейшн Amniotic adhesive cells
EP2346452A2 (en) * 2008-11-20 2011-07-27 Lifecell Corporation Method for treatment and prevention of parastomal hernias
ES2584053T3 (en) * 2008-11-20 2016-09-23 Janssen Biotech, Inc. Methods and compositions for cell binding and culture in flat substrates
JP2012509085A (en) 2008-11-20 2012-04-19 ヤンセン バイオテツク,インコーポレーテツド Culture of pluripotent stem cells on microcarriers
WO2010060031A1 (en) * 2008-11-21 2010-05-27 Anthrogenesis Corporation Treatment of diseases, disorders or conditions of the lung using placental cells
WO2010064702A1 (en) * 2008-12-05 2010-06-10 国立大学法人 東京大学 Biomarker for predicting prognosis of cancer
US20110245804A1 (en) 2008-12-05 2011-10-06 Regenerative Sciences, Llc Methods and Compositions to Facilitate Repair of Avascular Tissue
US8366748B2 (en) 2008-12-05 2013-02-05 Kleiner Jeffrey Apparatus and method of spinal implant and fusion
US20100168022A1 (en) * 2008-12-11 2010-07-01 Centeno Christopher J Use of In-Vitro Culture to Design or Test Personalized Treatment Regimens
US10179900B2 (en) * 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
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
ES2520393T3 (en) * 2008-12-19 2014-11-11 DePuy Synthes Products, LLC Cells derived from umbilical cord tissue to treat neuropathic pain and spasticity
CA2747794C (en) * 2008-12-19 2018-10-30 Advanced Technologies And Regenerative Medicine, Llc Treatment of lung and pulmonary diseases and disorders
JP6095893B2 (en) * 2008-12-19 2017-03-15 デピュイ・シンセス・プロダクツ・インコーポレイテッド Regeneration and repair of nerve tissue after injury
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
KR20110107357A (en) * 2008-12-30 2011-09-30 케이씨아이 라이센싱 인코포레이티드 Reduced pressure augmentation of microfracture procedures for cartilage repair
WO2010084943A1 (en) * 2009-01-23 2010-07-29 国立大学法人東京大学 Method for evaluating cultured cells
US9247943B1 (en) 2009-02-06 2016-02-02 Kleiner Intellectual Property, Llc Devices and methods for preparing an intervertebral workspace
WO2010093468A1 (en) * 2009-02-12 2010-08-19 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
US20120114618A1 (en) * 2009-03-26 2012-05-10 The Regents Of The University Of California Mesenchymal Stem Cells Producing Inhibitory RNA for Disease Modification
BRPI1013409A2 (en) * 2009-03-26 2018-01-16 Advanced Tech And Regenerative Medicine Llc human umbilical cord tissue cells as therapy for alzheimer's disease
US20120039857A1 (en) * 2009-04-06 2012-02-16 Capricor, Inc. Systems and methods for cardiac tissue repair
MX337823B (en) * 2009-05-13 2016-03-22 Medipost Co Ltd Tsp-1, tsp-2, il-17br and hb-egf associated with stem cell activities and applications thereof.
WO2010135527A2 (en) * 2009-05-20 2010-11-25 Humacyte, Inc. Elastin for soft tissue augmentation
EP2434896A4 (en) * 2009-05-28 2014-01-22 Univ Central Florida Res Found 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
RU2540016C2 (en) * 2009-07-20 2015-01-27 Янссен Байотек, Инк. Differentiating human embryonic stem cells
CA2768644A1 (en) 2009-07-20 2011-01-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
MX348077B (en) 2009-07-20 2017-05-25 Janssen Biotech Inc Differentiation of human embryonic stem cells.
ES2360434B1 (en) * 2009-07-21 2012-04-12 Universitat Internacional De Catalunya PLURIPOTENTIAL MOTHER CELLS OBTAINED FROM THE DENTAL PULP.
DK2456853T3 (en) * 2009-07-21 2021-02-01 Abt Holding Co USE OF STEM CELLS TO REDUCE LEUCOCYT EXTRAVASATION
WO2011022071A2 (en) * 2009-08-20 2011-02-24 The Regents Of The University Of California Cardiac compositions
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
US9060877B2 (en) 2009-09-18 2015-06-23 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US9629729B2 (en) 2009-09-18 2017-04-25 Spinal Surgical Strategies, Llc Biological delivery system with adaptable fusion cage interface
US10973656B2 (en) 2009-09-18 2021-04-13 Spinal Surgical Strategies, Inc. Bone graft delivery system and method for using same
US8906028B2 (en) 2009-09-18 2014-12-09 Spinal Surgical Strategies, Llc Bone graft delivery device and method of using the same
USD723682S1 (en) 2013-05-03 2015-03-03 Spinal Surgical Strategies, Llc Bone graft delivery tool
USD750249S1 (en) 2014-10-20 2016-02-23 Spinal Surgical Strategies, Llc Expandable fusion cage
US9173694B2 (en) 2009-09-18 2015-11-03 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
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
US8685031B2 (en) 2009-09-18 2014-04-01 Spinal Surgical Strategies, Llc Bone graft delivery system
US9186193B2 (en) 2009-09-18 2015-11-17 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
EP2483392B1 (en) * 2009-09-23 2015-11-11 Davinci Biosciences LLC Umbilical cord lining stem cells and methods and material for isolating and culturing same
CA2777663A1 (en) 2009-10-16 2011-04-21 University Of Medicine And Dentistry Of New Jersey Method for treating chronic nerve tissue injury using a cell therapy strategy
MX350966B (en) * 2009-10-29 2017-09-26 Janssen Biotech Inc Pluripotent stem cells.
US9855212B2 (en) 2009-10-30 2018-01-02 Abela Pharmaceuticals, Inc. Dimethyl sulfoxide (DMSO) or DMSO and methylsulfonylmethane (MSM) formulations to treat infectious diseases
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
EP2498798A4 (en) * 2009-11-10 2014-01-01 Univ Columbia Compositions and methods for wound treatment
WO2011062584A1 (en) * 2009-11-19 2011-05-26 Regents Of The University Of Minnesota Reducing inflammation using cell therapy
KR101867369B1 (en) 2009-12-23 2018-06-14 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
AU2010333840B2 (en) * 2009-12-23 2016-01-07 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
CA2787894A1 (en) 2010-01-26 2011-08-04 Stem Cells Spin S.A. Cell homogenate from stem cells derived from growing deer antlers, a method of obtaining it and its use
ES2646750T3 (en) * 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
US20110212065A1 (en) 2010-02-18 2011-09-01 Timothy Jansen Methods of manufacture of therapeutic products comprising vitalized placental dispersions
EP3769695A1 (en) * 2010-02-19 2021-01-27 LifeCell Corporation Abdominal wall treatment devices
JP6144049B2 (en) 2010-02-22 2017-06-07 バイオミメティック セラピューティクス,リミテッド ライアビリティ カンパニー Platelet-derived growth factor compositions and methods for treating tendon disorders
KR20130025375A (en) 2010-03-01 2013-03-11 얀센 바이오테크 인코포레이티드 Methods for purifying cells derived from pluripotent stem cells
TWI578993B (en) 2010-04-07 2017-04-21 安瑟吉納西斯公司 Angiogenesis using placental stem cells
TW201138792A (en) 2010-04-08 2011-11-16 Anthrogenesis Corp 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
CA2800610C (en) 2010-05-12 2019-09-24 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 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
ES2879246T3 (en) * 2010-07-02 2021-11-22 Univ North Carolina Chapel Hill Biomatrix scaffolds
EP2593117B1 (en) 2010-07-12 2019-03-20 University of Southern California Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
KR20130093091A (en) 2010-07-13 2013-08-21 안트로제네시스 코포레이션 Methods of generating natural killer cells
US8825388B2 (en) 2010-07-13 2014-09-02 Qualcomm Incorporated Indoor likelihood heatmap
SG187667A1 (en) * 2010-08-03 2013-03-28 Agency Science Tech & Res Microfabricated scaffold structures
JP2012031127A (en) * 2010-08-03 2012-02-16 Nagoya Univ Composition including umbilical cord matrix stromal cell
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
CN103154237B (en) 2010-08-31 2016-03-16 詹森生物科技公司 The differentiation of multipotential stem cell
EP2853589B1 (en) 2010-08-31 2017-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
KR101851956B1 (en) 2010-08-31 2018-04-25 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
JP2013536860A (en) * 2010-08-31 2013-09-26 クック・ジェネラル・バイオテクノロジー・エルエルシー Systemic, allogeneic stem cell therapy for the treatment of animal diseases
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
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
EP2658557A1 (en) 2010-12-31 2013-11-06 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
CN102614546B (en) * 2011-01-26 2014-10-15 香港中文大学 Cell sheet for tissue repair and bio-artificial tissue engineering, method of producing the same and method of using the same
ES2613547T3 (en) * 2011-03-18 2017-05-24 Microvascular Tissues, Inc. Allogeneic microvascular tissue for soft tissue treatments
KR101903339B1 (en) 2011-03-22 2018-10-01 플루리스템 리미티드 Methods for treating radiation or chemical injury
CA2834496A1 (en) 2011-04-28 2012-11-01 Aquero Company, Llc Lysine-based polymer coagulants for use in clarification of process waters
WO2012149468A2 (en) 2011-04-29 2012-11-01 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
EP2714111B1 (en) * 2011-05-31 2021-03-17 LifeCell Corporation Adipose tissue matrices
TWI602570B (en) 2011-06-01 2017-10-21 安瑟吉納西斯公司 Treatment of pain using placental stem cells
CA2837878A1 (en) 2011-06-10 2012-12-13 Tissuetech, Inc. Methods of processing fetal support tissues, fetal support tissue powder products, and uses thereof
WO2012177927A1 (en) 2011-06-21 2012-12-27 Mayo Foundation For Medical Education And Research Transgenic animals capable of being induced to delete senescent cells
CN103930542A (en) 2011-06-29 2014-07-16 生物修复疗法有限公司 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
US10865383B2 (en) 2011-07-12 2020-12-15 Lineage Cell Therapeutics, Inc. Methods and formulations for orthopedic cell therapy
WO2013022447A1 (en) * 2011-08-10 2013-02-14 Advanced Technologies And Regenerative Medicine, Llc Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation 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
JP2015502366A (en) 2011-12-13 2015-01-22 バック インスティテュート フォー リサーチ オン エイジング Methods for improving drug therapy
US9162011B2 (en) 2011-12-19 2015-10-20 Allosource Flowable matrix compositions and methods
EP3549615B1 (en) 2011-12-20 2020-12-16 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
DK2793965T3 (en) 2011-12-20 2019-05-20 Lifecell Corp TISSUE PRODUCTS WITH FLYING DUTIES
BR112014015417A8 (en) 2011-12-22 2017-07-04 Janssen Biotech Inc differentiation of human embryonic stem cells into single hormone insulin positive cells
MX362198B (en) * 2011-12-23 2019-01-08 Depuy Synthes Products Llc Detection of human umbilical cord tissue-derived cells.
DK3321355T3 (en) 2011-12-30 2021-11-08 Amit Patel PROCEDURES AND COMPOSITIONS FOR CLINICAL DETERMINATION OF AN ALLOGENIC CELL AND THERAPEUTIC APPLICATIONS
WO2013112350A1 (en) 2012-01-24 2013-08-01 Lifecell Corporation Elongated tissue matrices
EP3338548B1 (en) * 2012-02-23 2020-04-08 Celularity, Inc. Identification of antitumor compounds using placenta
US8940294B2 (en) 2012-03-02 2015-01-27 Tissuetech, Inc. Methods of isolating and culturing stem cells
KR20140131999A (en) 2012-03-07 2014-11-14 얀센 바이오테크 인코포레이티드 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
EP2841115B1 (en) 2012-04-24 2019-07-10 LifeCell Corporation Functionalized tissue matrices
JP2015521054A (en) 2012-06-05 2015-07-27 カプリコール,インコーポレイテッド Optimized methods for generating cardiac stem cells from heart tissue and their use in cardiac therapy
ES2897649T3 (en) 2012-06-08 2022-03-02 Janssen Biotech Inc Differentiation of human embryonic stem cells into pancreatic endocrine cells
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
US11090338B2 (en) 2012-07-13 2021-08-17 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
CA2881394A1 (en) 2012-08-13 2014-02-20 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
US10596202B2 (en) 2012-09-19 2020-03-24 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
WO2014047067A1 (en) 2012-09-19 2014-03-27 Tornier, 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
US9872937B2 (en) 2012-09-19 2018-01-23 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
WO2014052376A1 (en) 2012-09-26 2014-04-03 Lifecell Corporation Processed adipose tissue
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
SG10201709338RA (en) 2012-12-31 2017-12-28 Janssen Biotech Inc Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
CA2896658C (en) 2012-12-31 2021-06-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
MX2015008619A (en) 2012-12-31 2016-01-12 Janssen Biotech Inc Suspension and clustering of human pluripotent cells for differentiation into pancreatic endocrine cells.
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
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
EP3622960A1 (en) 2013-02-05 2020-03-18 Celularity, Inc. 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
US10329529B2 (en) 2013-03-14 2019-06-25 Celularity, Inc. Enhanced placental stem cells and uses thereof
EP2970882B1 (en) 2013-03-15 2018-11-28 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
JP2016523125A (en) 2013-05-30 2016-08-08 グラハム エイチ. クリーシー Local nervous 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
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
CA2932271C (en) 2013-12-06 2024-02-06 Allosource Methods 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
US20170216286A1 (en) 2014-01-28 2017-08-03 Mayo Foundation For Medical Education And Research Killing senescent cells and treating senescence-associated conditions using a src inhibitor and a flavonoid
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
WO2015116740A1 (en) 2014-01-28 2015-08-06 Buck Institute For Research On Aging Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders
EP3797742A1 (en) 2014-02-12 2021-03-31 Gyroscope Therapeutics Limited 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
WO2015138514A1 (en) 2014-03-10 2015-09-17 3-D Matrix, Ltd. Self-assembling peptide compositions
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
CA2948126A1 (en) * 2014-05-07 2015-11-12 Osiris Therapeutics, Inc. Therapeutic placental compositions, methods of making and methods of use
WO2015170347A2 (en) 2014-05-09 2015-11-12 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
US10219936B2 (en) 2014-09-11 2019-03-05 Orbit Biomedical Limited Therapeutic agent delivery device with advanceable cannula and needle
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
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
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
CA2962444C (en) 2014-10-03 2023-09-05 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
EP3206670A4 (en) 2014-10-14 2018-06-20 Samuel Lynch Compositions for treating wounds
KR101808762B1 (en) * 2014-10-29 2017-12-14 차의과학대학교 산학협력단 Placenta derived cell secreting C3 or C1r complement and Composition comprising the 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
EP3227682B1 (en) * 2014-12-05 2019-05-01 Meridigen Biotech Co., Ltd. Method of distinguishing mesenchymal stem cells
WO2016090215A2 (en) * 2014-12-05 2016-06-09 Janssen Biotech, Inc. Treatment of ocular conditions using progenitor cells
KR20170094368A (en) * 2014-12-16 2017-08-17 얀센 바이오테크 인코포레이티드 Treatment of retinal degeneration using progenitor cells
US20170080033A1 (en) * 2014-12-16 2017-03-23 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
JP6800854B2 (en) 2014-12-19 2020-12-16 ヤンセン バイオテツク,インコーポレーテツド Suspension culture 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
US20160243288A1 (en) 2015-02-23 2016-08-25 Tissuetech, Inc. Apparatuses and methods for treating ophthalmic diseases and disorders
SG11201706680SA (en) 2015-03-10 2017-09-28 Allergan Pharmaceuticals Holdings (Ireland) Unlimited Company Multiple needle injector
CA2979293C (en) 2015-03-11 2022-01-04 Timothy J. Kieffer Pancreatic endocrine progenitor cell therapies for the treatment of obesity and type 2 diabetes (t2d)
US11027047B2 (en) 2015-03-31 2021-06-08 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
JP2018516869A (en) 2015-05-20 2018-06-28 ティッシュテック,インク. Compositions and methods for preventing epithelial cell proliferation and epithelial-mesenchymal transition
EP3297694A1 (en) 2015-05-21 2018-03-28 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
WO2017019822A1 (en) * 2015-07-29 2017-02-02 Medivation Technologies, Inc. Pellet composition containing repair cells
WO2017019832A1 (en) 2015-07-29 2017-02-02 Medivation Technologies, Inc. Methods and compositions using repair cells and cationic dyes
CN108350427B (en) 2015-08-28 2022-03-11 日本乐敦制药株式会社 ROR1 positive mesenchymal stem cells and pharmaceutical composition comprising same
USD797290S1 (en) 2015-10-19 2017-09-12 Spinal Surgical Strategies, Llc Bone graft delivery tool
US11384328B2 (en) 2015-11-18 2022-07-12 President And Fellows Of Harvard College Cartridge-based system for long term culture of cell clusters
CN108291199A (en) * 2015-12-04 2018-07-17 詹森生物科技公司 Retinosis is treated using progenitor cells
KR20170076484A (en) * 2015-12-24 2017-07-04 삼성전자주식회사 Method of isolating senescent cell with high expression of Protocadherin gene
WO2017120092A1 (en) 2016-01-06 2017-07-13 3-D Matrix, Ltd. Combination compositions
EP3402543B1 (en) 2016-01-11 2021-09-08 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
WO2017136557A1 (en) * 2016-02-05 2017-08-10 Petrucci Gary M 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
KR102232054B1 (en) 2016-04-08 2021-03-26 알레간 인코포레이티드 Suction 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
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US10806629B2 (en) 2016-06-17 2020-10-20 Gyroscope Therapeutics Limited Injection device for subretinal delivery of therapeutic agent
US10646374B2 (en) 2016-06-17 2020-05-12 Orbit Biomedical Limited Apparatus and method to form entry bleb 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
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
EP3777907A1 (en) 2016-07-05 2021-02-17 LifeCell Corporation Tissue matrices incorporating multiple tissue types
SG11201811100RA (en) * 2016-07-05 2019-01-30 Janssen Biotech Inc Treatment of retinal vascular disease using progenitor cells
US20190184060A1 (en) * 2016-08-19 2019-06-20 Regentys Corporation 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
EP3515459A4 (en) 2016-09-20 2020-08-05 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
EP3534981A4 (en) * 2016-11-02 2020-06-24 Axogen Corporation 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
JPWO2018164228A1 (en) 2017-03-08 2020-01-09 ロート製薬株式会社 Pharmaceutical composition for preventing or treating a disease associated with fibrosis, comprising a ROR1-positive mesenchymal stem cell, a method for preparing the same, and prevention or treatment of a disease associated with fibrosis using ROR1-positive mesenchymal stem cell Treatment method
US11076984B2 (en) 2017-03-13 2021-08-03 Gyroscope Therapeutics Limited Method of performing subretinal drainage and agent delivery
EP3595680A1 (en) * 2017-03-17 2020-01-22 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
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
KR20190141695A (en) 2017-04-13 2019-12-24 센티 바이오사이언시스, 인코포레이티드 Combination Cancer Immunotherapy
EP3612191A4 (en) 2017-04-19 2020-12-30 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
JP7195033B2 (en) 2017-07-18 2022-12-23 ザ リサーチ ファウンデイション フォー ザ ステイト ユニバーシティー オブ ニューヨーク Biomarkers for intracranial aneurysms
US10251917B1 (en) 2017-09-19 2019-04-09 Gary M. Petrucci Methods and materials for treating tumors
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
AU2018351051A1 (en) 2017-10-18 2020-03-19 Lifecell Corporation Adipose tissue products and methods of production
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
WO2019079681A1 (en) 2017-10-20 2019-04-25 President And Fellows Of Harvard College Methods for producing mature adipocytes and methods of use thereof
WO2019083995A1 (en) * 2017-10-23 2019-05-02 Cell Medicine, Inc. Mesenchymal stem cell therapy of leigh syndrome
WO2019087130A2 (en) * 2017-11-03 2019-05-09 Janssen Biotech, Inc. Method of inhibiting angiogenesis
WO2019094365A1 (en) 2017-11-07 2019-05-16 Neurostim Oab, Inc. Non-invasive nerve 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
CN111565732A (en) 2018-01-12 2020-08-21 国立大学法人大阪大学 Agent for promoting normal differentiation-maturation of stratified squamous epithelial cell, agent for treating epithelial disease, and method for promoting normal differentiation-maturation of stratified squamous epithelial cell
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
WO2020081869A1 (en) 2018-10-17 2020-04-23 Senti Biosciences, Inc. Combinatorial cancer immunotherapy
JP2022520722A (en) 2019-01-23 2022-04-01 アステリアス バイオセラピューティクス インコーポレイテッド Dorsal-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
MX2021014654A (en) 2019-05-30 2022-03-11 Lifecell Corp Biologic breast implant.
EP3990100A4 (en) 2019-06-26 2023-07-19 Neurostim Technologies LLC Non-invasive nerve activator with adaptive circuit
CN114286860A (en) * 2019-08-29 2022-04-05 味之素株式会社 Method for producing mesenchymal stem cells from biological 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
WO2021071875A1 (en) * 2019-10-07 2021-04-15 University Of Utah Research Foundation Chondrogenic human mesenchymal stem cell (msc) sheets
IL292024A (en) 2019-10-11 2022-06-01 Gyroscope Therapeutics Ltd Dose clip assembly for syringe
JP2023506713A (en) 2019-12-16 2023-02-20 ニューロスティム テクノロジーズ エルエルシー Noninvasive nerve activator using booster charge delivery
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
EP4279919A1 (en) * 2021-01-12 2023-11-22 Cellatoz Therapeutics, Inc. Method for screening neuronal regeneration promoting cells having neuronal 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
WO2024052733A1 (en) 2022-09-06 2024-03-14 Gyroscope Therapeutics Limited Apparatus for subretinal administration of therapeutic agent via dual-curved needle

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
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
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
US5266480A (en) 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
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
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
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
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
JPH06104061B2 (en) 1989-02-10 1994-12-21 花王株式会社 Cell culture support material
DE69009237T2 (en) * 1989-02-27 1994-12-01 Takasago Perfumery Co Ltd Process for the preparation of optically active esters of 6-t-butoxy-3,5-dihydroxyhexanoic acid.
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
WO1992003917A1 (en) 1990-08-29 1992-03-19 Genpharm International 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
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
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.
CZ232593A3 (en) 1991-05-02 1994-07-13 Yeda Res & Dev Pharmaceutical preparation for preventing and/or therapy of pathological states
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
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young 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
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
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
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
US5834308A (en) 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
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
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
DE69531638T2 (en) 1994-06-06 2004-06-17 Osiris Therapeutics, Inc. BIOMATRIX FOR TISSUE REGENATION
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
JP4307552B2 (en) 1996-03-15 2009-08-05 ミューニン コーポレイション Extracellular matrix signaling molecules
US6223188B1 (en) * 1996-04-10 2001-04-24 Sun Microsystems, Inc. Presentation of link information as an aid to hypermedia navigation
WO1997041208A1 (en) 1996-04-26 1997-11-06 Case Western Reserve University Skin regeneration using 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
WO1998031316A1 (en) 1997-01-17 1998-07-23 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
EP2110431A1 (en) 1997-05-13 2009-10-21 Osiris Therapeutics, Inc. 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
AU738334B2 (en) * 1997-05-30 2001-09-13 Osteobiologics, Inc. Fiber-reinforced, porous, biodegradable implant device
ATE307195T1 (en) 1997-07-14 2005-11-15 Osiris Therapeutics Inc CARDIAC MUSCLE REGENERATION USING MESENCHYMAL STEM CELLS
US5902598A (en) * 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
EP1036163B1 (en) 1997-12-02 2010-08-18 Artecel Sciences, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
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
EP1076563B1 (en) 1998-03-16 2005-05-11 Cytovia, Inc. Dipeptide caspase inhibitors and the use thereof
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
AU3888699A (en) * 1998-05-07 1999-11-23 University Of South Florida Bone marrow cells as a source of neurons for brain and spinal cord repair
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
AU776514B2 (en) 1998-08-10 2004-09-09 General Hospital Corporation, The 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
WO2000038762A1 (en) 1998-12-24 2000-07-06 Biosafe S.A. Blood separation system particularly 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
CA2359821A1 (en) 1999-02-04 2000-08-10 Mcgill University Platform for the differentiation of cells
AU780794B2 (en) 1999-02-10 2005-04-14 Es Cell International Pte Ltd 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
CA2369758C (en) 1999-04-16 2007-04-03 Wm. Marsh Rice University Functionalized poly(propylene fumarate) and poly(propylene fumarate-co-ethylene glycol)
US6261600B1 (en) * 1999-04-30 2001-07-17 Drugtech Corporation Folic acid supplement
US6372494B1 (en) * 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
US6287340B1 (en) 1999-05-14 2001-09-11 Trustees Of Tufts College Bioengineered anterior cruciate ligament
AU4860900A (en) 1999-06-02 2000-12-18 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6329904B1 (en) * 1999-06-11 2001-12-11 Safety Through Cellular, Inc. Apparatus and method for providing weather and other alerts
US6261841B1 (en) * 1999-06-25 2001-07-17 The Board Of Trustees Of Northwestern University Compositions, kits, and methods for modulating survival and differentiation of multi-potential hematopoietic progenitor cells
US6355699B1 (en) 1999-06-30 2002-03-12 Ethicon, Inc. Process for manufacturing biomedical foams
US6333029B1 (en) 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US7621606B2 (en) 2001-08-27 2009-11-24 Advanced Cell Technology, Inc. Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
JP5398941B2 (en) 1999-08-05 2014-01-29 エイビーティー ホールディング カンパニー Pluripotent adult stem cell and method for isolating the same
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
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
AU7378600A (en) 1999-09-14 2001-04-17 Children's Medical Center Corporation Methods for treating muscular dystrophy
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
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
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
US6866843B2 (en) * 1999-12-06 2005-03-15 Viacell, Inc. Method of transplanting in a mammal and treating diabetes mellitus by administering a pseudo-islet like aggregate differentiated from a nestin-positive pancreatic stem cell
WO2001053503A1 (en) 2000-01-18 2001-07-26 Cornell Research Foundation, Inc. Neural 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
JP2003521935A (en) 2000-02-11 2003-07-22 フイラデルフイア・ヘルス・アンド・エデユケーシヨン・コーポレーシヨン Differentiation of bone marrow cells into neuronal cells and uses therefor
ATE473751T1 (en) 2000-02-11 2010-07-15 Schepens Eye Res Inst ISOLATION AND TRANSPLANTATION OF RETINAL STEM CELLS
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
AU2001245550A1 (en) * 2000-03-09 2001-09-17 Lee Walters Applications of immune system tolerance to treatment of various diseases
EP2298858A1 (en) 2000-03-16 2011-03-23 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
US20030212024A1 (en) 2000-05-12 2003-11-13 Keating Mark T 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
JP4679795B2 (en) * 2000-07-21 2011-04-27 株式会社セルシード Myocardial cell sheet, three-dimensional structure, myocardial tissue, and production method thereof
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
WO2002026941A2 (en) 2000-09-29 2002-04-04 Kooy Derek V D 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
US20020168763A1 (en) 2000-11-30 2002-11-14 Yan Wen Liang 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
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem 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
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
WO2002053193A2 (en) 2001-01-02 2002-07-11 The Charles Stark Draper Laboratory, Inc. Tissue engineering of three-dimensional vascularized using microfabricated polymer assembly technology
EP1366148A2 (en) 2001-01-24 2003-12-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE DEPARTMENT OF HEALTH &amp; HUMAN SERVICES 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
EP1367961B1 (en) 2001-02-06 2008-12-31 Massachusetts Institute Of Technology Peptide scaffold encapsulation of tissue cells and uses thereof
US7449180B2 (en) 2001-02-06 2008-11-11 John Kisiday Macroscopic scaffold containing amphiphilic peptides encapsulating cells
KR101132545B1 (en) * 2001-02-14 2012-04-02 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
EP1362095B1 (en) 2001-02-14 2015-05-27 Anthrogenesis Corporation 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
US20030008820A1 (en) * 2001-03-27 2003-01-09 Massachusetts Institute Of Technology Methods and products related to FGF dimerization
US7202080B2 (en) 2001-03-29 2007-04-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
US7838292B1 (en) 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
EP1379626A2 (en) 2001-04-19 2004-01-14 DeveloGen Aktiengesellschaft für entwicklungsbiologische Forschung A 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
CA2451838A1 (en) * 2001-05-25 2002-12-05 Cythera, Inc. Stem cell differentiation
WO2002098365A2 (en) 2001-06-07 2002-12-12 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
WO2003014317A2 (en) * 2001-08-08 2003-02-20 Celmed Biosciences Usa Compositions and methods for isolation, propagation, and differentiation of 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
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
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
EP1298201A1 (en) 2001-09-27 2003-04-02 Cardion AG Process for the production of cells exhibiting an islet-beta-cell-like state
EP1446477A4 (en) 2001-09-28 2006-06-07 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
JP2005506074A (en) * 2001-10-18 2005-03-03 イクシオン・バイオテクノロジー・インコーポレーテッド Conversion of hepatic stem and progenitor cells into functional pancreatic cells
US7129034B2 (en) * 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
WO2003039489A2 (en) * 2001-11-09 2003-05-15 Artecel Sciences, Inc. Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
AU2002365162A1 (en) 2001-11-14 2003-07-09 Northwestern University Self-assembly and mineralization of peptide-amphiphile nanofibers
JP4330995B2 (en) 2001-11-15 2009-09-16 チルドレンズ メディカル センター コーポレーション Methods for isolating, proliferating, and differentiating fetal stem cells from chorionic villi, amniotic fluid, and placenta, and methods for their therapeutic use
JP3728750B2 (en) * 2001-11-22 2005-12-21 ニプロ株式会社 Cultured skin and method for producing the same
CA2466478A1 (en) 2001-11-28 2003-06-05 Anges Mg, Inc. Gene therapy agents for neurodegenerative diseases
US6712850B2 (en) 2001-11-30 2004-03-30 Ethicon, Inc. Porous tissue scaffolds for the repair and regeneration of dermal tissue
JP2005511046A (en) 2001-12-04 2005-04-28 オーガノジェネシス インコーポレーテッド Cultured cells derived from 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
EP2264146A1 (en) 2001-12-07 2010-12-22 Geron Corporation Islet cells from human embryonic stem cells
AU2002364558A1 (en) * 2001-12-11 2003-06-23 Cytograft Tissue Engineering, Inc. 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
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
JP2005512592A (en) * 2001-12-21 2005-05-12 マウント・シナイ・ホスピタル Cellular composition, method for producing cellular composition, and method of using cellular composition
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
CA2473503C (en) 2002-01-14 2010-01-05 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
KR101176146B1 (en) * 2002-02-13 2012-08-22 안트로제네시스 코포레이션 Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
WO2003070749A2 (en) 2002-02-15 2003-08-28 Northwestern University Self-assembly of peptide-amphiphile nanofibers under physiological conditions
US7576065B2 (en) 2002-02-15 2009-08-18 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
US20050118714A1 (en) 2002-02-19 2005-06-02 Chul-Won Ha Isolation and culture-expansion methods of mesenchymal stem/progenitor cells from umbilical cord blood and differentation method of umbilical cord blood-derived mesenchymal stem/progenitor cells into various mesenchymal tissues
WO2003072728A2 (en) 2002-02-22 2003-09-04 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
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
EP1538913A2 (en) 2002-04-12 2005-06-15 Celgene 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
US20030235563A1 (en) 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
JP2005526838A (en) 2002-04-25 2005-09-08 ウイスコンシン アラムナイ リサーチ フオンデーシヨン Use of GDNF-secreting human neural stem cells to treat Parkinson's disease 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
AU2003233119A1 (en) * 2002-05-08 2003-11-11 Neuronova Ab Modulation of neural stem cells with s1p or lpa receptor agonists
BR0311360A (en) 2002-05-28 2006-06-06 Becton Dickinson Co methods for in vitro expansion and transdifferentiation of human pancreatic acinar cells into insulin producing cells
CN1668733A (en) 2002-05-30 2005-09-14 细胞基因公司 Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
WO2003104442A1 (en) 2002-06-07 2003-12-18 Es Cell International Pte Ltd Methods of regulating differentiation in stem cells
AU2003247514A1 (en) * 2002-06-11 2003-12-22 Roy Ogle Meningeal-derived stem cells
WO2004003561A1 (en) 2002-06-27 2004-01-08 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
US20050249731A1 (en) 2002-07-16 2005-11-10 Hadi Aslan Methods of implating mesenchymal stem cells for tissue repair and 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
CA2493363A1 (en) 2002-07-29 2004-02-05 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
AU2003262628A1 (en) 2002-08-14 2004-03-03 University Of Florida Bone marrow cell differentiation
US20060128014A1 (en) 2002-08-26 2006-06-15 Johan Haggblad Compositions and methods for culturing stem cells
WO2004020601A2 (en) * 2002-08-28 2004-03-11 University Of Florida Neurogenesis from hepatic stem cells
US7371576B2 (en) 2002-09-06 2008-05-13 Reneuron, Inc. CD56 positive human adult pancreatic endocrine progenitor cells
US6766863B2 (en) * 2002-09-20 2004-07-27 Hypro Corporation Fire fighting foam injection system with auto-start feature
US9969977B2 (en) * 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
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
AU2003287444A1 (en) 2002-10-31 2004-05-25 The General Hospital Corporation Repairing or replacing tissues or organs
AU2003296338A1 (en) 2002-12-05 2004-06-30 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
AU2004210853A1 (en) 2003-02-11 2004-08-26 Northwestern University Methods and materials for nanocrystalline surface coatings and attachment of peptide amphiphile nanofibers thereon
CA2515469C (en) 2003-02-11 2013-12-17 John E. Davies Progenitor cells from wharton's jelly of human umbilical cord
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
EP1636351A4 (en) 2003-05-21 2007-04-04 Gen Hospital Corp Microfabricated compositions and processes for engineering tissues containing multiple cell types
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue 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
US7413734B2 (en) 2003-06-27 2008-08-19 Ethicon, Incorporated Treatment of retinitis pigmentosa with human umbilical cord cells
US9572840B2 (en) * 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
CA2536909A1 (en) 2003-08-29 2005-03-10 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
CA2548305C (en) 2003-12-23 2014-07-15 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
WO2006101548A2 (en) 2004-12-21 2006-09-28 Ethicon, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060166361A1 (en) 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
AU2005322068B2 (en) 2004-12-23 2011-09-01 Ethicon Incorporated Treatment of stroke and other acute neural degenerative disorders using postpartum derived cells
EP1831355A2 (en) 2004-12-23 2007-09-12 Ethicon, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
EP1838842A2 (en) 2004-12-23 2007-10-03 Ethicon, Incorporated Treatment of osteochondral diseases using postpartum-derived cells and products thereof
PL1831356T3 (en) * 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US7741311B2 (en) 2005-01-03 2010-06-22 Shaker Mousa Composition and method for treating occlusive vascular diseases, nerve regeneration, and wound healing
AU2006208944A1 (en) 2005-01-28 2006-08-03 Imperial College Innovations Limited Methods for embryonic stem cell culture
JP5017253B2 (en) 2005-03-31 2012-09-05 ステムニオン,インコーポレイテッド Amnion-derived cell composition, preparation method and use thereof
US7923007B2 (en) 2005-08-08 2011-04-12 Academia Sinica Brain tissue damage therapies
WO2007070870A1 (en) 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
WO2007073552A1 (en) 2005-12-19 2007-06-28 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
WO2007076522A2 (en) * 2005-12-28 2007-07-05 Ethicon, Incorporated 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
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
WO2007081841A2 (en) 2006-01-06 2007-07-19 Stratagene California Reaction buffer composition for nucleic acid replication with packed dna polymerases
PT2366775E (en) 2006-03-23 2015-07-20 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
WO2007146106A2 (en) * 2006-06-05 2007-12-21 Cryo- Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
PT2078073E (en) 2006-10-12 2013-11-11 Ethicon Inc Kidney-derived cells and methods of use in tissue repair and regeneration
JP5670053B2 (en) 2006-11-13 2015-02-18 エシコン・インコーポレイテッドEthicon, Incorporated In vitro expansion of postpartum-derived cells using microcarriers
NZ597779A (en) 2007-02-12 2013-07-26 Anthrogenesis Corp 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
UA99152C2 (en) 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
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
US20090186358A1 (en) 2007-12-21 2009-07-23 Wyeth Pathway Analysis of Cell Culture Phenotypes and Uses Thereof
WO2009085860A1 (en) 2007-12-27 2009-07-09 Ethicon, Incorporated Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
TWI363852B (en) * 2008-03-28 2012-05-11 Ind Tech Res Inst Stacked light emitting device
ES2520393T3 (en) 2008-12-19 2014-11-11 DePuy Synthes Products, LLC Cells derived from umbilical cord tissue to treat neuropathic pain and spasticity
CA2747794C (en) 2008-12-19 2018-10-30 Advanced Technologies And Regenerative Medicine, Llc Treatment of lung and pulmonary diseases and disorders
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
JP6095893B2 (en) 2008-12-19 2017-03-15 デピュイ・シンセス・プロダクツ・インコーポレイテッド Regeneration and repair of nerve tissue after injury
BRPI1013409A2 (en) 2009-03-26 2018-01-16 Advanced Tech And Regenerative Medicine Llc human umbilical cord tissue cells as therapy for alzheimer's disease
CN103329308B (en) * 2011-01-19 2016-09-14 纳幕尔杜邦公司 There is the lithium battery separator of closing function

Also Published As

Publication number Publication date
WO2005001077A2 (en) 2005-01-06
WO2005003334A3 (en) 2005-04-07
ES2554343T3 (en) 2015-12-18
EP1641914A2 (en) 2006-04-05
US20060154367A1 (en) 2006-07-13
US20170143770A1 (en) 2017-05-25
US20050058631A1 (en) 2005-03-17
EP2322599B1 (en) 2015-04-15
CA2530732A1 (en) 2005-01-06
US9579351B2 (en) 2017-02-28
US10383898B2 (en) 2019-08-20
PL1641917T3 (en) 2016-11-30
US9717763B2 (en) 2017-08-01
PL1641916T3 (en) 2016-08-31
WO2005001079A2 (en) 2005-01-06
EP1641918A2 (en) 2006-04-05
US20100210013A1 (en) 2010-08-19
US20070009494A1 (en) 2007-01-11
PL1641915T3 (en) 2017-01-31
US20170166864A1 (en) 2017-06-15
US8277796B2 (en) 2012-10-02
ES2542070T3 (en) 2015-07-30
AU2004252570C1 (en) 2012-03-01
JP2007528706A (en) 2007-10-18
ES2564044T3 (en) 2016-03-17
WO2005001076A2 (en) 2005-01-06
EP2341131A2 (en) 2011-07-06
JP2007521793A (en) 2007-08-09
EP2338980A2 (en) 2011-06-29
CA2530533C (en) 2015-02-10
AU2004254616A1 (en) 2005-01-13
CA2530421A1 (en) 2005-01-06
EP1641913B1 (en) 2016-01-06
EP1641916B1 (en) 2016-02-17
AU2004281371A1 (en) 2005-04-28
PL1641913T3 (en) 2016-06-30
US10500234B2 (en) 2019-12-10
PL2338980T3 (en) 2016-04-29
US7524489B2 (en) 2009-04-28
US10758576B2 (en) 2020-09-01
WO2005001078A3 (en) 2005-04-21
US20060154366A1 (en) 2006-07-13
US7510873B2 (en) 2009-03-31
PL2338981T3 (en) 2016-01-29
ES2552226T3 (en) 2015-11-26
JP4950659B2 (en) 2012-06-13
CA2530412A1 (en) 2005-04-28
EP2399990B1 (en) 2015-07-22
US20170166863A1 (en) 2017-06-15
PL1649013T3 (en) 2016-07-29
CA2530732C (en) 2015-03-31
ES2569780T3 (en) 2016-05-12
WO2005003334A2 (en) 2005-01-13
ES2565582T3 (en) 2016-04-05
EP2338980B1 (en) 2015-04-22
US20050032209A1 (en) 2005-02-10
EP2322599A2 (en) 2011-05-18
US20070014771A1 (en) 2007-01-18
JP4950660B2 (en) 2012-06-13
PL2336298T3 (en) 2016-08-31
WO2005001079A3 (en) 2005-04-28
AU2004254616B2 (en) 2011-09-01
EP2338982A2 (en) 2011-06-29
US20060153816A1 (en) 2006-07-13
US10220059B2 (en) 2019-03-05
ES2550267T3 (en) 2015-11-05
EP1641915A2 (en) 2006-04-05
EP1649013A2 (en) 2006-04-26
ES2542069T3 (en) 2015-07-30
PL2322599T3 (en) 2015-10-30
EP2338981B1 (en) 2015-04-22
WO2005001077A3 (en) 2005-03-03
US11179422B2 (en) 2021-11-23
AU2004252571C1 (en) 2012-03-01
ES2568463T3 (en) 2016-04-29
AU2004252568A1 (en) 2005-01-06
AU2004254616C1 (en) 2012-03-01
AU2004252566A1 (en) 2005-01-06
EP2322599A3 (en) 2011-07-27
US10195233B2 (en) 2019-02-05
US20170326183A1 (en) 2017-11-16
WO2005038012A3 (en) 2005-09-15
US20100260843A1 (en) 2010-10-14
EP1641918B1 (en) 2016-01-06
US8318483B2 (en) 2012-11-27
AU2004252566B2 (en) 2011-09-15
AU2004252567B2 (en) 2011-10-06
PL1641914T3 (en) 2017-01-31
JP2007528705A (en) 2007-10-18
JP2007528703A (en) 2007-10-18
EP1641917B1 (en) 2016-04-27
PL1641918T3 (en) 2016-06-30
US20170239299A1 (en) 2017-08-24
WO2005001080A2 (en) 2005-01-06
US11191789B2 (en) 2021-12-07
US11000554B2 (en) 2021-05-11
JP4948165B2 (en) 2012-06-06
EP2341131A3 (en) 2011-08-03
EP1641913A2 (en) 2006-04-05
CA2530422C (en) 2016-01-05
EP2336298A3 (en) 2011-08-03
JP5148873B2 (en) 2013-02-20
PL2341131T3 (en) 2016-01-29
US20050054098A1 (en) 2005-03-10
US8658152B2 (en) 2014-02-25
JP2007521008A (en) 2007-08-02
AU2004252571B2 (en) 2011-09-15
EP2336298B1 (en) 2016-02-17
EP2338980A3 (en) 2011-08-10
ES2600555T3 (en) 2017-02-09
WO2005038012A2 (en) 2005-04-28
US20050058629A1 (en) 2005-03-17
US20060188983A1 (en) 2006-08-24
US9498501B2 (en) 2016-11-22
ES2597837T3 (en) 2017-01-23
EP1641914B1 (en) 2016-07-20
CA2530412C (en) 2016-02-09
AU2004252570A1 (en) 2005-01-06
CA2530421C (en) 2015-04-21
PL2338982T3 (en) 2016-01-29
US20140045263A1 (en) 2014-02-13
EP1649013B1 (en) 2016-01-20
ES2541604T3 (en) 2015-07-22
EP2338982B1 (en) 2015-08-26
EP2341131B1 (en) 2015-08-05
JP4950661B2 (en) 2012-06-13
AU2004281371C1 (en) 2012-10-11
US20190192577A1 (en) 2019-06-27
EP2399990A1 (en) 2011-12-28
EP2338981A3 (en) 2011-08-03
US20050037491A1 (en) 2005-02-17
US20070036767A1 (en) 2007-02-15
JP2007521009A (en) 2007-08-02
CA2530416C (en) 2015-08-25
AU2004252567A1 (en) 2005-01-06
ES2582342T3 (en) 2016-09-12
EP2336298A2 (en) 2011-06-22
PL2399990T3 (en) 2015-12-31
WO2005001078A2 (en) 2005-01-06
US8703121B2 (en) 2014-04-22
JP2007528702A (en) 2007-10-18
US20050058630A1 (en) 2005-03-17
CA2530255A1 (en) 2005-01-06
US20060153817A1 (en) 2006-07-13
US20060153818A1 (en) 2006-07-13
JP4948166B2 (en) 2012-06-06
JP4948164B2 (en) 2012-06-06
US7413734B2 (en) 2008-08-19
AU2004252571A1 (en) 2005-01-06
AU2004252570B2 (en) 2011-09-15
US10039793B2 (en) 2018-08-07
EP1641915B1 (en) 2016-07-27
CA2530255C (en) 2015-12-08
EP2338982A3 (en) 2011-08-03
AU2004252568B2 (en) 2011-06-30
US7560276B2 (en) 2009-07-14
WO2005001076A3 (en) 2005-03-31
US9504719B2 (en) 2016-11-29
CA2530416A1 (en) 2005-01-06
US20130022585A1 (en) 2013-01-24
EP1641916A2 (en) 2006-04-05
US20050019865A1 (en) 2005-01-27
AU2004281371B2 (en) 2012-05-10
WO2005001080A3 (en) 2005-03-24
US20180338999A1 (en) 2018-11-29
ES2564045T3 (en) 2016-03-17
CA2530533A1 (en) 2005-01-13
EP2338981A2 (en) 2011-06-29
EP1641917A2 (en) 2006-04-05
US20060234376A1 (en) 2006-10-19

Similar Documents

Publication Publication Date Title
US8703121B2 (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
AU2006325710A1 (en) Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20220627