CA2530412A1 - Cartilage and bone repair and regeneration using postpartum-derived cells - Google Patents

Cartilage and bone repair and regeneration using postpartum-derived cells Download PDF

Info

Publication number
CA2530412A1
CA2530412A1 CA002530412A CA2530412A CA2530412A1 CA 2530412 A1 CA2530412 A1 CA 2530412A1 CA 002530412 A CA002530412 A CA 002530412A CA 2530412 A CA2530412 A CA 2530412A CA 2530412 A1 CA2530412 A1 CA 2530412A1
Authority
CA
Canada
Prior art keywords
cell
cells
bone
derived
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
CA002530412A
Other languages
French (fr)
Other versions
CA2530412C (en
Inventor
Anthony J. Kihm
Agnieszka Seyda
Sridevi Dhanaraj
Ziwei Wang
Alexander M. Harmon
Ian Ross Harris
Darin J. Messina
Sanjay Mistry
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 CA2530412A1 publication Critical patent/CA2530412A1/en
Application granted granted Critical
Publication of CA2530412C publication Critical patent/CA2530412C/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 isolation and induction to differentiate to cells of a chondrogenic or osteogenic phenotype are provided by the invention. The invention further provides cultures and compositions of the postpartum-derived cells and products related thereto. The postpartum-derived cells of the invention and products related thereto have a plethora of uses, including but not limited to research, diagnostic, and therapeutic applications, for example, in the treatment of bone and cartilage conditions.

Description

2 PCT/US2004/020958 CARTILAGE AND BONE REPAIR AND REGENERATION
USING POSTPARTUM-DERIVED CELLS
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. Other 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 NP5], filed June 25, 2004, U.S.
Application No. [ETH-5073 NP7], filed June 25, 2004, and U.S. Provisional Application No.
60/555,908 [ETH 5127], filed March 24, 2004. [ETH 5127], filed March 24, 2004.
FIELD OF THE INVENTION
This invention relates to the field of mammalian cell biology and cell culture. In particular, the invention relates to cells derived from postpartum tissue having the potential to differentiate into chondrogenic and osteogenic lineages, and methods of preparation and use of those postpartum tissue-derived cells, including cell-based therapies for conditions of bone and cartilage.
BACKGROUND OF THE INVENTION
Diseases and conditions of bone and cartilage affect a large portion of the population.
Three types of cartilage are present in mammals and include: hyaline cartilage, fibrocartilage, and elastic cartilage. Hyaline cartilage consists of a gristly mass having a firm, elastic consistency, is translucent and pearly blue in color. Hyaline cartilage is predominantly found on the articulating surfaces of articulating joints. It is found also in epiphyseal plates, costal cartilage, tracheal cartilage, bronchial cartilage and nasal cartilage.
Fibrocartilage is essentially the same as hyaline cartilage except that it contains fibrils of type I
collagen that add tensile strength to the cartilage. The collagenous fibers are arranged in bundles, with the cartilage cells located between the bundles. Fibrocartilage is found commonly in the annulus fibrosis of the invertebral disc, tendonous and ligamentous insertions, menisci, the symphysis pubis, and insertions of joint capsules. Elastic cartilage also is similar to hyaline cartilage except that it contains fibers of elastin. It is more opaque than hyaline cartilage and is more flexible and pliant.
These characteristics are defined in part by the elastic fibers embedded in the cartilage matrix.
Typically, elastic cartilage is present in the pinna of the ears, the epiglottis, and the larynx.
The surfaces of articulating bones in mammalian joints are covered with articular cartilage. The articular cartilage prevents direct contact of the opposing bone surfaces and permits the near frictionless movement of the articulating bones relative to one another. Two types of articular cartilage defects are commonly observed in mammals and include full-thickness and partial-thickness defects. The two types of defects differ not only in the extent of physical damage but also in the nature of repair response each type of lesion elicits.
Full-thickness articular cartilage defects include damage to the articular cartilage, the underlying subchondral bone tissue, and the calcified layer of cartilage located between the articular cartilage and the subchondral bone. Full-thickness defects typically arise during severe trauma of the joint or during the late stages of degenerative joint diseases, for example, during osteoarthritis. Since the subchondral bone tissue is both innervated and vascularized, damage to this tissue is often painful. The repair reaction induced by damage to the subchondral bone usually results in the formation of fibrocartilage at the site of the full-thickness defect.
Fibrocartilage, however, lacks the biomechanical properties of articular cartilage and fails to persist in the joint on a long term basis.
Partial-thickness articular cartilage defects are restricted to the cartilage tissue itself.
These defects usually include fissures or clefts in the articulating sunace of the cartilage. Partial-thickness defects are caused by mechanical arrangements of the joint which in turn induce wearing of the cartilage tissue within the joint. In the absence of innervation and vasculature, partial-thickness defects do not elicit repair responses and therefore tend not to heal. Although painless, partial-thickness defects often degenerate into full-thickness defects.
Cartilage may develop abnormally or may be damaged by disease, such as rheumatoid arthritis or osteoarthritis, or by trauma, each of which can lead to physical deformity and debilitation. Whether cartilage is damaged from trauma or congenital anomalies, its successful clinical regeneration is often poor at best, as reviewed by Howell, et al.
Osteoarthritis: Diagnosis and Management, 2nd ed., (Philadelphia, W. B. Saunders, 1990) and Kelley, et al. Textbook of Rheumatology, 3rd ed., (Philadelphia, W. B. Saunders, 1959).

Bone conditions also are widespread. For example, there generally are two types of bone conditions: non-metabolic bone conditions, such as bone fractures, bone/spinal deformation, osteosarcoma, myeloma, bone dysplasia and scoliosis, and metabolic bone conditions, such as osteoporosis, osteomalacia, rickets, fibrous osteitis, renal bone dystrophy and Paget's disease of bone. Osteoporosis, a metabolic bone condition, is a systemic disease characterized by increased bone fragility and fracturability due to decreased bone mass and change in fine bone tissue structure, its major clinical symptoms including spinal kyphosis, and fractures of dorsolumbar bones, vertebral centra, femoral necks, lower end of radius, ribs, upper end of humerus, and others. In bone tissue, bone formation and destruction due to bone resorption occur constantly.
Upon deterioration of the balance between bone formation and bone destruction due to bone resorption, a quantitative reduction in bone occurs. Traditionally, bone resorption suppressors such as estrogens, calcitonin and bisphosphonates have been mainly used to treat osteoporosis.
Bone grafting is often used for the treatment of bone conditions. Indeed, more than 1.4 million bone grafting procedures are performed in the developed world annually. Most of these procedures are administered following joint replacement surgery or during trauma surgical reconstruction. The success or failure of bone grafting is dependent upon a number of factors including the vitality of the site of the graft, the graft processing, and the immunological compatibility of the engrafted tissue.
In view of the prevalence of bone and cartilage conditions, novel sources of bone and cartilage tissue for therapeutic, diagnostic, and research uses are in high demand.
SUMMARI~ OF THE INVENTION
The invention is generally directed to 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 into a cell of osteocyte or chondrocyte phenotypes.
In some embodiments, the present invention provides cells derived from human postpartum tissue substantially free of blood, capable of self-renewal and expansion in culture, having the potential to differentiate into a cell of an osteogenic or chondrogenic phenotype;
requiring L-valine for growth; capable of growth in about 5% to about 20%
oxygen; and further having at least one of the following characteristics:
production of at least one of GCP-2, tissue factor, vimentin, and alpha-smooth muscle actin;
lack of production of at least one of NOGO-A, GRO-alpha or oxidized low density lipoprotein receptor, as detected by flow cytometry;
-3-production of at least one of CD 10, CD 13, CD44, CD73, CD90, PDGFr-alpha, PD-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, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for 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;
tumor necrosis factor, alpha-induced protein 3 or expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor l, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and 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; cDNA DKFZp586M2022 (from clone DKFZp586M202?); mesenchyme homeobox 2;
sine oculis homeobox homolog 1; crystallin, alpha B; dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin;
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;
hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C; Iroquois homeobox protein 5;
hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; 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;
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); cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C);
hypothetical protein FLJ14054; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane
-4-protein 5; 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;
secretion of at least one of MCP-l, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIPla, RANTES, and T)IUV1P1;
lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIPIb, I309, MDC, and VEGF, as detected by ELISA; and the ability to undergo at least 40 population doublings in culture.
In certain embodiments, the postpartum-derived cell is an umbilical cord-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 LTMB 022803 (P17) (ATCC Accession No. PTA-6068). The postpartum-derived cells of the invention are preferably human cells.
The cells may be induced to differentiate to an osteogenic or chondrogenic phenotype.
Methods for inducing differentiation of postpartum-derived cells of the invention are contemplated. For example, the cells may be induced to differentiate to a cell having an osteogenic or chondrogenic phenotype. Methods of inducing differentiation of the cells of the invention preferably involve exposing the cells to one or more differentiation-inducing agents.
Osteogenesis inducing agents of the invention include bone morphogenic proteins (e.g., BMP-2, BMP-4) and transforming growth factor (TGF)-betal, and combinations thereof.
Chondrogenesis inducing agents of the invention include TGF-beta3, GDF-5, and a combination thereof. The invention includes the differentiation-induced cells and populations, compositions, and products thereof. Differentiation-induced cells of an osteogenic lineage preferably express at least one osteogenic lineage marker (e.g., osteocalcin, bone sialoprotein, alkaline phosphatase). Differentiation of PPDCs to an osteogenic lineage may be assessed by any means known in the art, for example but not limited to, measurement of mineralization (e.g., von Kossa staining). Differentiation-induced cells of a chondrogenic lineage preferably express at least one chondrogenic lineage marker (e.g., glycosaminoglycan, type II collagen).
Differentiation of PPDCs to a chondrogenic lineage may be assessed by any means known in the art, for example but not limited to, Safranin-O or hematoxylin/eosin staining.
-5-Populations of PPDCs are provided by the invention. The PPDCs may be differentiation-induced or undifferentiated. In some embodiments, a population of postpartum-derived cells is mixed with another population of cells. In some embodiments, the cell population is heterogeneous. A heterogeneous cell population of the invention may comprise at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% undifferentiated or differentiation-induced PPDCs of the invention. The heterogeneous cell populations of the invention may further comprise bone marrow cells, stem cells, chondroblasts, chondrocytes, osteoblasts, osteocytes, osteoclasts, bone lining cells, or other bone or cartilage cell progenitors.
Cell populations of the invention may be substantially homogeneous, i.e., comprises substantially only PPDCs (preferably at least about 96%, 97%, 98%, 99% or more PPDCs). The homogeneous cell population of the invention may comprise umbilical cord- or placenta-derived cells. Homogeneous populations of placenta-derived cells may be of neonatal or maternal lineage. Homogeneity of a cell population may be achieved by any method known in the art, for example, by cell sorting (e.g., flow cytometry), bead separation, or by clonal expansion.
The invention also provides heterogeneous and homogeneous cell cultures containing undifferentiated or differentiation-induced postpartum-derived cells of the invention.
Some embodiments of the invention provide a matrix for implantation into a patient seeded with one or more postpartum-derived cells of the invention or containing or treated with a cell lysate, conditioned medium, or extracellular matrix thereof. The PPDCs may be differentiation-induced or undifferentiated. The matux may contain or be treated with one or more bioactive factors including anti-apoptotic agents (e.g., EPO, EPO
mimetibody, TPO, IGF-I
and IGF-II. HGF, caspase inhibitors); anti-inflammatory agents (e.g., p38 MAPK
inhibitors, TGF-beta inhibitors, statins, IL,-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLI1VIUS, and NSAIDs (non-steroidal anti-inflammatory drugs;
e.g., TEPOXALIN, TOLMETIN, SUPROFEN); immunosupressive/immunomodulatory agents (e.g., calcineurin inhibitors, such as cyclosporine, tacrolimus; mTOR inhibitors (e.g., SIROLIMUS, EVEROLIMUS); anti-proliferatives (e.g., azathioprine, mycophenolate mofetil);
corticosteroids (e.g., prednisolone, hydrocortisone); antibodies such as monoclonal anti-IL-ZRalpha receptor antibodies (e.g., basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g., anti-thymocyte globulin (ATG); anti-lymphocyte globulin (ALG); monoclonal anti-T cell antibody OKT3));
anti-thrombogenic agents (e.g., heparin, heparin derivatives, urokinase, PPack (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); and anti-
-6-oxidants (e.g., probucol, vitamin A, ascorbic acid, tocopherol, coenzyme Q-10, glutathione, L-cysteine, N-acetylcysteine.
Also encompassed within the scope of the invention are PPDC-products, including extracellular matrices of PPDCs, cell lysates (e.g., soluble cell fractions) of PPDCs, and PPDC-conditioned medium.
In some embodiments the invention provides compositions of PPDCs and one or more bioactive factors, for example, but not linuted to growth factors, chondrogenic or osteogenic differentiation inducing factors, anti-apoptotic agents (e.g., EPO, EPO
mimetibody, TPO, IGF-I
and IGF-II, HGF, caspase inhibitors); anti-inflammatory agents (e.g., p38 MAPK
inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and NSAIDs (non-steroidal anti-inflammatory drugs; e.g., TEPOXALIN, TOLMETIN, SUPROFEN); immunosupressive/immunomodulatory agents (e.g., calcineurin inhibitors, such as cyclosporine, taerolimus; mTOR inhibitors (e.g., SIROLIMTJS, EVEROLIMUS); anti-proliferatives (e.g., azathioprine, mycophenolate mofetil);
corticosteroids (e.g., prednisolone, hydrocortisone); antibodies such as monoclonal anti-IL-2Ralpha receptor antibodies (e.g., basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g., anti-thymocyte globulin (ATG); anti-lymphocyte globulin (ALG); monoclonal anti-T cell antibody OKT3));
anti-thrombogenic agents (e.g., heparin, heparin derivatives, urokinase, PPack (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); and anti-oxidants (e.g., probucol, vitamin A, ascorbic acid, tocopherol, coenzyme Q-10, glutathione, L-cysteine, N-acetylcysteine).
Pharmaceutical compositions of the postpartum-derived cells, extracellular matrix produced thereby, cell lysates thereof, and PPDC-conditioned medium are included within the scope of the invention. The pharmaceutical compositions preferably include a pharmaceutically acceptable carrier or excipient. The pharmaceutical compositions are preferably for treating bone or cartilage conditions as defined herein. ' The invention further provides in some aspects methods of regenerating bone or cartilage tissue in a patient in need thereof by administering cells, matrices, or PPDC-products of the invention into a patient are provided.
Further provided by the invention are methods for treating a condition such as a bone or cartilage condition in a patient by administering one or more postpartum-derived cells, PPDC
populations, matrices, cell lysates, a combination of cell lysate and extracellular matrix, conditioned medium, or compositions of the invention. The PPDCs, whether differentiated or undifferentiated, or a combination thereof, extracellular matrix produced thereby, cell lysates thereof, a combination of cell lysate and extracellular matrix, matrices (e.g., scaffolds), conditioned medium, and compositions of the invention may be used in the treatment of bone or cartilage tissue conditions, for example, but not limited to congenital defects, bone fractures, meniscal injuues or defects, bone/spinal deformation, osteosarcoma, myeloma, bone dysplasia and scoliosis, osteoporosis, periodontal disease, dental bone loss, osteomalacia, rickets, fibrous osteitis, renal bone dystrophy, spinal fusion, spinal disc reconstruction or removal, Paget's disease of bone, meniscal injuries, rheumatoid arthritis, osteoarthritis, or a traumatic or surgical injury to cartilage or bone.
Also provided by the invention are kits comprising the postpartum-derived cells, PPDC-conditioned medium, lysate, and/or extracellular matrices of the invention.
The hits of the invention may further contain at least one component of a matrix, a second cell type, a hydrating agent, a ell culture substrate, a differentiation-inducing agent, cell culture media, and instructions, for example, for culture of the cells or administration of the cells and/or cell products.
In some embodiments, the invention provides methods for identifying compounds that modulate chondrogenesis or osteogenesis of a postpartum-derived cell comprising contacting a cell of the invention with said compound and monitoring the cell for a marker of chondrogenesis or osteogenesis. Also provided are methods for identifying compound toxic to a postpartum-derived cell of the invention by contacting said cell with said compound and monitoring survival of said cell.
Other features and advantages of the invention will be apparent from the detailed description and examples that follow.
DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
De rziti.oras Various terms used throughout the specification and claims we defined as set forth below.
Stern cells are undifferentiated cells defined by their ability at the single cell level to both self-renew and 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 irz vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple _g_ germ layers following transplantation and to contribute substantially to most, if not all, tissues following injection into blastocysts.
Stem cells are classified by their developmental potential as: (1) totipoterzt-able to give rise to all embryonic and extraembryonic cell types; (2) plur-ipotent - able to give rise to all embryonic cell types; (3) multipotent - 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); (4) oligopotent - able to give rise to a more restricted subset of cell lineages than multipotent stem cells; and (5) zzrzipotent- 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 sterrz cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself and, under normal circumstances, differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types. An embryonic 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. A postpar-turrz stem. 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 rnay 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).
Eraabryorzic 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. Extraenzbryonic tisszze is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryonic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta ( which itself forms from the chorion and the maternal decidua basalis).
Differentiation is the process by which an unspecialized ("uncommitted") or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example. A differentiated or differentiation-induced cell is one that has taken on a more specialized ("committed") position within the lineage of a cell. The term cornrnitted, 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. A lineage-specific marker refers to a characteristic specifically associated with the phenotype of cells of a lineage of interest and can be used to assess the differentiation of an uncommitted cell to the lineage of interest.
In a broad sense, a progerxitor 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 we 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 p~°ogenitor 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. This 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 ~tori-rertewing progenitor cell or as an irttenttediate progenitor oi°
precursor cell.
As used herein, the phrase differe~ttiates i.rtto a rnesodenrcal, ectodem~tal or ettdodemnal lineage refers to a cell that becomes committed to a specific mesodermal, ectodermal or endodeimal lineage, respectively. Examples of cells that differentiate into a mesodennal lineage or give rise to specific mesodermal cells include, but are not limited to, cells that are adipogenic, chondrogenic, cardiogenic, dermatogenic, hematopoetic, 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, and hepatogenic cells, cell that give rise to the lining of the intestine, and cells that give rise to pancreogenic and splanchogenic cells.
The cells of the invention are referred to herein as postpantcrra-derived cells (PPDCs).
Subsets of the cells of the present invention are referred to as pla.certta-derived cells (PDCs) or acrrabilical cord-derived cells (UDCs). PPDCs of the invention encompass undifferentiated and differentiation-induced cells. 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 lfz.
vitro (e.g., cultured in a growth medium to expand the population and/or to produce a cell line).
The ifz vitro manipulations of postpartum-derived cells and the unique features of the postpartum-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"). A pr-inzay~
cell culture is a culture of cells, tissues or organs taken directly from organisms and before the first subculture. Cells are expaizded 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 doubli~zg tizzze.
A cell Haze 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 may be referred to as a P10 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 linuted to the seeding density, substrate, medium, and time between passaging.
A coazditi,ozzed rrzedi.urzz is a medium in which a specific cell or population of cells has been cultured, and then removed. While the cells are cultured in the medium, they 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 troplzic factor is defined as a substance that promotes survival, growth, proliferation, maturation, differentiation, and/or maintenance of a cell, or stimulates increased activity of a cell.

When referring to cultured vertebrate cells, the term senescence (also replicative senescence or cellular senescence) refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's lir~zit). 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 irz vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide).
Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone. Thus, cells made quiescent by removing essential growth factors are able to resume growth and division when the growth factors are re-introduced, and thereafter carry out the same number of doublings as equivalent cells grown continuously. Similarly, when cells are frozen in liquid nitrogen after various numbers of population doublings and then thawed and cultured, they undergo substantially the same number of doublings as cells maintained unfrozen in culture. Senescent cells are not dead or dying cells;
they are actually resistant to programmed cell death (apoptosis), and have been maintained in their nondividing state for as long as three years. These cells are very much alive and metabolically active, but they do not divide. The nondividing state of senescent cells has not yet been found to be reversible by any biological, chemical, or viral agent.
As used herein, the term Growth craediunz refers to a culture medium sufficient for expansion of postpartum-derived cells. The culture medium of Growth medium preferably contains Dulbecco's Modified Essential Media (DMEM). More preferably, Growth medium contains glucose. Growth medium preferably contains DMEM-low glucose (DMEM-LG) (Invitrogen, Carlsbad, CA). Growth medium preferably contains about 15% (v/v) serum (e.g., fetal bovine semm, defined bovine serum). Growth medium preferably contains at least one antibiotic agent and/or antimycotic agent (e.g., penicillin, streptomycin, amphotericin B, gentamicin, nystatin; preferably, 50 units/milliliter penicillin G sodium and micrograms/milliliter streptomycin sulfate). Growth medium preferably contains ?-mercaptoethanol (Sigma, St. Louis MO). Most preferably, Growth medium contains DMEM-low glucose, serum, 2-mercaptoethanol, and an antibiotic agent.
As used herein, stafzdard growth coczditioczs refers to standard atmospheric conditions comprising about 5% CO~, a temperature of about 35-39°C, more preferably 37°C, and a relative humidity of about 100%.

The term isolated refers to a cell, cellular component, or a molecule that has been removed from its native environment.
The term about refers to an approximation of a stated value within a range of ~10°Io.
Borne corrditiorz (or irzjacry or disease) is an inclusive term encompassing acute and chronic and metabolic and non-metabolic conditions, disorders or diseases of bone. The term encompasses conditions caused by disease or trauma or failure of the tissue to develop normally.
Examples of bone conditions include but are not limited to congenital bone defects, bone fractures, meniscal injuries or defects, bone/spinal deformation, osteosarcoma, myeloma, bone dysplasia and scoliosis, osteoporosis, periodontal disease, dental bone loss, osteomalacia, rickets, fibrous osteitis, renal bone dystrophy, spinal fusion, spinal disc reconstruction or removal, and Paget's disease of bone.
Cartilage condition (or injury or disease) is an inclusive term encompassing acute and chronic conditions, disorders, or diseases of cartilage. The term encompasses conditions including but not limited to congenital defects, meniscal injuries, rheumatoid arthritis, osteoarthritis, or a traumatic or surgical injury to cartilage.
The term treating (or treatrzzerzt af) a bonze or cartilage condition refers to ameliorating the effects of, or delaying, halting or reversing the progress of, or delaying or preventing the onset of, a bone or cartilage condition as defined herein.
The term effective anzocrrzt refers to a concentration of a reagent or pharmaceutical composition, such as a growth factor, differentiation agent, trophic factor, cell population or other agent, that is effective for producing an intended result, including cell growth and/or differentiation irz vitro or in vivo, or treatment of a bone or cartilage condition as described herein. With respect to growth factors, an effective amount may range from about'1 nanogram/milliliter to about 1 microgram/milliliter. With respect to PPDCs as administergd to a patient irz vivo, an effective amount may range from as few as several hundred or fewer to as many as several million or more. In specific embodiments, an effective amount may range from 103-1011. It will be appreciated that the number of cells to be administered will vary depending on the specifics of the disorder to be treated, including but not limited to size or total volume/surface area to be treated, as well as proximity of the site of administration to the location of the region to be treated, among other factors familiar to the medicinal biologist.
The terms effective period (or time) and effective corzditiorzs refer to a period of time or other controllable conditions (e.g., temperature, humidity for in vitro methods), necessary or preferred for an agent or pharmaceutical composition to achieve its intended result.

The term pa.tiefzt or subject refers to animals, including mammals, preferably humans, who are treated with the pharmaceutical compositions or in accordance with the methods described herein.
The team nzatrix as used herein refers to a support for the PPDCs of the invention, for example, a scaffold (e.g., VICRYL, PCL/PGA, or RAD16) or supporting medium (e.g., hydrogel, extracellular membrane protein (e.g., MATRIGEL (BD Discovery Labware, Bedford, MA)).
The term plzarzzzaceutically acceptable carrier (or zzzedizzzcz), which may be used interchangeably with the term biologically cozzzpatible caz°zzer or znediunz, refers to reagents, cells, compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other complication commensurate with a reasonable benefit/risk ratio. As described in greater detail herein, pharmaceutically acceptable carriers suitable for use in the present invention include liquids, semi-solid (e.g., gels) and solid materials (e.g., cell scaffolds). As used herein, the term biodegradable describes the ability of a material to be broken down (e.g., degraded, eroded, dissolved) izz vivo. The term includes degradation izz vivo with or without elimination (e.g., by resorption) from the body. The senu-solid and solid materials may be designed to resist degradation within the body (zzon-biodegradable) or they may be designed to degrade within the body (biodegradable, bioerodable). A biodegradable material may further be baoz-esorbable oz-bioabsorbable, i.e., it may be dissolved and absorbed into bodily fluids (water-soluble implants are one example), or degraded and ultimately eliminated from the body, either by conversion into other mateuals or by breakdown and elimination through natural pathways.
Several terms are used herein with respect to cell replacement therapy. The terms azctologous transfer-, autologous trazzsplazztation, autograft and the like refer to treatments wherein the cell donor is also the recipient of the cell replacement therapy.
The terms allogezzei.c tra~zsfer; allogezzeic trazcsplaiztatiozz, allograft and the like refer to treatments wherein the cell donor is of the same species as the recipient of the cell replacement therapy, but is not the same individual. A cell transfer in which the donor's cells have been histocompatibly matched with a recipient is sometimes referred to as a syfzgezzeic trazzsfer. The terms xerzoge~zeic traczsfer;
xezzogefzeic tra~zsplazztatiozz, xezzograft and the like refer to treatments wherein the cell donor is of a different species than the recipient of the cell replacement therapy.
The following abbreviations are used herein:
ANG2 (or Azzg2) 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;
CKl8 for cytokeratin 18;
CXC ligan.d 3 for chemokine receptor ligand 3;
DAPI for 4' - 6-Diamidino-2-phenylindole-2HC1;
DMEM for Dulbecco's Minimal Essential Medium;
DMEM:Ig (or D~11EM:Lg, DMEM:LG) for DMEM with low glucose;
EDTA for ethylene diamine tetraace.tic acid;
EGF (or E) for epidermal growth factor;
EPO for erythropoietin;
FACS for fluorescent activated cell sorting;
FBS for fetal bovine seivm;
FGF (or F) for fibroblast growth factor;
GCP-~ for granulocyte chemotactic protein-2;
GDF-5 for growth and differentiation factor 5;
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;
h.MSC for Human mesenchymal stern cells;
HNF-Ialplaa for hepatoc5~te-specific transcription factor;
HUVEC for Human umbilical vein endothelial cells;
1309 for a chemokine and the ligand for the CCRB receptor and is responsible for chemoattraction of TH2 type T-cells;
IGF for insulin-like growth factor;
IL-6 for interleukin-6;
IL-8 for interleukin 8;
K19 for keratin 19;
K8 for keratin 8;
KGF for keratinocyte growth factor;
MCP-1 for monocyte chemotactic protein 1;

MDC for macrophage-derived chemokine;
MIPI alpha for macrophage inflammatory protein lalpha;
MIPI beta for macrophage inflammatory protein lbeta;
MMP for matrix metalloprotease (IvIl\~);
NISC for mesenchymal stem cells;
NHDF for Normal Human Dermal Fibroblasts;
NPE for Neural Progenitor Expansion media;
OxLDLR for oxidized low density lipoprotein receptor;
PBMC for peripheral blood mononuclear cell;
PBS for phosphate buffered saline;
PDC for placenta-derived cell;
PDGFbb for platelet derived growth factor;
PDGFr-alpha for platelet derived growth factor receptor alpha;
PD-L2 for programmed - death ligand 2;
PE for phycoerythrin;
PO for "per os" (by mouth);
PPDC for postpartum-derived cell;
Ra~ztes (or RANTES) for regulated on activation, normal T cell expressed and secreted;
rb for rabbit;
rh for recombinant human;
SC for subcutaneously;
SLID for severe combined immunodeficiency;
SDF-lalplza for stromal-derived factor lalpha;
SHH for sonic hedgehog;
SMA for smooth muscle actin;
SOP for standard operating procedure;
TARO for thymus and activation-regulated chemoltine;
TCP for tissue culture plastic;
TGFbeta2 for transforming growth factor beta?;
TGFbeta-3 for transforming growth factor beta-3;
TIMPl for tissue inhibitor of matrix metalloproteinase 1;
TPO for thrombopoietin;
TuJI for B 1II Tubulin;
UDC for umbilical cord-derived cell;

VEGF for vascular endothelial growth factor;
vWF for von Willebrand factor; and alphaFP for alpha-fetoprotein.
Descriptio~t, Various patents and other publications are cited herein and throughout the specification, each of which is incorporated by reference herein in its entirety.
In one aspect, the invention provides postpartum-derived cells (PPDCs) derived from postpartum tissue substantially free of blood. The PPDCs may be derived from placenta of a mammal including but not limited to human. The cells are capable of self-renewal and expansion in culture. The postpartum-derived cells have the potential to differentiate into cells of other phenotypes. The invention provides, in one of its several aspects cells that are derived from umbilical cord, as opposed to umbilical cord blood. The invention also provides, in one of its several aspects, cells that are derived from placental tissue.
The cells have been characterized as to several of their cellular, genetic, immunological, and biochemical properties. For example, the cells have been characterized by their growth by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
Derivation and expansion of postpartum-derived cells (PPDCs) According to the methods described herein, a mammalian placenta and umbilical cord are recovered upon or shortly after termination of either a full-term or pre-term pregnancy, for example, after expulsion after birth. Postpartum tissue can be obtained from any completed pregnancy, full-teL-~n or less than full-term, whether delivered vaginally, or through other means, for example, Cessarian section. The postpartum tissue may be transported from the birth site to a laboratory in a sterile container such as a flask, beaker, culture dish, or bag. The container may have a solution or medium, including but not limited to a salt solution, such as, for example, Dulbecco's Modified Eagle's Medium (DMEM) or phosphate buffered saline (PBS), or any solution used for transportation of organs used for transplantation, such as University of Wisconsin solution or perfluorochemical solution. One or more antibiotic andlor antimycotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin, may be added to the medium or buffer. The postpartum tissue may be rinsed with an anticoagulant solution such as heparin-containing solution. It is preferable to keep the tissue at about 4-10°C prior to extraction of PPDCs. It is even more preferable that the tissue not be frozen prior to extraction of PPDCs.
Isolation of PPDCs preferably occurs in an aseptic environment. Blood and debris are preferably removed from the postpartum tissue prior to isolation of PPDCs. For example, the postpartum tissue may be washed with buffer solution, such as but not limited to phosphate buffered saline. The wash buffer also may comprise one or more antimycotic and/or antibiotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin.
In some aspects of the invention, the different cell types present in postpartum tissue are fractionated into subpopulations from which the PPDCs can be isolated. This may be accomplished using techniques for cell separation including, but not limited to, enzymatic treatment to dissociate postpartum tissue into its component cells, followed by cloning and selection of specific cell types, for example but not limited to selection based on morphological and/or biochemical markers; selective growth of desired cells (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; differential adherence properties of the cells in the mixed population; filtration;
conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation);
unit gravity separation; countercurrent distribution; electrophoresis; and flow cytometry, for example, fluorescence activated cell sorting (FACS).
In a preferred embodiment, postpartum tissue comprising a whole placenta or a fragment or section thereof is disaggregated by mechanical force (mincing or shear forces), enzymatic digestion with single or combinatorial proteolytic enzymes, such as a matrix metalloprotease and/or neutral protease, for example, collagenase, trypsin, dispase, LIBERASE
(Boehringer Mannheim Corp., Indianapolis, IN), hyaluronidase, and/or pepsin, or a combination of mechanical and enzymatic methods. For example, the cellular component of the postpartum tissue may be disaggregated by methods using collagenase-mediated dissociation. Enzymatic digestion methods preferably employ a combination of enzymes, such as a combination of a matrix metalloprotease and a neutral protease. The matrix metalloprotease is preferably a collagenase. The neutral protease is preferably thermolysin or dispase, and most preferably is dispase. More preferably, enzymatic digestion of postpartum tissue uses a combination of a matrix metalloprotease, a neutral protease, and a mucolytic enzyme for digestion of hyaluronic acid, such as a combination of collagenase, dispase, and hyaluronidase or a combination of LIBERASE (Boehringer Mannheim Coip., Indianapolis, IN) and hyaluronidase.
Collagenase may be type 1, 2, 3, or 4. Other enzymes known in the art for cell isolation include papain, deoxyribonucleases, serine proteases, such as trypsin, chymotrypsin, or elastase, that may be used either on their own or in combination with other enzymes such as matrix metalloproteases, mucolytic enzymes, and neutral proteases. Serine proteases are preferably used consecutively following use of other enzymes. The temperature and period of time tissues or cells are in contact with serine proteases is particularly important. Serine proteases may be inhibited by alpha 2 microglobulin in serum and therefore the medium used for digestion is usually serum-free. EDTA and DNAse are commonly used in enzyme digestion procedures to increase the efficiency of cell recovery. The degree of dilution of the digestion may also greatly affect the cell yield as cells may be trapped within the viscous digest. The LIBERASE
(Boehringer Mannheim Corp., Indianapolis, IN) Blendzyme (Roche) series of enzyme combinations are very useful and may be used in the instant methods. Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources. The skilled artisan is also well-equipped to assess new, or additional enzymes or enzyme combinations for their utility in isolating the cells of the invention. Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer. In more preferred embodiments, the tissue is incubated at 37°G during the enzyme treatment of the disintegration step.
Postpa~.-tum tissue comprising the umbilical cord and placenta may be used without separation. Alternatively, the umbilical cord may be separated from the placenta by any means known in the art. In some embodiments of the invention, postpartum tissue is separated into two or more sections, such as umbilical cord and placenta. In some embodiments of the invention, placental tissue is separated into two or more sections, each section consisting predominantly of either neonatal, neonatal and maternal, or maternal aspect. The separated sections then are dissociated by mechanical and/or enzymatic dissociation according to the methods described herein. Cells of neonatal or maternal lineage may be identified by any means known in the art, for example, by karyotype analysis or in sitar hybridization for the Y-chromosome. Karyotype analysis also may be used to identify cells of normal karyotype.
Isolated cells or postpartum tissue from which PPDCs grow out may be used to initiate, or seed, cell cultures. Cells are transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen, gelatin, fibronectin, ornithine, vitronectin, and extracellular membrane protein (e.g., MATRIGEL (BD
Discovery Labware, Bedford, MA)). PPDCs are cultured in any culture medium capable of sustaining growth of the cells such as, but not limited to, DMEM (high or low glucose), Eagle's basal medium, Ham's F10 medium (F10), Ham's F-12 medium (F12), Iscove's modified Dulbecco's medium, Mesenchymal Stem Cell Growth Medium (MSCGM), DMEM/F12, RPMI 1640, advanced DMEM (Gibco), DMEM/MCDB201 (Sigma), and CELL-GRO FREE. The culture medium may be supplemented with one or more components including, for example, serum (e.g., fetal bovine serum (FBS), preferably about 2-15% (v/v); equine serum (ES);
human serum(HS));
beta-mercaptoethanol (BME), preferably about 0.001% (v/v); one or more growth factors, for example, platelet-derived growth factor (PDGF), insulin-like growth factor-1 (IGF-1), leukemia inhibitory factor (LIF), .epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), and erythropoietin (EPO); amino acids, including L-valine; and one or more antibiotic and/or antimycotic agents to control microbial contanunation, such as, for example, penicillin G, streptomycin sulfate, amphotericin B, gentanucin, and nystatin, either alone or in combination. The culture medium preferably comprises Growth medium (DMEM-low glucose, serum, BME, an antimycotic agent, and an antibiotic agent).
The cells are seeded in culture vessels at a density to allow cell growth. In a preferred embodiment, the cells are cultured at about 0 to about 5 percent by volume COZ
in air. In some preferred embodiments, the cells are cultured at about 2 to about 25 percent 02 in air, preferably about 5 to about 20 percent OZ in air. The cells preferably are cultured at about 25 to about 40°C, more preferably about 35°C to about 39°C ,and more preferably are cultured at 37°C. The cells are preferably cultured in an incubator. The medium in the culture vessel can be static or agitated, for example, using a bioreactor. PPDCs preferably are grown under low oxidative stress (e.g., with addition of glutathione, ascorbic acid, catalase, tocopherol, N-acetylcysteine). "Low oxidative stress", as used herein, refers to conditions of no or minimal free radical damage to the cultured cells.
Methods for the selection of the most appropriate culture medium, medium preparation, and cell culture techniques are well known in the art and are described in a vv.~iety of sources, including Doyle et al., (eds.), 1995, CELL & TISSUE CULTURE: LABORATORY
PROCEDURES, John Wiley & Sons, Chichester; and Ho and Wang (eds.), 1991, ANTNIAL CELL
BIOREACTORS, Butterworth-Heinemann, Boston, which are incorporated herein by reference.
The culture medium is changed as necessary, for example, by carefully aspirating the medium from the dish, for example, with a pipette, and replenishing with fresh medium.
Incubation is continued until a sufficient number or density of cells accumulate in the dish. The original explanted tissue sections may be removed and the remaining cells trypsinized using standard techniques or using a cell scraper. After trypsinization, the cells are collected, removed to fresh medium and incubated as above. In some embodiments, the medium is changed at least once at approximately 24 hours post-trypsinization to remove any floating cells. The cells remaining in culture are considered to be PPDCs.
After culturing the cells or tissue fragments for a sufficient period of time, PPDCs will have grown out, either as a result of migration from the postpartum tissue or cell division, or both. In some embodiments of the invention, PPDCs are passaged, or removed to a separate culture vessel containing fresh medium of the same or a different type as that used initially, where the population of cells can be mitotically expanded. PPDCs are preferably passaged up to about 100% confluence, more preferably about 70 to about S5% confluence. The lower limit of confluence for passage is understood by one skilled in the art. The placenta-derived cells of the invention may be utilized from the first subculture (passage 0) to senescence.
The preferable number of passages is that which yields a cell number sufficient for a given application. In certain embodiments, the cells are passaged 2 to 25 times, preferably 4 to 30 times, more preferably S to 15 times, more preferably 10 or 11 times, and most preferably 11 times. Cloning and/or subcloning may be performed to confirm that a clonal population of cells has been isolated.
Cells of the invention may be cryopreserved and/or stored prior to use.
Characterization of PPDCs PPDCs may be characterized, for example, by growth characteristics (e.g., population doubling capability, doubling time, passages to senescence), karyotype analysis (e.g., normal karyotype; maternal or neonatal lineage), flow cytometry (e.g., FACS
analysis), immunohistochemistry and/or immunocytochemistry (e.g., for detection of epitopes including but not limited to vimentin, desmin, alpha-smooth muscle actin, cytokeratin 1S, von Willebrand factor, CD34, GROalpha, GCP-2, oxidized low density lipoprotein receptor 1, and NOGO-A), gene expression profiling (e.g., gene chip arrays; polymerase chain reaction (for example, reverse transcriptase PCR, real time PCR, and conventional PCR)), protein arrays, protein secretion (e.g., by plasma clotting assay or analysis of PPDC-conditioned medium, for example, by Enzyme Linked ImmunoSorbent Assay (ELISA)), antibody analysis (e.g., ELISA;
antibody staining for cell surface markers including but not limited to CD10, CD13, CD31, CD34, CD44, CD45, CD73, CDSO, CDS6, CD90, CD117, CD141, CD17S, platelet-derived growth factor receptor alpha (PDGFr-alpha), HLA class I antigens (HLA-A, HLA-B, HLA-C), HLA
class II
antigens (HLA-DP, HLA-DQ, HLA-DR), B7-H2, and PD-LZ), mixed lymphocyte reaction (e.g., as measure of stimulation of allogeneic PBMCs), and/or other methods known in the art.

PPDCs can undergo at least 40 population doublings in culture. Population doubling may be calculated as [1n (cell final/cell initial)/ln 2]. Doubling time may be calculated as (time in culture (h)/population doubling).
Undifferentiated PPDCs preferably produce of at least one of NOGO-A, GCP-3, tissue factor, vimentin, and alpha-smooth muscle actin; more preferred are cells which produce each of GCP-2, tissue factor, vimentin, and alpha-smooth muscle actin. In some embodiments, two, three, four, or five of these factors are produced by the PPDCs.
In some embodiments, PPDCs lack production of at least one of NOGO-A, GRO-alpha, or oxidized low density lipoprotein receptor, as detected by flow cytometry.
In some embodiments, PPDCs lack production of at least two or three of these factors.
PPDCs may comprise at least one cell surface marker of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C. PPDCs preferably produce each of these surface markers. PPDCs may be characterized in their 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. PPDCs preferably lack production of each of these surface markers.
In some embodiments, PPDCs exhibit expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3; or at least one of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor 1, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113. Preferred PPDCs express, relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, increased levels of interleukin 8; reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2);
chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3; or increased levels of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor 1, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113. In PPDCs wherein expression, relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of interleukin S; 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, increased relative levels of at least one of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor 1, protein kinase C zeta, clone IMAGE:41?9671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113 are preferably not present. In PPDCs wherein expression, relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor 1, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113, increased relative levels 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 27; chemokine (C-X-C
motif) ligand 3;
and tumor necrosis factor, alpha-induced protein 3 are preferably not present.
PPDCs may 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 shock 27kDa protein 2; chemokine (C-X-C motif) ligand 12 (stromal cell-derived factor 1); elastin; cDNA DKFZp586M202'2 (from clone DKFZp586M2022);
mesenchyme homeobox 2; sine oculis homeobox homolog 1; crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1;
tetranectin; 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; hypothetical gene BC008967; collagen, type VIII, alpha 1;
tenascin C;
Iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2;
cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor l;
potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, alpha 7; DKFZP586L151 protein; transeriptional co-activator with PDZ-binding motif (TAZ);
sine oculis homeobox homolog 2; KIAA1034 protein; early growth response 3;
distal-less homeobox 5; hypothetical protein FLJ20373; aldo-keto reductase family l, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; fibronectin 1;
proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); cDNA clone EUROIMAGE 1968422; EphA3;
KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C
(atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5; EGF-containing fibulin-like extracellular matrix protein 1; BCL2/adenovirus E1B l9kDa interacting protein 3-like; AE
binding protein 1; cytochrome c oxidase subunit VBa 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 a gene array, for example on a Affymetrix GENECHIP.
PPDCs may secrete a variety of biochemically active factors, such as growth factors, chemokines, 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, MIPIa, RANTES, and TJMP1. PPDCs may be characterized in their lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIPIb, 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 postpartum-derived 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 GCP-2, NOGO-A, tissue factor, vimentin, and alpha-smooth muscle actin. More preferred are those cells producing three, four, or five of these proteins.
The skilled artisan will appreciate that cell markers are subject to vary somewhat under vastly different growth conditions, and that generally herein described are characterizations in Growth Medium, or variations thereof. Postpartum-derived cells that produce of at least one, two, three, or four of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C
are preferred. More preferred are those cells producing five, six, or seven of these cell surface markers. Still more preferred are postpartum-derived cells that can produce eight, nine, or ten of the foregoing cell surface marker proteins.
PPDCs that lack of production of at least one, two, three, or 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 preferred. PPDCs lacking production of at least five, six, seven, or eight or more of these markers are 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 eleven, twelve, or thirteen 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.
It is preferred that postpartum-derived cells exhibit expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of at least one, two, or three of interleukin 8; reticulon 1;
chemokine (C-X-C
motif) ligand 1 (melanoma growth stimulating activity, alpha); chemokine (C-X-C motif) ligand 6 (granulocyte ehemotactic protein 2); chemokine (C-X-C motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3; or at least one, two, or three of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor 1, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113. More preferred are those cells which exhibit elevated relative expression of four or five, and still more prefewed are cell capable of increased relative expression of six, seven, or eight of the foregoing genes of the respective gene sets. Most preferably, the cells exhibit expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for a combination of interleukin 8; reticulon l; chemokine (C-X-C
motif) ligand 1 melanoma rowth stimulating activit , al ha ; chemokine C-X-C moti 1i and 6 ranuloc a ( g b Y p ) ( ~ g (g Yt chernotaetic protein 2); chemokine (C-X-C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3 or a combination of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor l, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113.
For some embodiments, preferred are 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; cDNA
DKFZp586M2022 (from clone DKFZp5S6M2022); mesenchyme homeobox 2; sine oculis homeobox homolog 1; crystallin, alpha B; dishevelled associated activator of morphogenesis 2;
DKFZP586B2420 protein; similar to neuralin 1; tetranectin; src homology three (SH3) and cysteine rich domain; B-cell translocation gene 1, anti-proliferative;
cholesterol 25-hydroxylase;
runt-related transcription factor 3; hypothetical protein FLJ23191;
interleulcin 11 receptor, alpha;

procollagen C-endopeptidase enhancer; frizzled homolog 7; hypothetical gene BC008967;
collagen, type VIII, alpha 1; tenascin C; Iroquois homeobox protein 5;
hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; 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; 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); cDNA
clone EUROIMAGE
1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C
(atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; cDNA
DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5; EGF-containing fibulin-like extracellular matrix protein 1; BCL2ladenovirus 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. 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, 15 or 20 genes corresponding to those listed above. Presently more preferred are cell with reduced expression of at least 25, 30, or 35 of the genes corresponding to the listed sequences. Also more preferred are those postpartum-derived cells having expression that is reduced, relative to that of a human fibroblast, a mesenchymal stem cell, or an 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, M~la, RANTES, and T1ZVVIP1. Cells which secrete five, six, seven or eight of the listed proteins are also 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, PPDCs 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, MIPlb, 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.
Examples of placenta-derived cells of the invention were deposited with the American Type Culture Collection (ATCC, Manassas, VA) and assigned ATCC Accession Numbers as follows: (1) strain designation PLA 071003 (PS) 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.
Examples of umbilical cord-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 LTMB 022803 (P17) was assigned Accession No. PTA-6068.
PPDCs can be isolated. The invention also provides compositions of PPDCs, including populations of PPDCs. In some embodiments, the cell population is heterogeneous. A
heterogeneous cell population of the invention may comprise at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% PPDCs of the invention. In some embodiments, the heterogeneous cell populations of the invention may further comprise bone marrow cells, stem cells, chondroblasts, chondrocytes, andlor progenitor cells. In some embodiments, the heterogeneous cell populations of the invention may further comprise bone marrow cells, stem cells, osteoblasts, osteocytes, osteoclasts, bone lining cells, and/or progenitor cells. In some embodiments, the. population is substantially homogeneous, i.e., comprises substantially only PPDCs (preferably at least about 96%, 97%, 98%, 99% or more PPDCs). The homogeneous cell population of the invention may comprise umbilical cord- or placenta-derived cells.
Homogeneous populations of umbilical cord-derived cells may be free of cells of maternal lineage. Homogeneous populations of placenta-derived cells may be of neonatal or maternal lineage. Homogeneity of a cell population may be achieved by any method known in the art, for example, by cell sorting (e.g., flow cytometry), bead separation, or by clonal expansion.
Methods of the invention further include methods for producing a population of postpartum-derived cells by expanding a cell of the invention in culture. The postpartum-derived cells of the invention preferably expand in the presence of from about 5% to about 20% oxygen.
The postpartum-derived cells of the invention preferably are expanded in culture medium such as but not limited to Dulbecco's modified Eagle's medium (DMEM), mesenchymal stem cell growth medium, advanced DMEM (Gibco), DMEM/MCDB201 (Sigma), RPMI1640, CELL-_27_ GRO FREE, advanced DMEM (Gibco), DMEM/MCDB201 (Sigma), Ham's F10 medium, Ham's F12 medium; DMEM/F12, Iscove's modified Dulbecco's medium, or Eagle's basal medium. The culture medium preferably contains low or high glucose, about 2%-15% (v/v) serum, betamercaptoethanol, and an antibiotic agent. The culture medium may contain at least one of fibroblast growth factor, platelet-derived growth factor, vascular endothelial growth factor, and epidermal growth factor. The cells of the invention may be grown on an uncoated or coated surface. Surfaces for growth of the cells may be coated for example with gelatin, collagen (e.g., native or denatured), fibronectin, laminin, ornithine, vitronectin, or extracellular membrane protein (e.g., MATRIGEL). In some embodiments, a population of postpartum-derived cells is mixed with another population of cells.
Culture of PPDCs in a Chondrogenic Medium PPDCs may be induced to differentiate into a chondrogenic lineage by subjecting them to differentiation-inducing cell culture conditions. PPDCs may be cultured in a chondrogenic medium comprising specific exogenous chondrogenic growth factors (e.g., in culture), such as, for example, one or more of GDF-5 or transforming growth factor beta3 (TGF-beta3), with or without ascorbate.
Preferred chondrogenic medium is supplemented with an antibiotic agent, amino acids including proline and glutamine, sodium pyruvate, dexamethasone, ascorbic acid, and insulin/tranferrin/selenium. Chondrogenic medium is preferably supplemented with sodium hydroxide and/or collagen. Most preferably, chondrogenic culture medium is supplemented with collagen. The cells may be cultured at high or low density. Cells are preferably cultured in the absence of serum.
Culture of PPDCs in an Osteogenic Medium PPDCs may be induced to differentiate into an osteogenic lineage by subjecting them to differentiation-inducing cell culture conditions. In some embodiments, PPDCs are cultured in osteogenic medium such as, but not limited to, media (e.g., DMEM-low glucose) containing about 10-7 molar and about 10-~ molar dexamethasone in combination with about 10 micromolw to about 50 micromolar ascorbate phosphate salt (e.g., ascorbate-2-phosphate) and between about nanomolar and about 10 millimolar beta-glycerophosphate. The medium preferably includes serum (e.g., bovine serum, horse serum). Osteogenic medium also may comprise one or more antibiotic/antimycotic agents. The osteogenic medium is preferably supplemented with transforming growth factor-beta (e.g., TGF-betal) and/or bone rnorphogenic protein (e.g., BMP-2, BMP-4, or a combination thereof; most preferably BMP-4) Assessment of Differentiation PPDCs may be induced to differentiate to an ectodermal, endodermal, or mesodermal lineage. Methods to characterize differentiated cells that develop from the PPDCs of the invention, include, but are not limited to, histological, morphological, biochemical and immunohistochemical methods, or using cell surface markers, or genetically or molecularly, or by identifying factors secreted by the differentiated cell, and by the inductive qualities of the differentiated PPDCs.
Chondrogenic differentiation may be assessed, for example, by Safranin-O
staining for glycosaminoglycan expression by the cells or hematoxylin/eosin staining or by detection of a chondrogenic lienage marker (e.g., sulfated glycosaminoglycans and proteoglycans, keratin, chondroitin, Type II collagen) in the culture or more preferably in the cells themselves.
PPDCs may be analyzed for an osteogenic phenotype by any method known in the art, e.g., von Kossa staining or by detection of osteogenic markers such as osteocalcin, bone sialoprotein, alkaline phosphatase, osteonectin, osteopontin, type I collagen, bone morphogenic proteins, and/or core binding factor al in the culture or more preferably in the cells themselves..
Methods of using PPDCs or components or products thereof Genetic Engineering of PPDCs The cells of the invention can be engineered using any of a variety of vectors including, but not limited to, integrating viral vectors, e.g., retrovirus vector or adeno-associated viral vectors; non-integrating replicating vectors, e.g., papilloma virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors. Other methods of introducing DNA
into cells include the use of liposomes, electroporation, a particle gun, or by direct DNA
injection.
Hosts cells are preferably transformed or transfected with DNA controlled by or in operative association with, one or more appropriate expression control elements such as promoter or enhancer sequences, transcription terminators, polyadenylation sites, among others, and a selectable marker.
Following the introduction of the foreign DNA, engineered cells may be allowed to grow in enriched media and then switched to selective media. The selectable marker in the foreign DNA confers resistance to the selection and allows cells to stably integrate the foreign DNA as, for example, on a plasmid, into their chromosomes and grow to form foci which, in turn, can be cloned and expanded into cell lines.

This method can be advantageously used to engineer cell lines which express the gene product.
Any promoter may be used to drive the expression of the inserted gene. For example, viral promoters include, but are not limited to, the CMV promoter/enhancer, SV
40, papillomavirus, Epstein-Barr virus or elastin gene promoter. Preferably, the control elements used to control expression of the gene of interest should allow for the regulated expression of the gene so that the product is synthesized only when needed I72 viv0. If transient expression is desired, constitutive promoters are preferably used in a non-integrating and/or replication-defective vector. Alternatively, inducible promoters could be used to drive the expression of the inserted gene when necessary.
Inducible promoters include, but are not limited to, those associated with metallothionein and heat shock proteins.
The cells of the invention may be genetically engineered to "knock out" or "knock down"
expression of factors that promote inflammation or rejection at the implant site. Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are discussed below. "Negative modulation," as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level and/or activity of the target gene product in the absence of the modulatory treatment. The expression of a gene native to a chondrocyte or osteocyte can be reduced or knocked out using a number of techniques including, for example, inhibition of expression by inactivating the gene completely (commonly termed "knockout") using the homologous recombination technique. Usually, an exon encoding an important region of the protein (or an exon 5' to that region) is interrupted by a positive selectable marker, e.g., neo, preventing the production of normal mRNA from the target gene and resulting in inactivation of the gene. A gene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the sequences intervening the two regions can be deleted (Mombaerts et al., 1991, Pf~oc. Nat. Aca~l. Sci.. LLS.A. 88:3084).
Antisense, small interfering RNA, DNAzymes and ribozyme molecules which inhibit expression of the twget gene can also be used in accordance with the invention to reduce the level of target gene activity. For example, antisense RNA molecules which inhibit the expression of major histocornpatibility gene complexes (HLA) have been shown to be most versatile with respect to immune responses. Still further, triple helix molecules can be utilized in reducing the level of target gene activity.

These techniques are described in detail by L.G. Davis et al. (eds), 1994, BASIC
METHODS IN MoLECUL,AR BIOLOGY, 2nd ed., Appleton & Lange, Norwalk, Conn., which is incorporated herein by reference.
IL-1 is a potent stimulator of cartilage resorption and of the production of inflammatory mediators by chondrocytes (Campbell et al.., 1991, J. Inz~zzurz. 147: 1238).
Using any of the foregoing techniques, the expression of IL-1 can be knocked out or knocked down in the cells of the invention to reduce the risk of resorption of implanted cartilage or the production of inflammatory mediators by the cells of the invention. Likewise, the expression of MHC class II
molecules can be knocked out or knocked down in order to reduce the risk of rejection of the implanted tissue.
Once the cells of the invention have been genetically engineered, they may be directly implanted into the patient to allow for the treatment of a bone or cartilage condition, for example, rheumatoid or joint disease, or to produce an anti-inflammatory gene product such as, far example, peptides or polypeptides corresponding to the idiotype of neutralizing antibodies for GM-CSF, TNF, IL-1, IL-2, or other inflammatory cytokines.
Alternatively, the genetically engineered cells may be used to produce new tissue i~a vitro, which is then implanted in the subject, as described herein.
Secretiofz of Trophic factors The secretion of growth factors by PPDCs may provide trophic support for a second cell type ifz vitro or i.rz vivo. PPDCs may secrete, for example, at least one of monocyte chemotactic protein 1 (MCP-1), interleukin-6 (IL6), interleukin 8 (IL-S), GCP ~, hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), fibroblast growth factor (FGF), heparin binding epidermal growth factor (HB-EGF), brain-derived neurotrophic factor (BDNF), thrombopoietin (TPO), macrophage inflammatory protein 1 alpha (MIPla), RANTES, and tissue inhibitor of matrix metalloproteinase 1 (TIIVVIPl), which can be augmented by a variety of techniques, including ex vi.vo cultivation of the cells in chemically defined medium.
In some aspects of the invention, a population of PPDCs supports the survival, proliferation, growth, maintenance, maturation, differentiation, or increased activity of cells including stem cells, such as embryonic stem cells, bone marrow cells, chondrocytes, chondroblasts, and mixtures thereof. In some aspects of the invention, a population of PPDCs supports cells including stem cells, such as embryonic stem cells, bone maiTOw cells, osteoblasts, osteocytes, osteoclasts, bone lining cells, and mixtures thereof. In some aspects of the invention, a population of PPDCs supports cells including stem cells, such as embryonic stem cells, bone marrow cells, chondrocytes, chondroblasts, osteoblasts, osteocytes, osteoclasts, bone lining cells, and mixtures thereof. In other embodiments, the population is substantially homogeneous, i.e., comprises substantially only PPDCs (preferably at least about 96%, 97%, 98%, 99% or more PPDCs)..
PPDCs have the ability to support survival, growth, and differentiation of other cell types in co-culture. In some embodiments, PPDCs are co-cultured izz vitro to provide trophic support to other cells, including but not limited to stem cells, osteocytes, osteoblasts, osteoclasts, bone lining cells, chondrocytes, chondroblasts, and/or bone marrow cells, or combinations thereof.
For co-culture, it may be desirable for the PPDCs and the desired other cells to be co-cultured under conditions in which the two cell types are in contact. This can be achieved, for example, by seeding the cells as a heterogeneous population of cells in culture medium or onto a suitable culture substrate. Alternatively, the PPDCs can first be grown to confluence and employed as a substrate for the second desired cell type in culture. In this latter embodiment, the cells may further be physically separated, e.g., by a membrane or similar device, such that the other cell type may be removed and used separately following the co-culture period. In other embodiments, the desired other cells are cultured in contact with the conditioned medium, extracellular matrix, and/or cell lysate of the PPDCs. Use of PPDCs in co-culture to promote expansion and differentiation of other cell types may find applicability in research and in clinical/therapeutic areas. For instance, PPDC co-culture may be utilized to facilitate growth and differentiation of cells of a given phenotype in culture, for example, chondrocytes or osteocytes, for basic research purposes or for use in drug screening assays, for example.
PPDC co-culture may also be utilized for ex viva expansion of cells of an osteogenic or chondrogenic phenotype for later administration for therapeutic purposes. For example, cells may be harvested from an individual, expanded ex vi.vo in co-culture with PPDCs, then returned to that individual (autologous transfer) or another individual (syngeneic or allogeneic transfer). In these embodiments, it will be appreciated that, following ex viva expansion, the mixed population of cells comprising the PPDCs could be administered to a patient in need of treatment, for example, of a bone or cartilage condition as described herein. Alternatively, in situations where autologous transfer is appropriate or desirable, the co-cultured cell populations may be physically separated in culture, enabling removal of the autologous cells for administration to the patient.
Cozzditioned znedizanz of PPDCs Another embodiment of the invention features use of PPDCs for production of conditioned medium, either from undifferentiated PPDCs or from PPDCs incubated under conditions that stimulate differentiation into a chondrogenic or osteogenic lineage. Such conditioned media are contemplated for use in in vitro or ex vivo culture of cells, for example, stem or progenitor cells, including but not limited to bone marrow cells, osteoblasts, osteocytes, osteoclasts, bone lining cells, chondroblasts, and chondrocytes, or ifa vi.vo to support transplanted cells comprising homogeneous or heterogeneous populations of PPDCs and/or stem cells, osteocytes, osteoblasts, osteoclasts, bone lining cells, chondrocytes, chondroblasts, and bone marrow cells, for example.
Therapeutic applications of PPDCs PPDCs of the invention may be used to treat patients requiring the repair or replacement of cartilage or bone tissue resulting from disease or trauma or failure of the tissue to develop normally, or to provide a cosmetic function, such as to augment facial or other features of the body. Treatment may entail the use of the cells of the invention to produce new cartilage tissue or bone tissue. For example, the undifferentiated or chondrogenic differentiation-induced cells of the invention may be used to treat a cartilage condition, for example, rheumatoid arthritis or osteoa~-thritis or a traumatic or surgical injury to cartilage. As another example, the undifferentiated or osteogenic differentiation-induced cells of the invention may be used to treat bone conditions, including metabolic and non-metabolic bone diseases. Examples of bone conditions include meniscal tears, spinal fusion, spinal disc removal, spinal reconstruction, bone fractures, bone/spinal deformation, osteosarcoma, myeloma, bone dysplasia, scoliosis, osteoporosis, periodontal disease, dental bone loss, osteomalacia, rickets, fibrous osteitis, renal bone dystrophy, and Paget's disease of bone.
The cells of the invention may be administered alone or as admixtures with other cells.
Cells that may be administered in conjunction with PPDCs include, but are not limited to, other multipotent or pluripotent cells or chondrocytes, chondroblasts, osteocytes, osteoblasts, osteoclasts, bone lining cells, stem cells, or bone marrow cells. The cells of different types may be admixed with the PPDCs immediately or shortly prior to administration, or they may be co-cultured together for a period of time prior to administration.
The PPDCs may be administered with other beneficial drugs or biological molecules (growth factors, trophic factors). When PPDCs are administered with other agents, they may be administered together in a single pharmaceutical composition, or in separate pharmaceutical _33_ compositions, simultaneously or sequentially with the other agents (either before or after administration of the other agents). Bioaetive factors which may be co-administered include anti-apoptotic agents (e.g., EPO, EPO mimetibody, TPO, IGF-I and IGF-II, HGF, caspase inhibitors);
anti-inflammatory agents (e.g., p38 MAPK inhibitors, TGF-beta inhibitors, statins, IL-6 and 1L-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and NSAIDs (non-steroidal anti-inflammatory drugs; e.g., TEPOXALIN, TOLMETIN, SUPROFEN);
immunosupressive/immunomodulatory agents (e.g., calcineurin inhibitors, such as cyclosporine, tacrolimus; mTOR inhibitors (e.g., SIROLIMUS, EVEROLIMUS); anti-proliferatives (e.g., azathioprine, mycophenolate mofetil); corticosteroids (e.g., prednisolone, hydrocortisone);
antibodies such as monoclonal anti-IL-2Ralpha receptor antibodies (e.g., basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g., anti-thymocyte globulin (ATG); anti-lymphocyte globulin (ALG); monoclonal anti-T cell antibody OKT3)); anti-thrombogenic agents (e.g., heparin, heparin derivatives, urokinase, PPack (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); and anti-oxidants (e.g., probucol, vitamin A, ascorbic acid, tocopherol, coenzyme Q-10, glutathione, L-cysteine, N-acetylcysteine) as well as local anesthetics. As another example, the cells may be co-administered with scar inhibitory factor as described in U.S. Patent No. 5,827,735, incorporated herein by reference.
In one embodiment, PPDCs are administered as undifferentiated cells, i.e., as cultured in Growth Medium. Alternatively, PPDCs may be administered following exposure in culture to conditions that stimulate differentiation toward a desired phenotype, for example, a chondrogenic or osteogenic phenotype.
The cells of the invention may be surgically implanted, injected, delivered (e.g., by way of a catheter or syringe), or otherwise administered directly or indirectly to the site in need of repair or augmentation. The cells may be administered by way of a matrix (e.g., a three-dimensional scaffold). The cells may be administered with conventional pharmaceutically acceptable carriers. Routes of administration of the cells of the invention or compositions or components (e.g., ECM, cell lysate, conditioned medium) thereof include intramuscular, ophthalmic, parenteral (including intravenous), intraarterial, subcutaneous, oral, and nasal administration. Particular routes of parenteral administration include, but are not limited to, intramuscular, subcutaneous, intraperitoneal, intracerebral, intraventricular, intracerebroventricular, intrathecal, intracisternal, intraspinal andlor peri-spinal routes of administration.

When cells are administered in semi-solid or solid devices, surgical implantation into a precise location in the body is typically a suitable means of administration.
Liquid or fluid pharmaceutical compositions, however, may be administered to a more general location (e.g., throughout a diffusely affected area, for example), from which they migrate to a particular location, e.g., by responding to chemical signals.
Other embodiments encompass methods of treatment by administering pharmaceutical compositions comprising PPDC cellular components (e.g.,'cell lysates or components thereof) or products (e.g., extracellular matrix, trophic and other biological factors produced naturally by PPDCs or through genetic modification, conditioned medium from PPDC culture).
Again, these methods may further comprise administering other active agents, such as anti-apoptotic agents (e.g., EPO, EPO mimetibody, TPO, IGF-I and IGF-II, HGF, caspase inhibitors);
anti-inflammatory agents (e.g., p38 MAPK inhibitors, TGF-beta inhibitors; statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and NSA>Ds (non-steroidal anti-inflammatory drugs; e.g., TEPOXALIN, TOLMETIN, SUPROFEN);
immunosupressive/immunomodulatory agents (e.g., calcineurin inhibitors, such as cyclosporine, tacrolimus; mTOR inhibitors (e.g., SLROLIMLTS, EVEROLIMUS); anti-proliferatives (e.g., azathioprine, .mycophenolate mofetil); corticosteroids (e.g., prednisolone, hydrocortisone);
antibodies such as monoclonal anti-IL,-2Ralpha receptor antibodies (e.g., basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g., anti-thymocyte globulin (ATG); anti-lymphocyte globulin (ALG); monoclonal anti-T cell antibody OKT3)); anti-thrombogenic agents (e.g., heparin, heparin derivatives, urokinase, PPack (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); and anti-oxidants (e.g., probucol, vitamin A, ascorbic acid, tocopherol, coenzyme Q-10, glutathione, L-cysteine, N-acetylcysteine), local anesthetics, and scar inhibitory factor as described in U.S. Patent No.
5,827,735, incorporated herein by reference.
Dosage forms and regimes for administering PPDCs or any of the other pharmaceutical compositions described herein are developed in accordance with good medical practice, taking into account the condition of the individual patient, e.g., nature and extent of the condition being treated, age, sex, body weight and general medical condition, and other factors known to medical practitioners. Thus, the effective amount of a pharmaceutical composition to be administered to a patient is determined by these considerations as known in the art.

In some embodiments of the invention, it may not be necessary or desirable to immunosuppress a patient prior to initiation of cell therapy with PPDCs. In addition, PPDCs have been shown not to stimulate allogeneic PBMCs in a mixed lymphocyte reaction.
Accordingly, transplantation with allogeneic, or even xenogeneic, PPDCs may be tolerated in some instances.
However, in other instances it may be desirable or appropriate to pharmacologically immunosuppress a patient prior to initiating cell therapy. This may be accomplished through the use of systemic or local irmnunosuppressive agents, or it may be accomplished by delivering the cells in an encapsulated device. PPDCs may be encapsulated in a capsule that is permeable to nutrients and oxygen required by the cell and therapeutic factors the cell is yet impermeable to immune humoral factors and cells. Preferably the encapsulant is hypoallergenic, is easily and stably situated in a target tissue, and provides added protection to the implanted structure. These and other means for reducing or eliminating an immune response to the transplanted cells are known in the art. As an alternative, PPDCs may be genetically modified to reduce their immunogenicity.
Survival of transplanted PPDCs in a living patient can be detemnined through the use of a variety of scanning techniques, e.g., computerized axial tomography (CAT or CT) scan, magnetic resonance imaging (MRI) or positron emission tomography (PET) scans.
Determination of transplant survival can also be done post nzortem by removing the target tissue, and examining it visually or through a microscope. Alternatively, cells can be treated with stains that are specific for cells of a specific lineage. Transplanted cells can also be identified by prior incorporation of tracer dyes such as rhodamine- or fluorescein-labeled microspheres, fast blue, bisbenzanude, ferric microparticles, or genetically introduced reporter gene products, such as beta-galactosidase or beta-glucuronidase.
Functional integration of transplanted PPDCs into a subject can be assessed by exanuning restoration of the function that was damaged or diseased, for example, restoration of joint or bone function, or augmentation of function.
Compositions and Pharmaceutical cof~zpositiorzs Compositions of PPDCs and related products (e.g., extracellular matrix, lysate, soluble cell fraction, conditioned medium), including for example pharmaceutical compositions, are included within the scope of the invention. Compositions of the invention may include one or more bioactive factors, for example but not limited to a growth factor, a differentiation-inducing factor, a cell survival factor such as caspase inhibitor, an anti-inflammatory agent such as p38 kinase inhibitor, or an angiogenic factor such as VEGF or bFGF. Some examples of bioactive factors include PDGF-bb, EGF, bFGF, IGF-l, and LIF. In some embodiments, undifferentiated or differentiation-induced PDPCs are cultured in contact with the bioactive factor. In some embodiments, undifferentiated PPDCs remain undifferentiated upon contact with the bioactive factor. In other embodiments, the bioactive factor induces differentiation of the PPDCs.
Pharmaceutical compositions of the invention may comprise homogeneous or heterogeneous populations of PPDCs, extracellular matrix or cell lysate thereof, or conditioned medium thereof in a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers for the cells of the invention include organic or inorganic carrier substances suitable which do not deleteriously react with the cells of the invention or compositions or components thereof. To the extent they are biocompatible, suitable pharmaceutically acceptable carriers include water, salt solution (such as Ringer's solution), alcohols, oils, gelatins, and carbohydrates, such as lactose, amylose, or starch, fatty acid esters, hydroxymethylcellulose, and polyvinyl pyrolidine.
Such preparations can be sterilized, and if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, and coloring. Pharmaceutical can-iers suitable for use in the present invention , are known in the art and are described, for example, in Pharmaceutical Sciences (17'h Ed., Mack Pub. Co., Easton, PA) and WO 96/05309, each of which are incorporated by reference herein.
The dosage (e.g., the number of cells to be administered) and frequency of administration of the pharmaceutical compositions of the invention will depend upon a number of factors, including but not limited to, the nature of the condition to be treated, the extent of the symptoms of the condition, characteristics of the patient (e.g., age, size, gender, health).
For example but not by way of limitation, PPDCs, extracellular matrix or cell lysates thereof, conditioned medium, compositions, and matrices produced according to the invention can be used to repair or replace damaged or destroyed cartilage tissue, to augment existing cartilage tissue, to introduce new or altered tissue, to modify artificial prostheses, or to join biological tissues or structures. For example, some embodiments of the invention would include (i) hip prostheses coated with replacement cartilage tissue constructs grown in three-dimensional cultures; (ii) knee reconstruction with cartilage tissue constructs; (iii) prostheses of other joints requiring reconstruction and/or replacement of articular cartilage; and (iv) cosmetic reconstruction with cartilage tissue constructs.
For example, the evaluation of internal derangements of articular cartilage in for example, the knee, hip, elbow, ankle and the glenohumeral joint, may be performed by arthroscopic techniques. In some embodiments, the injured or deteriorated portion of cartilage tissue is removed, for example, by arthroscopic surgery, followed by cartilage grafting. Cartilage tissue constructs may also be employed in reconstructive surgery for different types of joints.
Detailed procedures have been described in Resnick, D., and Niwayama, G., (eds), 1988, Diagnosis of Bone and Joint Disorders, 2d ed., W. B. Sanders Co., which is incorporated herein by reference.
Repair or replacement of damaged cartilage may be enhanced by fixation of the implanted cells and/or cartilage tissue at the site of repair. Various methods can be used to fix the new cells and/or cartilage tissue in place, including: patches derived from biocompatible tissues, which can be placed over the site; bioabsorbable sutures or other fasteners, e.g., pins, staples, tacks, screws and anchors; non-absorbable fixation devices, e.g., sutures, pins, screws and anchors; adhesives.
As another example but not by way of limitation, PPDCs, extracellular matrix or cell lysates thereof, conditioned medium, and the bone tissue produced according to the invention can be used to repair or replace damaged or destroyed bone tissue, to augment existing bone tissue, to introduce new or altered tissue, or to modify artificial prostheses. The cells of the invention may be administered alone, in a pharmaceutically acceptable carrier, or seeded on or in a matrix as described herein.
Use of PPDCs for Tra.~aspla~atatio~z The treatment methods of the subject invention involve the implantation of PPDCs or trans-differentiated cells into individuals in need thereof. The cells of the present invention may be allogeneic or autologous and may be delivered to the site of therapeutic need or "home" to the site.
The cells of the present invention may be differentiated ire vitro prior to implantation in a patient. Irr vitoo differentiation allows for controlled application of bioactive factors.
The cells of the present invention may be induced to differentiate ira situ or may be introduced irr vivo to provide trophic support to endogenous cells. The appropriate cell implantation dosage in humans can be determined from existing information relating to either the activity of the cells or the density of cells for bone or cartilage replacement. This information is also useful in calculating an appropriate dosage of implanted material.
Additionally, the patient can be monitored to determine if additional implantation can be made or implanted material reduced accordingly.
To enhance the differentiation, survival or activity of implanted cells, additional bioactive factors may be added including growth factors, anti-apoptotic agents (e.g., EPO, EPO

mimetibody, TPO, IGF-I and IGF-II, HGF, caspase inhibitors); anti-inflammatory agents (e.g., p38 MAPK inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMIJS, and NSAIDs (non-steroidal anti-inflammatory drugs;
e.g., TEPOXALIN, TOLMETIN, SUPROFEN); immunosupressive/immunomodulatory agents (e.g., calcineurin inhibitors, such as cyclosporine, tacrolimus; mTOR
inhibitors (e.g., SIROLIMUS, EVEROLIMLTS); anti-proliferatives (e.g., azathioprine, mycophenolate mofetil);
corticosteroids (e.g., prednisolone, hydrocortisone); antibodies such as monoclonal anti-IL.-2Ralpha receptor antibodies (e.g., basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g., anti-thymocyte globulin (ATG); anti-lymphocyte globulin (ALG);
monoclonal anti-T cell antibody OKT3)); anti-thrombogenic agents (e.g., heparin, heparin derivatives, urokinase, PPack (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); and anti-oxidants (e.g., probucol, vitamin A, ascorbic acid, tocopherol, coenzyme Q-10, glutathione, L-cysteine, N-acetylcysteine), and local anesthetics. To enhance vascularization and survival of transplanted bone tissue, angiogenic factors such as VEGF, PDGF or bFGF can be added either alone or in combination with endothelial cells or their precursors including CD34+, CD34+/CD117+ cells.
Alternatively, PPDCs to be transplanted may be genetically engineered to express such growth factors, antioxidants, antiapoptotic agents, anti-inflammatory agents, or angiogenic factors.
PPDCs can be. used to treat diseases or conditions of bone or cartilage or to augment or replace bone or cat-tilage. The disease or conditions to be treated include but we not limited to osteoarthritis, osteoporosis, rheumatoid arthritis, chondrosis deformans, dental and oral cavity disease (e.g., tooth fracture and defects), joint replacement, congenital abnormalities, bone fracture, and tumors (benign and malignant).
One or more other components may be added to transplanted cells, including selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs. Alternatively, the cells of the invention may be genetically engineered to express and produce for growth factors. Details on genetic engineering of the cells of the invention are provided infra. Bioactive factors which may be usefully incorporated into the cell formulation include anti-apoptotic agents (e.g., EPO, EPO mimetibody, TPO, IGF-I and IGF-II, HGF, caspase inhibitors); anti-inflammatory agents (e.g., p38 MAPK
inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and NSAIDs (non-steroidal anti-inflammatory drugs; e.g., TEPOKALIN, TOLMETIN, SUPROFEN); immunosupressive/immunomodulatory agents (e.g., calcineurin inhibitors, such as cyclosporine, tacrolimus; mTOR inhibitors (e.g., SIROLIMUS, EVEROLIMLTS); anti-proliferatives (e.g., azathioprine, mycophenolate mofetil);
corticosteroids (e.g., prednisolone, hydrocortisone); antibodies such as monoclonal anti-IL,-2Ralpha receptor antibodies (e.g., basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g., anti-thymocyte globulin (ATG); anti-lymphocyte globulin (ALG); monoclonal anti-T cell antibody OKT3));
anti-thrombogenic agents (e.g., heparin, heparin derivatives, urokinase, PPack (dextrophenylalanine proline arginine chloromethylketone), antithrombin compounds, platelet receptor antagonists, anti-thrombin antibodies, anti-platelet receptor antibodies, aspirin, dipyridamole, protanune, hirudin, prostaglandin inhibitors, and platelet inhibitors); and anti-oxidants (e.g., probucol, vitamin A, ascorbic acid, tocopherol, coenzyme Q-10, glutathione, L-cysteine, N-acetylcysteine), and local anesthetics. For example, the cells may be co-administered with scar inhibitory factor as described in U.S. Patent No. 5,827,735, incorporated herein by reference.
Fonnulatiorz of PPDCs for Transplarztatiozz In a non-limiting embodiment, a formulation comprising the cells of the invention is prepared for administration directly to the site where the production of new cartilage or bone, tissue is desired. For example, and not by way of limitation, the cells of the invention may be suspended in a hydrogel solution for injection. Examples of suitable hydrogels for use in the invention include self-assembling peptides, such as RAD16. Alternatively, the hydrogel solution containing the cells may be allowed to harden, for instance in a mold, to form a matrix having cells dispersed therein prior to implantation. Or, once the matrix has hardened, the cell formations may be cultured so that the cells are mitotically expanded prior to implantation. The hydrogel is an organic polymer (natural or synthetic) which is cross-linked via covalent, ionic, or hydrogen bonds to create a three-dimensional open-lattice structure which entraps water molecules to form a gel. Examples of materials which can be used to form a hydrogel include polysaccharides such as alginate and salts thereof, peptides, polyphosphazines, and polyacrylates, which are crosslinked ionically, or block polymers such as polyethylene oxide-polypropylene glycol block copolymers which are crosslinked by temperature or pH, respectively. In some embodiments, the support for the PPDCs of the invention is biodegradable.
In some embodiments of the invention, the formulation comprises an rn sitez polymerizable gel, as described, for example, in U.S. Patent Application Publication 2003/0022676; Anseth et al., J. Control Release, 78(1-3):199-209 (2002); Wang et al., Biomaterials, 24(22):3969-SO (2003).
In some embodiments, the polymers are at least partially soluble in aqueous solutions, such as water, buffered salt solutions, or aqueous alcohol solutions, that have charged side groups, or a monovalent ionic salt thereof. Examples of polymers with acidic side groups that can be reacted with cations are poly(phosphazenes), poly(acrylic acids), poly(methacrylic acids), copolymers of acrylic acid and methacrylic acid, polyvinyl acetate), and sulfonated polymers, such as sulfonated polystyrene. Copolymers having acidic side groups formed by reaction of acrylic or methacrylic acid and vinyl ether monomers or polymers can also be used. Examples of acidic groups are carboxylic acid groups, sulfonic acid groups, halogenated (preferably fluorinated) alcohol groups, phenolic OH groups, and acidic OH groups.
Examples of polymers with basic side groups that can be reacted with anions are polyvinyl amines), polyvinyl pyridine), polyvinyl imidazole), and some imino substituted polyphosphazenes. The ammonium or quaternary salt of the polymers can also be formed from the backbone nitrogens or pendant imino groups. Examples of basic side groups are amino and imino groups.
Alginate can be ionically cross-linked with divalent cations, in water, at room temperature, to form a hydrogel matrix. Due to these mild conditions, alginate has been the most commonly used polymer for hybridoma cell encapsulation, as described, for example, in U.S.
Patent No. 4,352,S83 to Lim. In the Lim process, an aqueous solution containing the biological materials to be encapsulated is suspended in a solution of a water soluble polymer, the suspension is formed into droplets which are configured into discrete microcapsules by contact with multivalent cations, then the surface of the microcapsules is crosslinked with polyamino acids to foam a semipermeable membrane around the encapsulated materials.
Polyphosphazenes are polymers with backbones consisting of nitrogen and phosphorous separated by alternating single and double bonds. Each phosphorous atom is covalently bonded to two side chains.
The polyphosphazenes suitable for cross-linking have a majority of side chain groups which are acidic and capable of forming salt bridges with di- or trivalent canons. Examples of preferred acidic side groups are carboxylic acid groups and sulfonic acid groups. Hydrolytically stable polyphosphazenes are formed of monomers having carboxylic acid side groups that are crosslinked by divalent or trivalent cations such as Ca2+ or A13+. Polymers can be synthesized that degrade by hydrolysis by incorporating monomers having imidazole, amino acid ester, or glycerol side groups. For example, a polyanionic poly[bis(carboxylatophenoxy)]phosphazene (PCPP) can be synthesized, which is cross-linked with dissolved multivalent rations in aqueous media at room temperature or below to form hydrogel matrices.
Biodegradable polyphosphazenes have at least two differing types of side chains, acidic side groups capable of forming salt bridges with multivalent rations, and side groups that hydrolyze under i~z vivo conditions, e.g., imidazole groups, amino acid esters, glycerol and glucosyl.
Hydrolysis of the side chain results in erosion of the polymer. Examples of hydrolyzing side chains are unsubstituted and substituted imidizoles and amino acid esters in which the group is bonded to the phosphorous atom through an amino linkage (polyphosphazene polymers in which both R groups are attached in this manner are known as polyaminophosphazenes). For polyimidazolephosphazenes, some of the "R" groups on the polyphosphazene backbone are imidazole rings, attached to phosphorous in the backbone through a ring nitrogen atom. Other "R" groups can be organic residues that do not participate in hydrolysis, such as methyl phenoxy groups or other groups shown in the scientific paper of Allcock, et al., Macromolecule 10:82 (1977). Methods of synthesis of the hydrogel materials, as well as methods for preparing such hydrogels, we known in the art.
Other components may also be included in the formulation, including but not limited to any of the following: (1) buffers to provide appropriate pH and isotonicity;
(2) lubricants; (3) viscous materials to retain the cells at or near the site of administration, including, for example, alginates, agars and plant gums; and (4) other cell types that may produce a desired effect at the site of administration, such as, for example, enhancement or modification of the formation~of tissue or its physicochemical characteristics, or as support for the viability of the cells, or inhibition of inflammation or rejection. The cells may be covered by an appropriate wound covering to prevent cells from leaving the site. Such wound coverings are known as those of skill in the art.
Fornzcclatiocz of a Cartilage or Bone Tissue Patclz Culture or co-cultures of PPDCs in a pre-shaped well enables the manufacture of a tissue patch of pre-determined thickness and volume. The volume of the resulting tissue patch is dependent upon the volume of the well and upon the number of PPDCs in the well. Tissue of optimal pre-determined volume may be prepared by routine experimentation by altering either or both of the aforementioned parameters.
The cell contacting surface of the well may be coated with a molecule that discourages adhesion of PPDCs to the cell contacting surface. Preferred coating reagents include silicon based reagents i.e., dichlorodimethylsilane or polytetrafluoroethylene based reagents, i.e., TEFLON. Procedures for coating materials with silicon based reagents, specifically dichlorodimethylsilane, are well known in the art. See for example, Sambrook et al. (1989) "Molecular Cloning A Laboratory Manual", Cold Spring Harbor Laboratory Press, the disclosure of which is incorporated by reference herein. It is appreciated that other biocompatible reagents that prevent the attachment of cells to the surface of the well may be useful in the practice of the instant invention.
Alternatively, the well may be cast from a pliable or moldable biocompatible material that does not permit attachment of cells per se. Preferred materials that prevent such cell attachment include, but are not limited to, agarose, glass, untreated cell culture plastic and polytetrafluoroethylene, i.e., TEFLON. Untreated cell culture plastics, i.e., plastics that have not been treated with or made from materials that have an electrostatic charge are commerciallyy available, and may be purchased, for example, from Falcon Labware, Becton-Dickinson, Lincoln Park, N.J. The aforementioned materials, however, are not meant to be limiting. It is appreciated that any other pliable or moldable biocompatible material that inherently discourages the attachment of PPDCs may be useful in the practice of the instant invention.
The size and shape of the well may be determined by the size and shape of the tissue defect to be repaired. For example, it is contemplated that the well may have a cross-sectional surface area of 25 cm'. This is the average cross-sectional surface area of an adult, human femoral chondyle. Accordingly, it is anticipated that a single piece of cartilage may be prepared in accordance with the invention in order to resurface the entire femoral chondyle. The depth of the well is preferably greater than about 0.3 cm and preferably about 0.6 cm in depth. The thickness of natural ai-ticular cartilage in an adult articulating joint is usually about 0.3 cm.
Accordingly, the depth of the well should be lwge enough to permit a cartilage patch of about 0.3 cm to form. The well should be deep enough to contain culture medium overlaying the tissue patch.
It is contemplated that a tissue patch prepared in accordance with the invention may be "trimmed" to a pre-selected size and shape by a surgeon performing surgical repair of the damaged tissue. Trimming may be performed with the use of a sharp cutting implement, i.e., a scalpel, a pair of scissors or an arthroscopic device fitted with a cutting edge, using procedures well known in the art.
The pre-shaped well may be cast in a block of agarose gel under aseptic conditions.
Agarose is an economical, biocompatible, pliable and moldable material that can be used to cast pre-shaped wells, quickly and easily. As mentioned above, the dimensions of the well may dependent upon the size of the resulting tissue plug that is desired.
A pre-shaped well may be prepared by pouring a hot solution of molten LT
agarose (BioRad, Richmond, Calif.) into a tissue culture dish containing a cylinder, the cylinder having dimensions that mirror the shape of the well to be formed. The size and shape of the well may be chosen by the artisan and may be dependent upon the shape of the tissue defect to be repaired.
Once the agarose has cooled and solidified around the cylinder, the cylinder is carefully removed with forceps. The surface of the tissue culture dish that is exposed by the removal of the cylinder is covered with molten agarose. This seals the bottom of the well and provides a cell adhesive surface at the base of the well. When the newly added molten LT agarose cools and solidifies, the resulting pre-shaped well is suitable for culturing, and inducing the differentiation of PPDCs.
It is appreciated, however, that alternative methods may be used to prepare a pre-shaped well useful in the practice. of the invention.
PPDCs in suspension may be seeded into and cultured in the pre-shaped well.
The PPDCs may be induced to differentiate to a chondrogenic or osteogenic phenotype in culture in the well or may have been induced to differentiate prior to seeding in the well. The cells may be diluted by the addition of culture medium to a cell density of about 1 x 105 to 1x109 PPDGs per milliliter.
The cells may form a cohesive plug of cells. The cohesive plug of cells may be removed from the well and surgically implanted into the tissue defect. It is anticipated that undifferentiated PPDCs may differentiate in situ thereby to form tissue i~a vivo.
Cartilage and bone defects may be identified inferentially by using computer aided tomography (CAT scanning); ~i-ray examination, magnetic resonance imaging (MRI), analysis of synovial fluid or serum markers or by any other procedures known in the art. Defects in mammals also are readily identifiable visually during arthroscopic examination orduring open surgery of the joint. Treatment of the defects can be effected during an arthroscopic or open surgical procedure using the methods and compositions disclosed herein.
Accordingly, once the defect has been identified, the defect may be treated by the following steps of (1) surgically implanting at the pre-determined site a tissue patch prepared by the methodologies described herein, and (2) permitting the tissue patch to integrate into pre-determined site.
The tissue patch optimally has a size and shape such that when the patch is implanted into the defect, the edges of the implanted tissue contact directly the edges of the defect. In addition, the tissue patch may be fixed in place during the surgical procedure. This can be effected by surgically fixing the patch into the detect with biodegradable sutures and/or by applying a bioadhesive to the region interfacing the patch and the defect.
In some instances, damaged tissue maybe surgically excised prior the to implantation of the patch of tissue.
T~ansplarrtati.on of PPDCs r~si.rag Scaffolds The cells of the invention or co-cultures thereof may be seeded onto or into a three-dimensional scaffold and implanted itr vivo, where the seeded cells will proliferate on the framework and form a replacement cartilage or bone tissue in vi.vo in cooperation with the cells of the patient.
In some aspects of the invention, the matrix comprises decellularized tissue, such as extracellular matrix, cell lysates (e.g., soluble cell fractions), or combinations thereof, of the PPDCs. In some embodiments, the matrix is biodegradable. In some aspects of the invention, the matrix comprises natural or synthetic polymers. Matrices of the invention include biocompatible scaffolds, lattices, self-assembling structures and the like, whether biodegradable or not, liquid or solid. Such matrices are known in the arts of cell-based therapy, surgical repair, tissue engineering, and wound healing. Preferably the matrices are pretreated (e.g., seeded, inoculated, contacted with) with the cells, extracellular matrix, conditioned medium, cell lysate, or combination thereof, of the invention. More preferably the matrices are populated with cells in close association to the matrix or its spaces. In some aspects of the invention, the cells adhere to the matrix. In some embodiments, the cells are contained within or bridge interstitial spaces of the matrix. Most preferred are those seeded matrices wherein the cells are in close association with the matrix and which, when used therapeutically, induce or support ingrowth of the patient's cells and/or proper angiogenesis. The seeded matrices can be introduced into a patient's body in any way known in the art, including but not limited to implantation, injection, surgical attachment, transplantation with other tissue, injection, and the like. The matrices of the invention may be configured to the shape andlor size of a tissue or organ i~a vivo.
For example, but not by way of limitation, the scaffold may be designed such that the scaffold structure: (1) supports the seeded cells without subsequent degradation; (2) supports the cells 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 iar vitro, at which point, the scaffold is degraded. A review of scaffold design is provided by Hutmacher, J. Biomat. Sci. Polymer- Edra., 12(1):107-124 (2001).

Scaffolds of the invention can be administered in combination with any one or more growth factors, cells, for example stem cells, bone marrow cells, chondrocytes, chondroblasts, osteocytes, osteoblasts, osteoclasts, bone lining cells, or their precursors, drugs or other components described above that stimulate tissue formation or otherwise enhance or improve the practice of the invention. The PPDCs to be seeded onto the scaffolds may be genetically engineered to express growth factors or drugs.
The cells of the invention can be used to produce new tissue in vitro, which can then be implanted, transplanted or otherwise inserted into a site requiring tissue repair, replacement or augmentation in a patient.
In a non-limiting embodiment, the cells of the invention are used to produce a three-dimensional tissue construct ir1 vitro, which is then implanted i~z vivo. As an example of the production of three-dimensional tissue constructs, see U.S. Patent No.
4,963,439, which is incorporated herein by reference. For example, the cells of the invention may be inoculated or "seeded" onto a three-dimensional framework or scaffold, and proliferated or grown in. vitro to form a living tissue that can be implanted in vivo.
The cells of the invention can be grown freely in a culture vessel to sub-confluency or confluency, lifted from the culture and inoculated onto a three-dimensional framework.
Inoculation of the three-dimensional framework with a high concentration of cells, e.g., approximately 10b to 5 x 10~ cells per milliliter, will result in the establishment of the three-dimensional support in relatively shorter periods of time.
Examples of scaffolds which may be used in the present invention include nonwoven mats, porous foams, or self assembling peptides. Nonwoven mats may, for example, be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the tradename VICRYL (Ethicon, Inc., Somerville, NJ).
Foams, composed of, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, formed by processes such as freeze-drying, or lyophilized, as discussed in U.S.
Patent No. 6,355,699, are also possible scaffolds. Hydrogels such as self-assembling peptides (e.g., RAD16) may also be used. These materials are frequently used as supports for growth of tissue.
The three-dimensional framework may be made of ceramic materials including, but not limited to: mono-, di-, tri-, alpha-tri-, beta-tri-, and tetra-calcium phosphate, hydroxyapatite, fluoroapatites, calcium sulfates, calcium fluorides, calcium oxides, calcium carbonates, magnesium calcium phosphates, biologically active glasses such as BIOGLASS
(University of Florida, Gainesville, FL), and mixtures thereof. There are a number of suitable porous biocompatible ceramic materials cmTently available on the commercial market such as SURGIBON (Unilab Surgibone, Inc., Canada), ENDOBON (Merck Biomaterial France, France), CEROS (Mathys, A. G., Bettlach, Switzerland), and INTERPORE (Interpore, Irvine, CA, United States), and mineralized collagen bone grafting products such as HEALOS
(Orquest, Inc., Mountain View, CA) and VITOSS, RHAKOSS, and CORTOSS (Orthovita, Malvern, PA).
The framework may be a mixture, blend or composite of natural and/or synthetic materials. In some embodiments, the scaffold is in the form of a cage. In a preferred embodiment, the scaffold is coated with collagen.
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 stmctures. In addition, the three-dimensional framework may be molded into a useful shape, such as that of the external portion of the ear, a bone, joint or other specific structure in the body to be repaired, replaced or augmented.
In another preferred embodiment, the cells of the invention are seeded onto a framework comprising a prosthetic device for implantation into a patient, as described in U.S. Patent No.
6,200,606, incorporated herein by reference. As described therein, a variety of clinically useful prosthetic devices have been developed for use in bone and cartilage grafting procedures. (see e.g. Bone Grafts and Bone Substitutions. Ed. M. B. Habal & A. H. Reddi, W. B.
Saunders Co., 1992). For example, effective knee and hip replacement devices have been and continue to be widely used in the clinical environment. Many of these devices are fabricated using a variety of inorganic materials having low immunogenic activity, which safely function in the body.
Examples of synthetic materials which have been tried and proven include titanium alloys, calcium phosphate, ceramic hydroxyapatite, and a variety of stainless steel and cobalt-chrome alloys. These materials provide structural support and can form a scaffolding into which host vascularization and cell migration can occur. The present invention provides a source of cells which may be used to "seed" such prosthetic devices. In the preferred embodiment PPDCs are first mixed with a carrier material before application to a device. Suitable carriers well known to those skilled in the art include, but are not limited to, gelatin, fibrin, collagen, starch, polysaccharides, saccharides, proteoglycans, synthetic polymers, calcium phosphate, or ceramics.

The framework may be treated prior to inoculation of the cells of the invention in order to enhance cell attachment. For example, prior to inoculation with the cells of the invention, nylon matrices could be treated with 0.1 molar acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon. Polystyrene could be similarly treated using sulfuric acid.
In addition, the external surfaces of the three-dimensional framework may be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma coating the framework or addition of one or more proteins (e.g., collagens, elastic fibers, reticular fibers), glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, among others.
In some embodiments, the scaffold is comprised of or is treated with materials that render it non-thrombogenic. These treatments and materials may also promote and sustain endothelial growth, migration, and extracellular matrix deposition. Examples of these materials and treatments include but are not limited to natural materials such as basement membrane proteins such as laminin and Type N collagen, synthetic materials such as ePTFE, and segmented polyurethaneurea silicones, such as PURSPAN (The Polymer Technology Group, Inc., Berkeley, GA). 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.
In some embodiments, the surface of the scaffold is textured. For example, in some aspects of the invention, the scaffold is provided with a groove and ridge pattern. The grooves are preferably less than about 500 microns, more preferably less than about 100 microns, and most preferably between about 10 nanometers and 10 microns. Such "mierogrooves" allow the cells to align and/or migrate guided by the surface grooves. See, e.g., Odontology. 2001;89(1):2-11. The textured scaffold may be used, for example, as a dental implant.
In some embodiments, it is important to re-create in culture the cellular microenvironment found lit VL1~0, such that the extent to which the cells of the invention are grown prior to implantation irz vivo or use izz vi.tr-o may vary. In addition, growth factors, chondrogenic differentiation inducing agents, osteogenic inducing agents, and angiogenic factors may be added to the culture medium prior to, during, or subsequent to inoculation of the cells to trigger differentiation and tissue formation by the PPDCs or co-cultures thereof.
The three-dimensional framework rnay be modified so that the growth of cells and the production of tissue thereon is enhanced, or so that the risk of rejection of the implant is reduced.

Thus, one or more biologocally active compounds, including, but not limited to, anti-inflammatories, immunosuppressants or growth factors, may be added to the framework.
Therapeutic Uses for Extracellular Matrix or Cell Lysate Derived Front PPDCs As an alternative to implanting the cells of the invention, or living tissue produced therefrom, a subject in need of tissue repair, replacement, or augmentation may benefit from the administration of a component or product of PPDCs, such as the extracellular matrix (ECM) or cell lysate produced by those cells.
In some embodiments, after the cells of the invention have been cultured i~t vitro, such as, for example, by using a three-dimensional scaffold system described herein, such that a desired amount of ECM has been secreted onto the framework. Once ECM is secreted onto the framework, the cells may be removed. The ECM may be processed for further use, for example, as an injectable preparation.
In some embodiments, the cells are killed and cellular debris (e.g., cellular membranes) is removed from the framework. This process may be carried out in a number of different ways.
For example, the living tissue can be flash-frozen in liquid nitrogen without a cryopreservative, or the tissue can be immersed in sterile distilled water so that the cells burst in response to osmotic pressure. Once the cells have been killed, the cellular membranes may be disrupted and cellular debris removed by treatment with a mild detergent rinse, such as EDTA, CHAPS or a zwitterionic detergent. An advantage to using a mild detergent rinse is that it solubilizes membrane-bound proteins, which are often highly antigenic.
Alternatively, the tissue can be enzymatically digested and/or extracted with reagents that break down cellular membranes. Example of such enzymes include, but are not limited to, hyaluronidase, dispase, proteases, and nucleases (for example, deoxyribonuclease and ribonuclease). Examples of detergents include non-ionic detergents such as, for example, alkylaryl polyether alcohol (TRITON~ X-100), octylphenoxy polyethoxy-ethanol (Rohm and Haas Philadelphia, PA), BRIJ-35, a polyethoxyethanol lauryl ether (Atlas Chemical Co., San Diego, CA), polysorbate 20 (TWEEN 20~), a polyethoxyethanol sorbitan monolaureate (Rohm and Haas), polyethylene lauryl ether (Rohm and Haas); and ionic detergents such as, for example, sodium dodecyl sulphate, sulfated higher aliphatic alcohols, sulfonated alkanes and sulfonated alkylarenes containing 7 to 22 carbon atoms in a branched or unbranched chain.
Scaffolds comprising the ECm may be used therapeutically as described above.
Alternatively, ECM may be collected from the scaffold. The collection of the ECM can be accomplished in a variety of ways, depending, for example, on whether the scaffold is bioaegradable or non-bioaegraaabie. For example, if the framework is non-biodegradable, the ECM can be removed by subjecting the framework to sonication, high pressure water jets, mechanical scraping, or mild treatment with detergents or enzymes, or any combination of the above.
If the framework is biodegradable, the ECM can be collected, for example, by allowing the framework to degrade or dissolve in solution. Alternatively, if the biodegradable framework is composed of a material that can itself be injected along with the ECM, the framework and the ECM can be processed ifa toto for subsequent injection. Alternatively, the ECM
can be removed from the biodegradable framework by any of the methods described above for collection of ECM
from a non-biodegradable framework. All collection processes are preferably designed so as not to denature the ECM or cell lysate produced by the cells of the invention.
Once the ECM has been collected, it may be processed further. The ECM can be homogenized to fine particles using techniques well known in the art such as, for example, by sonication, so that they can pass through a surgical needle. ECM components can be crosslinked, if desired, by gamma irradiation. Preferably, the ECM can be irradiated between 0.25 to 2 mega rads to sterilize and crosslink the ECM. Chemical crosslinking using agents that are toxic, such as glutaraldehyde, is possible but not generally preferred.
r Cell lysates prepwed from the populations of the postpartum-derived cells also have many utilities. In one embodiment, whole cell lysates are prepared, e.g., by disrupting cells without subsequent separation of cell fractions. In another embodiment, a cell membrane fraction is separated from a soluble fraction of the cells by routine methods known in the art, e.g., centrifugation, filtration, or similar methods. Use of soluble cell fractions in. vi.oo allows the beneficial intracellular milieu to be used in a patient without triggering rejection or an adverse response. 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 other solution. 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, partially purified, combined with pharmaceutically acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions. Cell lysates may be used in vitro or iaa vi.vo, alone or for example, with cells. The cell lysates, if introduced in vivo, may be introduced locally at a site or treatment, or remotely to provide, for example needed cellular growth factors to a patient.
The amounts and/or ratios of proteins may be adjusted by mixing the ECM or cell lysate produced by the cells of the invention with ECM or cell lysate of one or more other cell types. In addition, biologically active substances such as proteins, growth factors and/or drugs, can be incorporated into the ECM or cell lysate preparation. Exemplary biologically active substances include anti-inflammatory agents and growth factors which promote healing and tissue repair.
Cells may be co-administered with the ECM or cell lysates of the invention.
ECM or cell lysate of PPDCs may be formulated for administration as described above for PPDCs.
The above described process for preparing injectable ECM or cell lysate is preferably canied out under sterile conditions using sterile materials. The processed ECM
or cell lysate in a pharmaceutically acceptable carrier can be injected intradermally or subcutaneously to treat bone or cartilage conditions, for example, by augmenting tissue or repairing or correcting congenital anomalies, acquired defects or cosmetic defects.
Use of PPDCs for In Vitro Screening of Drrcg Effrcacy or Toxicity The cells and tissues of the invention may be used i~z vitro to screen a wide variety of compounds for effectiveness and eytotoxicity of pharmaceutical agents, growth/regulatory factors, anti-inflammatory agents. To this end, the cells of the invention, or tissue cultures described above, are maintained in vitro and exposed to the compound to be tested. The activity of a cytotoxic compound can be measured by its ability to damage or kill cells in culture. This may readily be assessed by vital staining techniques. The effect of growth/regulatory factors may be assessed by analyzing the number of living cells in vitro, e.g., by total cell counts, and differential cell counts. This may be accomplished using standard cytological and/or histological techniques, including the use of immunocytochemical techniques employing antibodies that define type-specific cellular antigens. The effect of various drugs on the cells of the invention either in suspension culture or in the three-dimensional system described above may be assessed.
The cells and tissues of the invention may be used as model systems for the study of physiological or pathological conditions. For example, joints that are immobilized suffer relatively quickly in a number of respects. The metabolic activity of chondrocytes appears affected as loss of proteoglycans and an increase in water content are soon observed. The normal white, glistening appearance of the cartilage changes to a dull, bluish color, and the cartilage thickness is reduced. However, the amount of this change that is due to nutritional deficiency versus the amount due to upset in the stress-dependent metabolic homeostasis is not yet clear.

The cells and tissues of the invention may be used to determine the nutritional requirements of cartilage under different physical conditions, e.g., intermittent pressurization, and by pumping action of nutrient medium into and out of the cartilage construct. This may be especially useful in studying underlying causes for age-related or injury-related decrease in tensile strength of, for example, articular cartilage, e.g., in the knee, that predispose the weakened cartilage to traumatic damage.
The cells and tissues of the invention may also be used to study the mechanism of action of cytokines, growth factors and inflammatory mediators, e.g., IL-1, TNF and prostaglandins. In addition, cytotoxic and/or pharmaceutical agents can be screened for those that are most efficacious for a particular patient, such as those that reduce or prevent resorption of cartilage or bone otherwise enhance the balanced growth thereof. Agents that prove to be efficacious in vitro could then be used to treat the patient therapeutically.
Use of PPDCs to Produce Biological Molecules In a further embodiment, the cells of the invention can be cultured ira vitro to produce biological products in high yield. For example, such cells, which either naturally produce a particular biological product of interest (e.g., a growth factor, regulatory factor, or peptide hormone), or have been genetically engineered to produce a biological product, could be clonally expanded using, for example, the three-dimensional culture system described above. If the cells excrete the biological product into the nutrient medium, the product can be readily isolated from the spent or conditioned medium using standard separation techniques, e.g., such as differential protein precipitation, ion-exchange chromatography, gel filtration chromatography, electrophoresis, and high performance liquid chromatography. A "bioreactor"
may be used to take advantage of the flow method for feeding, for example, a three-dimensional culture iu vitro.
Essentially, as fresh media is passed through the three-dimensional culture, the biological product is washed out of the culture and may then be isolated from the outflow, as above.
Alternatively, a biological product of interest may remain within the cell and, thus, its collection may require that the cells be lysed. The biological product may then be purified using any one or more of the above-listed techniques.
Kits The PPDCs and components and products thereof can conveniently be employed as part of a kit, for example, for culture or implantation. Accordingly, the invention provides a kit including the PPDCs and additional components, such as a matrix (e.g., a scaffold), 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 powdered form), antibiotic compounds, hormones, and the like. While the kit can include any such components, preferably it includes all ingredients necessary for its intended use. If desired, the lit also can include cells (typically cryopreserved), which can be seeded into the lattice as described herein.
In another aspect, the invention provides kits that utilize the PPDCs, PPDC
populations, components and products of PPDCs in various methods for augmentation, regeneration, and repair as described above. In some embodiments, the kits may include one or more cell populations, including at least PPDCs and a pharmaceutically acceptable carrier (liquid, semi-solid or solid). The kits also optionally may include a means of administering the cells, for example by injection. The kits further may include instructions for use of the cells. Kits prepared for field hospital use, such as for military use, may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute injuries. Kits for assays and iaa vitro methods as described herein may contain one or more of (1) PPDCs or components or products of PPDCs, (2) reagents for practicing the in vitro method, (3) other cells or cell populations, as appropriate, and (4) instructions for conducting the in. vitro method.
Cryopreservation and Banking PPDCs PDCs of the invention can be cryopreserved and maintained or stored in a "cell bank".
Cryopreservation of cells of the invention may be earned out according to known methods. For example, but not by way of limitation, cells may be suspended in a "freeze medium" such as, for example, culture medium further comprising 0 to 95 percent FBS and 0 to 10 percent dimethylsulfoxide (DMSO), with or without 5 to 10 percent glycerol, at a density, for example, of about 0.5 to 10 x 106 cells per milliliter. The cryopreservation medium may comprise cryopreservation agents including but not limited to methylcellulose. The cells are dispensed into glass or plastic ampoules that are then sealed and transferred to the freezing chamber of a controlled rate freezer. The optimal rate of freezing may be determined empirically. A
programmable rate freezer for example, can give a change in temperature of -1 to -10°C per minute. The preferred cryopreservation temperature is about -80°C to about -180°C, more preferably is about -90°C to about -160°C, and most preferably is about -125 to about -140°C.
Cryopreserved cells preferably are transferred to liquid nitrogen prior to thawing for use. In some embodiments, for example, once the ampoules have reached about -90°C, they are transferred to a liquid nitrogen storage area. Cryopreserved cells can be stored for a period of years.
The cryopreserved cells of the invention constitute a bank of cells, portions of which can be "withdrawn" by thawing and then used as needed. Thawing should generally be carried out rapidly, for example, by transfernng an ampoule from liquid nitrogen to a 37°C water bath. The thawed contents of the ampoule should be immediately transferred under sterile conditions to a culture vessel containing an appropriate medium such as DMEM conditioned with 10 percent FBS.
In yet another aspect, the invention also provides for banking of tissues, cells, cellular components and cell populations of the invention. As discussed above, the cells are readily cryopreserved. The invention therefore provides methods of cryopreserving the cells in a bank, wherein the cells are stored frozen and associated with a complete characterization of the cells based on immunological, biochemical and genetic properties of the cells. The cells so 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 co-cultures, in the presence or absence of a matrix or support. While for some applications it may be preferable to use cells freshly prepared, the remainder can be cryopreserved and banked by freezing the cells and entering the information in the computer to associate the computer entry with the samples. Even where it is not necessary to match a source or donor with a recipient of such cells, for immunological proposes, 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 banked sample, the sample is retrieved , and readied for therapeutic use. Cell lysates or components prepared as described herein may also be preserved (e.g., cryopreserved, lyophilized) and banked in accordance with the present invention.
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.
EXAMPLES

EhAMPLE 1 Derivation of Cells from Postpartum Tissues The objective of this study was to derive populations of cells from placental and umbilical cord tissues. Postpartum umbilical cord and placenta were obtained upon birth of either a full term or pre-term pregnancy. Cells were harvested from 5 separate donors of umbilical cord and placental tissue. Different methods of cell isolation were tested fox their ability to yield cells with: I) the potential to differentiate into cells with different phenotypes, or 2) the potential to provide critical trophic factors useful for other cells and tissues.
Metlaorls & Materials Umbilical cord cell derivation. 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 umbilical cord was washed in phosphate buffered saline (PBS;
Invitrogen, Carlsbad, CA) in the presence of antimycotic and antibiotic (100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B
(Invitrogen Carlsbad, CA)). The tissues were then mechanically dissociated in 150 cm' 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 antimycotic and antibiotic (100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin 8 (Invitrogen)) and digestion enzymes.
In some experiments, an enzyme mixture of collagenase and disease was used ("C:D;"
collagenase (Sigma, St Louis, MO), 500 Units/milliliter; and disease (Invitrogen), 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 Unitshnilliliter; 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, NY) at 225 rpm for 2 hrs.
After digestion, the tissues were centrifuged at 150 x g for 5 minutes, and 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#AND18475; Hyclone, Logan, UT), 0.001% (v/v) 2-mercaptoethanol (Sigma), 100 Units/milliliter of penicillin, 100 microgram/milliliter streptomycin, 0.25 microgram/milliliter amphotericin B ( Invitrogen, Carlsbad, CA). The cell suspension was filtered through a 70-micrometer nylon cell strainer (BD Biosciences). An additional 5 milliliter rinse comprising Growth medium was passed through the strainer. The cell suspension was then passed through a 40-micrometer nylon cell strainer (BD Biosciences) and chased with a rinse of an additional 5 milliliters of Growth medium.
The filtrate was resuspended in Growth medium (total volume 50 milliliters) and centrifuged at 150 x g for 5 minutes. The supernatant was aspirated, and the cells were re.suspended 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 Trypaii 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 (DMEM-Low glucose (Invitrogen), 15 percent (v/v) defined bovine serum (Hyclone, Logan, UT;
Lot#AND18475), 0.001 percent (v/v) 2-mercaptoethanol (Sigma), 100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B
(Invitrogen)).
After about 2-4 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/cm2.
Cells were grown in a humidified incubator with 5 percent carbon dioxide and 20 percent oxygen at 37°C.
Placental Cell Isolation. Placental tissue was obtained from NDRI
(Philadelphia, PA).
The tissues were from a pregnancy and were obtained at the time of a normal surgical delivery.
Placental cells were isolated as described for umbilical cord cell isolation.
The following example 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 (100 U/milliliter penicillin, 100 microgram/milliliter streptomycin, 0.25 microgramlmilliliter amphotericin B; Invitrogen) to remove blood and debris.
The placental tissue was then dissected into three sections: top-line (neonatal side or aspect), mid-line (mixed cell isolation neonatal and maternal or vinous region), and bottom line (maternal side or aspect).
The separated sections were individually washed several times in PBS with antibiotic/antimycotic to further remove blood and debris. Each section was then mechanically dissociated in 150 cm2 tissue culture plates in the presence of 50 nulliliters 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 or DMEM-High glucose medium containing antimycotic and antibiotic (100 Units/milliliter penicillin, 100 microgramslnulliliter streptomycin, 0.25 micrograms/milliliter amphotericin B (Invitrogen)) and digestion enzymes. In some experiments an enzyme mixture of eollagenase and dispase ("C:D") was used containing collagenase (Sigma, St Louis, MO) at 500 Units/milliliter and dispase (Invitrogen) at 50 Units/milliliterin DMEM-Low glucose medium. In other experiments a mixture of collagenase, dispase, and hyaluronidase (C:D:H) was used (collagenase, 500 Units/nulliliter; dispase, 50 Units/milliliter; and hyaluronidase (5igma), 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 milliliter of Growth medium (DMEM-Low glucose (Invitrogen), 15 % (v/v) fetal bovine serum (FBS; defined bovine serum;
Lot#AND18475; Hyclone, Logan, UT), 0.001% (v/v) 2-mercaptoethanol (Sigma, St.
Louis, MO), antibiotic/antimycotic (100 U/nulliliter penicillin, 100 microgram/milliliter streptomycin, 0.25 microgram/rnilliliter amphotericin B; Invitrogen)). The cell suspension was filtered through a 70 micrometer nylon cell strainer (BD Biosciences), chased by a rinse with an additional 5 milliliters of Growth medium. The total cell suspension was passed through a 40 micrometer nylon cell strainer (BD Biosciences) followed with an additional 5 milliliters of Growth medium as a rinse.
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 then cultured at standard conditions.

LIBERASE (Boehringer Mannheim Corp., Indianapolis, IN) Cell Isolation. Cells were isolated from umbilical cord in DMEM-Low glucose medium with LIBERASE
(Boehringer Mannheim Corp., Indianapolis, IN) (2.5 milligrams per milliliter, Blendzyme 3;
Roche Applied Sciences, Indianapolis, IN) and hyaluronidase (5 Units/milliliter, Sigma).
Digestion of the tissue and isolation of the cells was as described for other protease digestions above using a LIBERASE (Boehringer Mannheim Corp., Indianapolis, IN)/hyaluronidase mixture in place of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE
(Boehringer Mannheim Corp., Indianapolis, IN) 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. Attempts were made to isolate pools of cells from umbilical cord 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. From these experiments a cell population was isolated that readily expanded.
Isolation of cells from Cord Blood. Cells have also been isolated from cord blood samples attained from NDRI. The isolation protocol used here was that of International Patent Application W002/29971 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 for5 seconds, and incubated for 2 minutes at ambient temperature. The soluble cell fraction was centrifuged (10 minutes at 200 x g). The cell pellet was resuspended in complete minimal essential medium (Gibco, Carlsbad CA) containing 10 percent fetal bovine serum (Hyclone, Logan UT), 4 millimolar glutamine (Mediatech Herndon, VA ), 100 Units penicillin per 100 milliliters and 100 micrograms streptomycin per 100 milliliters (Gibco, Carlsbad, CA). The resuspended cells were centrifuged (10 minutes at 200 x g), the supernatant was aspirated, and the cell pellet was washed in complete medium. Cells were seeded directly into either T75 flasks (Corning, NY), T75 laminin-coated flasks, or T175 fibronectin-coated flasks (both Becton Dickinson, Bedford, MA).
Isolation of postpartum-derived cells using different enzyme combinations and growth conditions. To determine whether cell populations can be isolated under different conditions and expanded under a variety of conditions immediately after isolation, cells were digested in Growth medium with or without 0.001 percent (v/v) 2-mercaptoethanol (Sigma, St.
Louis, MO), using the enzyme combination of C:D:H, according to the procedures provided above. Placenta-derived cells so isolated were seeded under a variety of conditions. All cells were grown in the presence of penicillinlstreptomycin. (Table 1-2).
Isolation of postpartum-derived cells using different enzyme combinations and growth conditions. In all conditions, cells attached and expanded well between passage 0 and 1 (Table 1-2). Cells in conditions 5 to S and 13 to 16 were demonstrated to proliferate. well up to 4 passages after seeding at which point they were cryopreserved. All cells were banked.
Results Cell isolation using different enzyme combinations. The combination of C:D:H
provided the best cell yield following isolation and generated cells which expanded for many more generations in culture than the other conditions (Table 1-1). 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 collagen that was tested.
Isolation of postpartum-derived cells using different enzyme combinations and growth conditions. Cells attached and expanded well between passage 0 and 1 under all conditions tested for enzyme digestion and growth (Table 1-2). Cells in experimental conditions 5-S and 13-1C proliferated well up to 4 passages after seeding, at which point they were cryopreserved. All cells were banked.
Isolation of cells from residual blood in the cords. Nucleated cells attached and grew rapidly. These cells were analyzed by flow cytometry and were similar to cells obtained by enzyme digestion.
Isolation of cells from Cord 131ood. The preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks.
The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.

Summary. Populations of cells can be isolated from umbilical cord and placental tissue most efficiently using the enzyme combination collagenase (a matrix metalloprotease), dispase (neutral protease), and hyaluronidase (a mucolytic enzyme which breaks down hyaluronic acid).
LTBERASE (Boehringer Mannheim Corp., Indianapolis, IN), which is a Blendzyme, may also be used. In the present study Blendzyme 3 which is collagenase (4 Wunsch units/g) and thermolysin (1714 casein LTnitslg) was also used together with hyaluronidase to isolate cells.
These cells expand readily over many passages when cultured in Growth medium on gelatin-coated plastic.
Postpartum-derived cells were isolated from residual blood in the cords but not from 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.
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-derived cells under varying conditions:
ConditionMedium 15% FBS BME Gelatin 2070 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 YY N

4 DMEM-Lg Y Y N N (5%) N

DMEM-Lg N (2%) Y N (Laminin)Y EGF/FGF (20 ng/mL) 6 DMEM-Lg N (2%) Y N (Laminin)N (5%) EGFIFGF (20 ng/mL)
7 DMEM-Lg N (2%) Y N Y PDGF/VEGF
(Fibronectin)
8 DMEM-Lg N (2%) Y N N (5%) PDGF/VEGF
(Fibronectin)
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/mL) 14 DMEM-Lg N (2%) N N (Laminin)N (5%) EGF/FGF (20 ng/mL) DMEM-Lg N (~%) N N Y PDGF/VEGF
(Fibronectin) 16 DMEM-Lg N (2%) N N N (5%) PDGF/VEGF
(Fibronectin) Reference 1. HO, Tony, W. ;KOPEN, Gene, C. ;RIGHTER, William, F. ;RUTKOWSKI, J., Lynn ;HERRING, W., Joseph ;RAGAGLIA, Vanessa ;WAGNER, Joseph W02003025149 A2 CELL POPULATIONS WHICH CO-EXPRESS CD49C AND CD90, NEURONYX, INC.
Application No. US0229971 US, Filed 20020920, A2 Published 20030327 ,A3 Published 20031218.

Evaluation of Growth Media for Postpartum-derived Gells Several cell culture media were evaluated for their ability to support the growth of placenta-derived cells. The growth of placenta-derived cells in normal (20%) and low (5%n) oxygen was assessed after 3 days using the MTS colorimetric assay.
Metlzocls & Materials Placenta-derived cells at passage 8 (P8) were seeded at 1 x 103 cells/well in 96 well plates in Growth medium (DMEM-low glucose (Gibco, Carlsbad CA), 15% (v/v) fetal bovine serum (Cat. #SH30070.03; Hyclone, Logan, UT), 0.001% (v/v) betamercaptoethanol (Sigma, St. Louis, MO), 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco)). After 8 hours, the medium was changed as described in Table 2-1, and cells were incubated in normal (20%, v/v) or low (5%, v/v) oxygen at 37°C, 5% COZ for 48 hours. MTS
was added to the culture. medium (CELLTITER 96 AQueous One Solution Cell Proliferation Assay, Promega, Madison, WI) for 3 hours and the absorbance measured at 490 nanometers (Molecular Devices, Sunnyvale CA).
Table 2-1 Culture medium Culture Medium Supplier Added fetal bovine serum (v/v) DMEM-low glucose Gibco Carlsbad CA 0, 2, 10 DMEM-high glucose Gibco 0, 2, 10 RPMI 1640 Mediatech, Inc. 0, 2, 10 Herndon, VA

Cell gro-free (Serum-free,Mediatech, Inc. -Protein-free) Ham's F10 Mediatech, Inc. 0, 2, 10 MSCGM (complete with Cambrex, Walkersville,0, 2, 10 serum) MD

Complete-serum free Mediatech, Inc. -w/albunun Growth medium NA -Ham's F12 Mediatech, Inc. 0, 2, 10 Iscove's Mediatech, Inc. 0, 2, 10 Basal Medium Eagle's Mediatech, Inc. 0, 2, 10 DMEM/F12 (1:1) Mediatech, Inc. 0, 2, 10 Resrclts Standard curves for the MTS assay established a linear correlation between an increase in absorbance and an increase in cell number. The absorbance values obtained were converted into estimated cell numbers and the change (%) relative to the initial seeding was calculated.
The Effect of Serum. The addition of serum to media at normal oxygen conditions resulted in a reproducible dose-dependent increase in absorbance and thus the viable cell number. The addition of serum to complete MSCGM resulted in a dose-dependent decrease in absorbance. In the media without added serum, cells grew in Cellgro, Ham's F10, and DMEM.

The Effect of Oxygen. Reduced oxygen appeared to increase the growth rate of cells in Growth Medium, Ham's F10, and MSCGM.
In decreasing order of growth, the media resulting in the best growth of the cells were Growth medium >MSCGM> Iscove's+10% FBS = DMEM-HG +10% FBS = Ham's F12 +10%
FBS = RPMI 1640 +10% FBS.
Summary. Postpartum-derived cells may be grown in a variety of culture media in normal or low oxygen. Short-term growth of placenta-derived cells was determined in 12 basal media with 0, 2, and 10% (v/v) serum in 5% or 20% O~. In general placenta-derived cells did not grow in serum-free conditions with the exceptions of Ham's F10 and Cellgro-free, which are also protein-free. Growth in these serum-free media was approximately 25-33, %
of the maximal growth observed with Growth medium containing 15% serum. This study demonstrates that placenta-derived cells may be grown in serum-free conditions and that Growth medium is one of several media (10% serum in Iscove's, RPMI or Ham's F12 media) that can be used to grow placenta-derived cells.
The most promising serum-free media was CELLGRO-FREE, a serum and protein-free medium without hormones or growth factors, which is designed for the growth of mammalian cells in vitro (Mediatech product information).
Complete-serum free medium also developed for serum-free culture was not as effective in supporting growth of the placenta-derived cells. Complete-serum free was developed by Mediatech, based on a 50/50 mix of DMEM/F12 with smaller percentages of RPMI
1640 and McCoy's 5A. This medium also contains selected trace elements and high molecular weight carbohydrates, extra vitamins, a non-animal protein source, and a small amount of BSA (1 gram/liter). It does not contain any insulin, transfernn, cholesterol, or growth or attachment factors. It is bicarbonate buffered for use with 5% CO~. Originally designed for hybridomas and suspension cell lines, it may be suitable for some anchorage dependent cell lines.

Growth of Postpartum-derived cells in Medium containing D-Valine It has been reported that medium containing D-valine instead of the normal L-valine isoform can be used to selectively inhibit the growth of fibroblast-like cells in culture (Hongpaisan, 2000; Sordillo et al., 1988). The growth of postpartum-derived cells in medium containing D-valine in the absence of L-valine was evaluated.
Nlethods & Mate~zals Placenta-derived cells (P3), fibroblasts (P9), and umbilical cord-derived cells (P5) were seeded at 5 x 103 cells/cm2 in gelatin-coated T75 flasks (Corning, Corning, NY). After 24 hours the medium was removed and the cells were washed with phosphate buffered saline (PBS) (Gibco, Carlsbad, CA) to remove residual medium. The medium was replaced with a Modified Growth medium (DMEM with D-valine (special order Gibco), 15% (v/v) dialyzed fetal bovine serum (Hyclone, Logan, UT), 0.001% (v/v) betamercaptoethanol (Sigma), 50 LTnits/milliliter penicillin, 50 microgram/milliliter streptomycin (Gibco)).
Resrclts Placenta-derived, umbilical cord-derived, and fibroblast cells seeded in the D-valine-containing medium did not proliferate, unlike cells seeded in Growth medium containing dialyzed serum. Fibroblasts changed morphologically, increasing in size and changing shape.
All of the cells died and eventually detached from the flask surface after 4 weeks.
Summary. Postpa~.-tum-derived cells require L-valine for cell growth and for long-term viability. L-valine is preferably not 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 Ifat. 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 hzt Rep.1'~:355-64.

Cryopreservation Media for Postpartum-derived Cells The objective of this study was to determine a suitable cryopreservation medium for the cryopreservation of postpartum-derived cells.
Methods & Materials Placenta-derived cells grown in Growth medium (DMEM-low glucose (Gibco, Carlsbad CA), 15% (v/v) fetal bovine serum (Cat. #SH30070.03, Hyclone, Logan, UT), 0.001% (v/v) betamercaptoethanol (Sigma, St. Louis, MO), 50 Units/milliliter penicillin, 50 microgram/milliliter streptomycin (Gibco)), in a gelatin-coated T75 flask were washed with phosphate buffered saline (PBS; Gibco) and trypsinized using 1 milliliter Trypsin/EDTA
(Gibco). The trypsinization was stopped by adding 10 milliliter Growth medium.
The cells were centrifuged at 150 x g, supernatant removed, and the cell pellet was resuspended in 1 milliliter Growth medium. An aliquot of cell suspension, 60 microliter, was removed and added to 60 microliter, trypan blue (Sigma). The viable cell number was estimated using a hemocytometer.
The cell suspension was divided into four equal aliquots each containing 88 x 104 cells each. The cell suspension was centrifuged and resuspended in 1 milliliter of each media below and transferred into Cryovials (Nalgene).
1.) Growth medium +10% (v/v) DMSO (Hybrimax, Sigma, St. Louis, MO) 2.) Cell Freezing medium w/DMSO, w/methylcellulose, serum-free (C6295, Sigma, St. Louis, MO) 3.) Cell Freezing medium serum-free (C2639, Sigma, St. Louis, MO) 4.) Cell Freezing Medium w/glycerol (C6039, Sigma, St. Louis, MO) The cells were cooled at approximately 1 °C/min overnight in a -80°C freezer using a "Mr Frosty" freezing container according to the manufacturer's instructions (Nalgene, Rochester, NY). Vials of cells were transfeiTed into liquid nitrogen for 2 days before thawing rapidly in a 37°C water bath. The cells were added to 10 milliliterGrowth medium and centrifuged before the cell number and viability was estimated as before. Cells were seeded onto gelatin-coated flasks at 5,000 cells/cm2 to determine whether the cells would attach and proliferate.
Resrclts The initial viability of the cells to be cryopreserved was assessed by trypan blue staining to be 100%.
There was a conunensurate reduction in cell number with viability for C6?95 due to cells lysis. The viable cells cryopreserved in all four solutions attached, divided, and produced a confluent monolayer within 3 days. There was no discernable difference in estimated growth rate.
Summary. The cryopreservation of cells is one procedure available for preparation of a cell bank or a cell product. Four cryopreservation mixtures were compared for their ability to protect human placenta-derived cells from freezing damage. Dulbecco's modified Eagle's medium (DMEM) and 10% (v/v) dimethylsulfoxide (DMSO) is the preferred medium of those compared for cryopreservation of placenta-derived cells.

Growth Characteristics of Postpartum-derived Cells The cell expansion potential of postpartum-derived cells was compared to other populations of isolated stem cells. The art of cell expansion to senescence is refewed to as Hayflick's limit (Hayflick L. The longevity of cultured human cells. J. Ana.
Geraatr. Soc.
22(1):1-12, 1974; Hayflick L. The strategy of senescence. Gerontologist 14(1):37-45), 1974).
Postpartum-derived cells are highly suited for therapeutic use because they can be readily expanded to sufficient cell numbers.
Materials afad Metlaods Gelatin-coating flasks. Tissue culture plastic flasks were coated by adding 20 milliliter 2% (w/v) porcine gelatin (Type B: 225 Bloom; Sigma, St Louis, MO) to a T75 flask (Corning, Corning, NY) for 20 minutes at room temperature. After removing the gelatin solution, 10 milliliter phosphate-buffered saline (PBS) (Invitrogen, Carlsbad, CA) were added and then aspirated.
Comparison of expansion potential of postpartum-derived cells to other cell populations. For comparison of growth expansion potential, the following cell populations were utilized: r) Mesenchymal stem cells (MSC; Cambrex, Walkersville, MD); ii) Adipose-derived cells (U.S. Patent No. 6,555,374 B1; U.S. Patent Application Publication No.
US2004/0058412);
iii) Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Carnbrex, Walkersville, MD); iv) Umbilical cord-derived cells; and v) Placenta-derived cells. Cells were initially seeded at 5,000 cells/cm'' on gelatin-coated T75 flasks in DMEM-Low glucose growth medium ((Invitrogen, Carlsbad, CA), with 15 % (v/v) defined bovine serum (Hyclone, Logan, UT;
Lot#AND18475), 0.001 % (v/v) 2-mercaptoethanol (Sigma, St. Louis, MO), 100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 nucrograms/milliliter amphotericin B;
Invitrogen, Carlsbad, CA). 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 microliters, Sigma, St. Louis MO). Viable cell numbers were estimated using a hemocytometer.
Following counting, cells were seeded at 5,000 cells/cmz onto gelatin-coated T
75 flasks in 25 milliliter of fresh Growth medium. Cells were grown under standard atmosphere with 5 %
carbon dioxide at 37 °C. 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 [1n (cell final/cell initial)/ln 2] and doubling time (time in culture (h)/population doubling) were calculated. For the purposes of determining optimal cell expansion, the total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor = cell final/cell initial).
Expansion potential of cell banks at low density. The expansion potential of cells banked at passage 10 was also tested. A different set of conditions was used.
Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, MD), umbilical cord-derived cells, and placenta-derived cells were tested. These cell populations had been banked at passage previously, having been seeded at 5,000 cells/cm2 and grown to confluence 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 conditons, counted using Trypan Blue staining. Thawed cells were then seeded at 1,000 cells/cm2 in Growth medium (DMEM-Low glucose (Invitrogen, Carlsbad, CA) with 15 percent (v/v) defined bovine serum (Hyclone, Logan, UT; Lot#AND18475), 0.001 percent 2-rnercaptoethanol ( Sigma, St. Louis, MO), antibiotic/antimycotic (100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphoteiicin B (Invitrogen, Carlsbad, CA)). 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 (1n (cell final/cell initial)/ln2) and doubling time (time in culture (h)lpopulation doubling) were calculated. The total cell yield per passage was determined by multiplying total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor = cell final/cell initial).
Expansion of postpartum-derived cells at low density from initial cell seeding. The expansion potential of freshly isolated postpartum-derived cell cultures under low cell seeding conditions was tested in another experiment. Umbilical cord- and placenta-derived cells were isolated as described herein. Cells were seeded at 1000 cells/cm' and passaged as described above until senescence. Cells were grown under standard atmospheric conditions at 37°C.
Growth medium was changed twice per week. Cells were passaged as they reached about S5%
confluence. At each passage, cells were trypsinized and counted by Trypan Blue staining. The cell yield, population doubling (1n (cell final/cell initial)/ln 2), and doubling time (time in culture (h)/population doubling) were calculated for each passage. The total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor= cell final/cell initial). Cells were grown on gelatin- and non-gelatin-coated flasks.
Expansion of Clonal Neonatal or Maternal Placenta-derived Cells. Cloning may be used in order to expand a population of neonatal or maternal cells successfully from placental tissue. Following isolation of three different cell populations from the placenta (neonatal aspect, maternal aspect, and vinous region), these cell populations are expanded under standard growth conditions and then karyotyped to reveal the identity of the isolated cell populations. By isolating the cells from a mother who delivers a boy, it is possible to distinguish between the male and female chromosomes by performing metaphase spreads. These experiments can be used to demonstrate that top-line cells are karyotype positive for neonatal phenotype, mid-line cells are karyotype positive for both neonatal and maternal phenotypes, and bottom-line cells are karyotype positive for maternal cells.
Expansion of cells in low oxygen culture conditions. It has been demonstrated that low 02 cell culture conditions can improve cell expansion in certain circumstances (Csete, Marie;
Doyle, John; Wold, Barbara J.; McKay, Ron; Studer, Lorenz. Low oxygen culturing of central nervous system progenitor cells. US20040005704). In order to determine if cell expansion of postpatrtum-derived cells could be improved by altering cell culture conditions, cultures of umbilical cord-derived cells were grown in low oxygen conditions. Cells were seeded at 5,000 cells/cm' 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.
Evaluation of other growth conditions. In other experiments, postpartum-derived cells were expanded on non-coated, collagen-coated, fibronectin-coated, laminin-coated, and extracellular membrane protein (e.g.,MATRIGEL (BD Discovery Labware, Bedford, MA))-coated plates. Cultures have been demonstrated to expand well on these different matrices.
Results Comparison of expansion potential of postpartum-derived cells vs. other stem cell and non-stem cell populations. Both umbilical cord-derived and placenta-derived cells expanded for greater than 40 passages generating cell yields of > 1E17 cells in 60 days. In contrast, MSCs and fibroblasts senesced after < 25 days and <60 days, respectively. Although both adipose-derived and omental cells expanded for almost 60 days, they generated total cell yields of 4.5E12 and 4.24E13 respectively. Thus, when seeded at 5,000 cells/cm' under the experimental conditions utilized, postpartum-derived cells expanded much better than the other cell types grown under the same conditions (Table 5-1).
Expansion of potential of cell banks at low density. Umbilical cord-derived, placenta-derived, and fibroblast cells expanded for greater than 10 passages generating cell yields of >
1E11 cells in 60 days (Table 5-2). After 60 days under these conditions, the fibroblasts became senescent, whereas the umbilical cord-derived and placenta-derived cell populations senesced after 80 days, completing >50 and > 40 population doublings, respectively.
Expansion of postpartum-derived cells at low density from initial cell seeding.
Postpartum-derived cells were seeded at low density (1,000 cells/cm2) on gelatin-coated and uncoated plates or flasks. Growth potential of these cells under these conditions was good. The cells expanded readily in a log phase growth. The rate of cell expansion was similar to that observed when postpartum-derived cells were seeded at 5,000 cells/cm' on gelatin-coated flasks in Growth medium. No differences were observed in cell expansion potential between culturing on either uncoated flasks or gelatin-coated flasks. However, cells appeared phenotypically much smaller on gelatin-coated flasks, and more, larger cell phenotypes were observed on uncoated flasks.
Expansion of Clonal Neonatal or Maternal Placenta-Derived Cells. A clonal neonatal or maternal cell population can be expanded from placenta-derived cells isolated from the neonatal aspect or the maternal aspect, respectively, of the placenta. Cells 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 performed to ensure that a clonal population of cells has been identified. For subcloning experiments, cells are trypsinized and reseeded at 0.5 cells/well. The subclones that grow well are expanded in gelatin-coated T25 flasks at 5,000 cells cm' /flask . Cells are passaged at 5,000 cells cm' /T75 flask.
The growth characteristics of a clone may be plotted to demonstrate cell expansion.
Karyotyping analysis can confirm that the clone is either neonatal or maternal.
Expansion of cells in low oxygen culture conditions. Postpartum-derived cells expanded well under the reduced oxygen conditions. Culturing under low oxygen conditions does not appear to have a significant effect on cell expansion for postpartum-derived cells.
Standard atmospheric conditions have already proven successful for growing sufficient numbers of cells, and low oxygen culture is not required for the growth of postpartum-derived 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-derived cells can be expanded in culture for such purposes. Comparisons were made of the growth of postpartum-derived cells in culture to that of other cell populations including mesenchymal stem cells. The data demonstrated that postpartum-derived cell lines as developed herein can expand for greater than 40 doublings to provide sufficient cell numbers, for example, for pre-clinical banks. Furthermore, these postpartum-derived cell populations can be expanded well at low or high density. This study has demonstrated that mesenchymal stem cells, in contrast, cannot be expanded to obtain large quantities of cells.
The current cell expansion conditions of growing isolated postpartum-derived cells at densities of about 5,000 cells/cm' 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 suggests that the cells can be readily expanded using lower density culture conditions (e.g. 1,000 cells/cm2). Postpartum-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for growth. Presently, culturing postpartum-derived cells under standard atmospheric conditions is prefewed for generating large pools of cells. However, when the culture conditions are altered, postpartum-derived cell expansion can likewise. be altered. This strategy may be used to enhance the proliferative and differentiative capacity of these cell populations.
Under the conditions utilized, while the expansion potential of MSC and adipose-derived cells is limited, postpa~~tum-derived cells expand readily to large numbers.
References 1) Hayflick L. The longevity of cultured human cells. JA~ra Ger-iatn Soc. 1974 Jan;22(1):1-12.
2) Hayflick L. The strategy of senescence. Gerontologist. 1974 Feb;l4(1):37-45.
3) Patent US20040058412 4) Patent US20040048372 6) Csete, Marie; (Ann Arbor, MI) ; Doyle, John; (South Pasadena, CA) ; Wold, Barbara J.; (San Marino, CA) ; McKay, Ron; (Bethesda, MD) ; Studer, Lorenz; (New York, NY). Low oxygen culturing of central nervous system progenitor cells. US20040005704.
Table 5-1: Growth characteristics for different cell populations grown to senescence Cell Type Senescence Total PopulationTotal Cell Doublings Yield MSC 24 days 8 4.72 E7 Adipose- 57 days 24 4.5 E12 derived cells (Artecel, U.S.

Patent No.

6,555,374) Fibroblasts 53 days 26 2.S2 E13 Umbilical 65 days 42 6.15 E17 cord-derived cells Placenta- 80 days 46 2.49 E19 derived cells Table 5-2: Growth characteristics for different cell populations using low density growth expansion from passage 10 to senescence Cell Type SenescenceTotal PopulationTotal Cell , Doublings Yield Fibroblast 80 days 43.68 2.59 E11 (P10) Umbilical 80 days 53.6 1.25 E14 cord-derived cells (P 10) Placenta-derived60 days 32.96 6.09 E12 cells (P10) Karyotype analysis of PPDCs Cell lines used in cell therapy are preferably homogeneous and free from anyy contaminating cell type. Human cells used in cell therapy should have a normal chromosome number (46) and structure. To identify postpartum-derived placental and umbilical cord cell lines that are homogeneous and free from cells of non-postpartum tissue origin, karyotypes of cell samples were analyzed.
Materials and Methods PPDCs from postpartum tissue of a male neonate were cultured in Growth medium (DMEM-low glucose (Gibco Carlsbad, CA), 15% (v/v) fetal bovine serum (FBS) (Hyclone, Logan, UT), 0.001% (v1v) betamercaptoethanol (Sigma, St. Louis, MO), and 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco, Carlsbad, CA)).
Postpartum tissue from a male neonate (X,Y) was selected to allow distinction between neonatal-derived cells and maternal-derived cells (X,X). Cells were seeded at 5,000 cells per square centimeter in Growth medium in a T25 flask (Corning, Corning, NY) and expanded to about 80%
confluence. A T25 flask containing cells was filled to the neck with Growth medium. Samples were delivered to a clinical cytogenetics lab by courier (estimated lab to lab transport time is one hour).
Chromosome analysis was performed by the Center for Human & Molecular Genetics at the New Jersey Medical School, Newark, NJ. Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two) . A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells were counted and analyzed when a heterogeneous karyotype number (four) was identified.
Results All cell samples sent for chromosome analysis were interpreted by the cytogenetics laboratory staff as exhibiting a normal appearance. Three of the sixteen cell lines analyzed exhibited a heterogeneous phenotype (XX and XY) indicating the presence of cells derived from both neonatal and maternal origins (Table 6-1). 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 ( ' ' XY), indicating a previously undetected presence of cells of maternal origin.
Table 6-1. Karyotype results of PPDCs.
Metaphase Metaphase Number of cells cells Tissue passagecounted analyzed karyotypes ISCN Karyotype Placenta22 20 5 2 46 , XX

Umbilical23 20 5 2 46, XX

Umbilical6 20 5 2 46, XY

Placenta 2 20 5 2 46, XX

Umbilical3 20 5 2 46, XX

Placenta-N0 20 5 2 46, XY

Placenta-V0 20 5 2 46, XY

46, XY[18]/46, Placenta-M0 21 5 4 XX[3]

Placenta-M4 20 5 ~ 2 46,XX

46, XY[5]/46, Placenta-N9 ~ 25 5 4 XX[20]

Placenta-N
C1 1 20 5 2 46, XY

Placenta-N
1 6 46, XY[2]/46, C3 0 ' XX[18]

Placenta-N
C4 1 20 5 2 46, XY

Placenta-N
C 15 1 20 5 2 46, XY

Placenta-N
C20 1 20 5 2 46, XY

Placenta-N
C22 1 I 20 I 5 I 2 I 46, XY

Key: N- Neonatal side; V- vinous region; M- maternal side; (:- clone Summary. Chromosome analysis identified placenta- and umbilical cord-derived PPDCs whose karyotypes appear normal as interpreted by a clinical cytogenetic laboratory.
Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.

Evaluation of Human Postpartum-derived Cell Surface Markers by Flow Cytometry 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-derived cell lines isolated from the placenta and umbilical cord were characterized by flow cytometry, thereby providing a profile for the identification of the cells of the invention.
Materials arzd Met)zods Media. Cells were cultured in DMEM-low glucose Growth medium (Gibco Carlsbad, CA), with 15% (vlv) fetal bovine serum (FBS); (Hyclone, Logan, UT), 0.001%
(v/v) betamercaptoethanol (Sigma, St. Louis, MO), and 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco, Carlsbad, CA).

Culture Vessels. Cells were cultured in plasma-treated T75, T150, and T225 tissue culture flasks (Corning, Corning, NY) until confluent. The growth surfaces of the flasks were coated with gelatin by incubating 2% (w/v) gelatin (Sigma, St. Louis, MO) for 20 minutes at room temperature.
Antibody Staining. Adherent cells in flasks were washed in phosphate buffered saline (PBS); (Gibco, Carlsbad, CA) and detached with Trypsin/EDTA (Gibco, Carlsbad, CA). Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1x10' per nulliliter. In accordance with the manufacturer's specifications, antibody to the cell surface marker of interest (Table 7-1) 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 and centrifuged to remove unbound antibody. Cells were resuspended in 500 microliter PBS and analyzed by flow cytometry.
Flow Cytometry Analysis. Flow cytometry analysis was performed with a FACScalibur instrument (Becton Dickinson, San Jose, CA).
Antibodies to Cell Surface Markers. The following antibodies to cell surface markers were used.
Table 7-1. Antibodies to Cell Surface markers Antibody Manufacture Catalog Number CD10 BD Pharmingen (San Diego, CA) 555375 CD13 BD Pharmingen (San Diego, CA) 555394 CD31 BD Pharmingen (San Die o, CA) 555446 CD34 BD Pharmingen (San Diego, CA) 555821 CD44 BD Pharmingen (San Diego, CA) 555478 CD45RA BD Pharmingen (San Diego, CA) 555489 CD73 BD Pharmingen (San Diego, CA) 550257 CD90 BD Pharminaen (San Diego, CA) 555596 CD117 BD Biosciences (San Jose, CA) 340529 CD141 BD Pharminaen (San Die o, CA) 559781 PDGFr-alphaBD Pharnungen (San Diego, CA) 556002 HLA-A, B, BD Pharmingen (San Diego, CA) 555553 C

HLA-DR, DP, BD Pha~~mingen (San Diego, CA) 555558 DQ

I G-FITC Sigma (St. Louis, MO) F-6522 IgG- PE Sigma (St. Louis, MO) P-4685 Placenta- and Umbilical Cord-Derived Cell Comparison. Placenta-derived cells were compared to umbilical cord-derived cells at passage 8.
Passage to Passage Comparison. Placenta- and umbilical cord cells were analyzed at passages 8, 15, and 20.

Donor to Donor Comparison. To compare differences among donors, placenta-derived cells from different donors were compared to each other, and umbilical cord-derived cells from different donors were compared to each other.
Surface Coating Comparison. Placenta-derived cells cultured on gelatin-coated flasks were compared to placenta-derived cells cultured on uncoated flasks. Umbilical cord-derived cells cultured on gelatin-coated flasks were compared to umbilical cord-derived cells cultured on uncoated flasks.
Digestion Enzyme Comparison. Four treatments used for isolation and preparation of cells were compared. Cells derived from postpartum tissue by treatment with 1) collagenase; 2) collagenase/dispase; 3) collagenase/hyaluronidase; and 4) collagenase/hyaluronidase/dispase were compared.
Placental Layer Comparison. Cells isolated from the maternal aspect of placental tissue were compared to cells isolated from the villous region of placental tissue and cells isolated from the neonatal fetal aspect of placenta.
Resrclts Placenta-derived cells were compared to Umbilical cord-derived cells. Placenta-and umbilical cord-derived cells analyzed by flow cytometry showed positive for production of CD10, CD13, CD44, CD73, CD 90, 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 fluorescence values of positive curves was accounted. The mean (i.e., CD13) and range (i.e., CD90) of the positive curves showed some variation, but the curves appeared normal, confirming a homogeneous population.
Both curves individually exhibited values greater than the IgG control.
Passage to Passage Comparison of Placenta-derived cells. Placenta-derived cells at passages 8, 15, and 20 analyzed by flow cytometry all were positive for production of CD10, CD13, CD44, CD73, CD 90, 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 production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ having fluorescence values consistent with the IgG control.
Passage to Passage Comparison of Umbilical cord-derived cells. Umbilical cord-derived cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG
control.
Donor to Donor Comparison of Placenta-derived cells. Placenta-derived cells isolated from separate donors analyzed by flow cytometry each expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HI.A-A, B, C, with increased values of fluorescence relative to the IgG
control. The cells were negative for production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence value consistent with the IgG
control.
Donor to Donor Comparison of Umbilical cord-derived cells. Umbilical cord-derived cells isolated from separate donors analyzed by flow cytometry each showed positive for production of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control.
The Effect of Surface Coating with Gelatin on Placenta-derived Cells. Placenta-derived cells expanded on either gelatin-coated or uncoated flasks analyzed by flow cytometry all expressed of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the. IgG control. These cells were negative for production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ indicated by fluorescence values consistent with the IgG control.
The Effect of Surface Coating with Gelatin on Umbilical cord-derived Cells.
Umbilical cord-derived cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for production of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG
control. These cells were negative for production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
Evaluation of Effect of Enzyme Digestion Procedure Used for Preparation and Isolation of the Cells on the Cell Surface Marker Profile. Placenta-derived cells isolated using various digestion enzymes analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, 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 production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control.
Placental Layer Comparison . Cells derived from the maternal, villous, and neonatal layers of the placenta, respectively, analyzed by flow cytometry showed positive for production 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 production of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control.
Summary. Analysis of placenta- and umbilical cord-derived postpartum cells by flow cytometry has established of an identity of these cell lines. Placenta- and umbilical cord-derived postpartum cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A,B,C
and negative for CD31, CD34, GD45, 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 homogeneous population which has positive expression of the markers.

Analysis of Postpartum Tissue-Derived Cells by Affymetrix GeneChip~ Arrays Affymetrix GeneChip~ arrays were used to compare gene expression profiles of umbilical cord- 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 a~ad Methods Isolation and Culture of Cells Postpar-tuni tissLre-derived cells. Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDRI, Philadelphia, PA) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 1. Cells were cultured in Growth medium (Dulbecco's Modified Essential Media (DMEM-low glucose; Invitrogen, Carlsbad, CA) with 15% (v/v) fetal bovine serum (Hyclone, Logan UT), 100 Units/milliliter penicillin, 100 micrograms/milliliter streptomycin (Invitrogen, Carlsbad, CA), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis MO)) on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37°C in standard atmosphere.
Fibroblasts. Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, MD; Lot number 9F0844) and were obtained from ATCC CRL-1501 _77_ (CCD39SK). Both lines were cultured in DMEMlFI2 medium (Invitrogen, Carlsbad, CA) with
10% (v/v) fetal bovine serum (Hyclone) and 100 Units/milliliter penicillin, micrograms/milliliter streptomycin (Invitrogen). The cells were grown on standard tissue-treated plastic.
Haerrrarz Meserzclzyrzzal Sterrz. 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% COZ.
Hrcnzarz Ileac Crest Bo~ae Marro~a~ 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 micromolar NH4Cl, 10 micromolar KHC03, and 0.1 micromolar EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500 x g. The supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10 % (v/v) fetal bovine serum and 4 micromolar glutamine.
The cells were centrifuged again and the cell pellet was resuspended in fresh medium. The viable mononuclear cells were counted using trypan-blue exclusion (Sigma, St. Louis, MO). The mononuclear cells were seeded in tissue-cultured plastic flasks at 5 x 104 cells/cm'. The cells were incubated at 37°C with 5% COZ at either standard atmospheric 02 or at 5% 02. Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture.
The adherent cells were maintained in culture.
Isolation of mRNA and Gene Chip Analysis. Actively growing cultures of cells were removed from the flasks with a cell scraper in cold phosphate buffered saline (PBS). The cells were centrifuged for 5 minutes at 300 x g. The supernatant was removed and the cells were resuspended in fresh PBS and centrifuged again. The supeznatant 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 HG-U133A GENECHIP oligonucleotide array (Affymetrix, Santa Clara CA). The hyybridization and data collection was performed according to the manufacturer's specifications. Analyses were performed using "Significance Analysis of Microarrays" (SAM) version 1.21 computer software (Stanford University, www-stat.stanford.edu/~tibs/SAM; Tusher, V.G. et al., 2001, Proc. Natl. Acad. Sci.
USA 98: 5116-5121).
_78_ 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 8-1.
Table 8-1. Cells analyzed by the microatray study. The cells lines are listed by their identification code along with passage at the time of analysis, cell Growth substrate. and growth media.
Cell Population Passage Substrate Media Umbilical (022803)2 Gelatin DMEM, 15% FBS, BME

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

Umbilical (071003)4 Gelatin DMEM, IS% FBS, BME

Placenta (042203)12 Gelatin DMEM, 15% FBS, BME

Placenta (042903)4 Gelatin DMEM, 15% FBS, BME

Placenta (071003)3 Gelatin DMEM, IS% FBS, BME

ICBM (070203) 3 Plastic ME M 10% FBS
(5%
Oz) ICBM (062703) 5 Plastic MEM 10% FBS
(std Oz) ICBM (062703 5 Plastic MEM 10% FBS
)(5%
~2) hMSC (Lot 2F1655)3 Plastic MSCGM

hMSC (Lot 2F1656)3 Plastic MSCGM

hMSC (Lot 2F1657)3 Plastic M SCGM

hFibroblast (9F0844)9 Plastic DMEM-F12, 10 % FBS

hFibroblast (ATCC4 Plastic DMEM-F12, 10 CIZL-1501) % FBS

The data were evaluated by a Principle Component Analysis, analyzing the 290 genes that were differentially expressed in the cells. This analysis allows for a relative comparison for the similatxties between the populations. Table 8-2 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 8-2. The Euclidean Distances for the Cell Pairs. The Euclidean distance was calculated for the cell types using the 290 genes that were differentially expressed between the cell types.

Similarity between the cells is inversely proportional to the Euclidean distance.
Cell Pair ~ Euclidean Distance ICBM-hMSC 24.71 Placenta-umbilical 25.52 ICBM-Fibroblast 36.44 ICBM- lacenta 37.09 Fibroblast-MSC 39.63 ICBM-Umbilical 40.15 Fibroblast- 41.59 Umbilical MSC-Placenta 42. S4 MSC-Umbilical 46.86 ICBM-placenta 48.41 Tables 8-3, 8-4, and 8-5 show the expression of genes increased in placenta-derived cells (Table 8-3), increased in umbilical cord-derived cells (Table 8-41, and reduced in umbilical cord-and placenta-derived cells (Table S-5). The column entitled "Probe Set )D"
refers to the manufacturer's identification code for the sets of several oligonucleotide probes located on a particular site on the chip, which hybridize to the named gene (column "Gene Name"), comprising a sequence that can be found within the NCBI (GenBank) database at the specified accession number (column "NCBI Accession Number").
Table 8-3. Genes shown to have specifically increased expression in the placenta-derived cells as compared to the other cell lines assayed.
Genes Increased in Placenta-Derived Cells Probe Gene Name NCBI Accession Number Set ID

209732_atC-type (calcium dependent, carbohydrate-AF070642 recognition domain) lectin, supertamily member 2 activation-induced 206067_s_atWilms tumor 1 NM 024426 207016_s_ataldehyde dehydrogenase 1 family,AB015228 member A2 206367_atrenin NM 000537 210004_atoxidised low density lipoproteinAF035776 (lectin-like) receptor 214993_atHomo sapiens, clone IMAGE:4179671,AF070642 mRNA, partial cds 202178_atprotein kinase C, zeta NM 002744 209780_athypothetical protein DKFZp564F013AL136883 204135_atdownregulated in ovarian cancerNM 014890 213542_atHomo sapiens mRNA; cDNA DKFZp547K1113AI246730 (from clone DKFZp547K1113) Table 8-4. Genes shown to have specifically increased expression in umbilical cord -derived cells as compared to the other cell lines assayed.
Genes Increased in Umbilical Cord-Derived Cells Probe Gene Name NCBI Accession Number Set ID

202859 interleukin 8 NM 000584 x at 211506 interleukin 8 AF043337 s at 210222 reticulon 1 BC000314 s at 204470_atchemokine (C-X-C motif) ligandNM 001511 1 (melanoma growth stimulating activity 206336_atchemokine (C-X-C motif) ligandNM 002993 6 (granulocyte chemotactic protein 2) 207850_atchemokine (C-X-C motif) ligandNM 002090 203485 reticulon 1 NM 021136 at 202644_s_attumor necrosis factor, alpha-inducedNM_006290 protein 3 Table 8-5. Genes that were shown to have decreased expression in the umbilical cord- and placenta-derived cells as compared to the other cell lines assayed.
Genes Decreased in Umbilical Cord-and Placenta-Derived Cells Probe Gene name NCBI Accession Number Set ID

210135_s_atshort stature homeobox 2 AF022654.1 205824_atheat shock 27kDa protein 2 NM 001541.1 209687_atchemokine (C-X-C motif) ligand019495.1 12 (stromal cell-derived factor 1 ) 203666_atchemokine (C-X-C motif) ligandNM 000609.1 12 (stromal cell-derived factor 1 ) 212670_atelastin (supravalvular aortic AA479278 stenosis, Williams-Beuren syndrome) 213381 Homo Sapiens mRNA; cDNA DKFZp586M2022N91149 at (from clone DKFZp586M2022) 206201 mesenchyme homeobox 2 (growth NM 005924.1 s at arrest-specific homeobox) 205817_atsine oculis homeobox homolog NM_005982.1 1 (Drosophila) 209283_atcrystallin, alpha B AF007162.1 212793 dishevelled associated activatorBF513244 at of morphogenesis 2 213488_atDKFZP586B2420 protein AL050143.1 209763 similar to neuralin 1 AL049176 at 205200_attetranectin (plasminogen bindingNM 003278.1 protein) 205743_atsrc homology three (SH3) and NM 003149.1 cysteine rich domain 200921 B-cell translocation gene 1, NM 001731.1 s at anti-proliferative 206932 cholesterol25-hydroxylase NM 003956.1 at 204198_s_atrunt-related transcription AA541630 factor 3 219747_athypothetical protein FLJ23191 NM 024574.1 204773_atinterleukin 11 receptor, alphaNM 004512.1 202465_atprocollagen C-endopeptidase NM 002593.2 enhancer 203706 frizzled homolog 7 (Drosophila)NM 003507.1 s at 212736_athypothetical gene BC008967 BE299456 214587_atcollagen, type VIII, alpha BE877796 201645_attenascin C (hexabrachion) NM 002160.1 210239 Iroquois homeobox protein 5 090304.1 at 203903_s_athephaestin NM 014799.1 205816_atintegrin, beta 8 NM_002214.1 203069_atsynaptic vesicle glycoprotein NM 014849.1 213909 Homo sapiens cDNA FLJ12280 AU147799 at fis, clone 206315_atcytokine receptor-like factor NM 004750.1 204401 potassium intermediate/small NM 002250.1 at conductance calcium-activated channel, subfamily N, member 216331 integrin, alpha 7 AK022548.1 at 209663_s_atintegrin, alpha 7 AF072132.1 213125 DKFZP586L151 protein AW007573 at 202133_attranscriptional co-activator AA081084 with PDZ-binding motif (TAZ) 206511_s_atsine oculis homeobox homolog NM 016932.1 2 (Drosophila) 213435_atKIAA1034 protein AB028957.1 206115_atearly growth response 3 NM 004430.1 213707 distal-less homeobox 5 NM 005221.3 s at 218181 hypothetical protein FLJ20373 NM 017792.1 s at 209160_ataldo-keto reductase family AB018580.1 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II) 213905 biglycan AA845258 x_at 201261 biglycan BC002416.1 _x_at 202132_attranscriptional co-activator AA081084 with PDZ-binding motif (TAZ) 214701 fibronectin 1 AJ276395.1 s at 213791 proenkephalin NM 006211.1 at 205422_s_atintegrin, beta-like 1 (with NM 004791.1 EGF-like repeat domains) 214927_atHomo sapiens mRNA full length AL359052.1 insert cDNA
clone EUROIMAGE 1968422 206070_s_atEphA3 AF213459.1 212805_atKIAA0367 protein AB002365.1 219789_atnatriuretic peptide receptor A1628360 C/guanylate cyclase C (atrionatriuretic peptide receptor C) 219054_athypothetical protein FLJ14054 NM 024563.1 213429_atHomo sapiens mRNA; cDNA DKFZp564B222AW025579 (from clone DKFZp564B222) 204929_s_atvesicle-associated membrane NM 006634.1 protein 5 (myobrevin) 201843_s_atEGF-containing fibulin-like NM 004105.2 .
extracellular matrix protein 1 ~ ' 221478_atBCL2/adenovirus E1 B l9kDa interactingAL132665.1 protein 3-like 201792_atAE binding protein 1 NM 001129.2 204570_atcytochrome c oxidase subunit NM 001864.1 Vlla polypeptide 1 (muscle) 201621 neuroblastoma, suppression of NM 005380.1 at tumorigenicity 1 202718_atinsulin-like growth factor bindingNM 000597.1 protein 2, 36kDa Tables 8-6, 8-7, and 8-S show the expression of genes increased in human fibroblasts (Table 8-6), ICBM cells (Table 8-7), and MSCs (Table 8-8).
Table 8-6. Genes that were shown to have increased expression in fibroblasts as compared to the other cell lines assayed.
Genes increased in fibroblasts dual specificity phosphatase ~3.
hIAA0527 rotein Homo sa iens cDNA: FLJ23224 fis, clone ADSU02206 dynein, cyto lasmic, intermediate olype tide 1 ankyrin 3, node of Ranvier (ankyrin G) inhibin, beta A (activin A, activin AB al ha olype tide) ectonucleotide pyrophosphatase/phosphodiesterase 4 (putative function) KIAA1053 rotein microtubule-associated rotein 1A
zinc fin er rotein 41 HSPC019 rotein Homo sa iens cDNA: FLJ23564 fis, clone LNG10773 Homo sapiens mRNA; cDNA DKFZp564A072 (from clone DKFZp564A072) LIM rotein (similar to rat rotein kinase C-bindin eni ma) _g4_ inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase com lex-associated ip-otein by othetical rotein FLJ22004 Human (clone CTG-A4) mRNA se uence ESTs, Moderately similar to cytokine receptor-like factor 2;
cytokine rece for CRL2 recursor [Homo sa iens]
transformin growth factor, beta 2 by othetical rotein MGC29643 antigen identified by monoclonal antibody MRC OX-2 Table 8-7. Genes that were shown to have increased expression in the ICBM-derived cells as compared to the other cell lines assayed.
Genes Increased In ICBM Cells cardiac ankyrin re eat rotein MHC class I region ORF
integrin, al ha 10 by othetical rotein 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 rotein 1-1 hip ocalcin-like 1 jagged 1 (Alagille syndrome) proteogl can 1, secretory granule Table S-S. Genes that were shown to have increased 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 rece for subfamily 4, rou A, member 2 v-fos FBJ munne osteoswcoma viral onco ene homolo by othetical rotein DC42 nuclear rece for subfamily 4, grou A, member 2 FBJ murine osteosarcoma viral oncogene homolog B
WNT1 inducible signalin athway protein 1 MCF.2 cell line derived transforming se uence otassium channel, subfamily K, member 15 cartilage aired-class homeo rotein 1 Homo sa iens cDNA FLJ12232 fis, clone MAMMA1001206 Homo sapiens cDNA FLJ34668 fis, clone LIVER2000775 jun B roto-oncoaene B-cell CLL/lym homa 6 (zinc finger rotein 51) zinc fin er rotein 36, C3H ty e, homolo (mouse) Summary. The GENECHil' analysis was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas. The study also included two different lines of dermal fibroblasts, tlu-ee lines of mesenchymal stem cells, and three lines of ileac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed by AffyMetrix GENECHIP that contained oligonucleotide probes for 2,000 genes.
Results showed that 290 genes are differentially expressed in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells.
Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord.
The expression of selected genes has been confirmed by PCR in Example 9. 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.
Reference Lockhart et al., Expression monitoring by hybridization to high-density oligonucleotide arrays. Nat. Biotech~zol. 1996, 14(13):1675-1680.

Cell Markers in Postpartum-derived Cells 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 Affymetnx GENECHIP array). Six "signature" genes were identified: oxidized LDL receptor 1, interleukin-8, renin, reticulon, chemokine 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 to identify accordance/discordance 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 neonatal as identified by karyotyping analysis), umbilical cord-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) were grown in Growth medium (DMEM-low glucose (Gibco, Carlsbad, CA), 15% (v/v) fetal bovine serum (Cat.
#SH30070.03;
Hyclone, Logan, UT), 0.001% (v/v) beta-mercaptoethanol (Sigma, St. Louis, MO), Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco, Carlsbad, CA) in a gelatin-coated T75 flask. Mesenchymal Stem Cells (MSCs) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, MD).
For the IL-8 secretion 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 an additional 8 hours in 10 milliliter of serum starvation medium (DMEM-low glucose (Gibco, Carlsbad, CA), 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco, Carlsbad, CA), and 0.1% (w/v) Bovine Serum Albumin (BSA; Sigma, St.
Louis, MO)).
After this treatment, RNA was extracted and the supernatants were centrifuged at 150 x g for 5 minutes to remove cellular debris. Supernatants were then frozen at -80°C for ELISA analysis.
Cell culture for ELISA assay. Postpartum cells derived from placenta and umbilical cord, as well as human fibroblasts derived from human neonatal foreskin, were cultured in Growth medium in gelatin-coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen.
Cells were thawed and transferred to 15 milliliter centrifuge tubes. After centrifugation at 150 x g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 milliliter culture medium and counted. Cells were grown in a 75 cm2 flask containing 15 milliliter of Growth medium at 375,000 cell/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation medium was collected at the end of incubation, centrifuged at 14,000 x g for 5 minutes, and stored at -20 °C.
To estimate the number of cells in each flask, 2 milliliter of tyrpsin/EDTA
(Gibco, Carlsbad, CA) was added to each flask. After cells detached from the flask, trypsin activity was neutralized with 8 milliliter of Growth medium. Cells were transferred to a 15 milliliter 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 manufacturer.
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 microliter 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 microliter DEPC-treated water and stored at -80°C. RNA was also extracted from human placenta and umbilical cord. Tissue (30 milligram) was suspended in 700 microliter 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 microliter of DEPC-treated water and stored at -80°C.
Reverse transcription. 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 ~0°C.
Genes identified by cDNA microarray as uniquely regulated in postpartum-derived 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-DEMAND
gene expression products: oxidized LDL receptor (Hs00234028); renin (Hs00166915); reticulon (Hs00382515); CXC ligand 3 (Hs00171061); GCP-2 (Hs00605742); IL-8 (Hs00174103); and GAPDH 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 was analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).
Conventional PCR. Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, MA) to confirm the results from real-time PCR.
PCR was peuormed using 2 microliter of cDNA solution, lx TAQ polymerase (tradename 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 renin (94°C for 15 seconds, 53°C
for 15 seconds and 72°C for 30 seconds for 3S 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 1. Primer concentration in the final PCR reaction was 1 micromolar except for GAPDH
_8g_ which was 0.5 micromolar. 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 mn 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 POLAROID camera (VWR International, South Plainfield, NJ).
Table 9-1: Primers used Primer name Primers Oxidized LDL receptorS 5 -GAGAAATCCAAAGAGCAAATGG-3( ID NO
: ' ' SEQ :

) A: 5'-AGAATGGAAAACTGGAATAGG-3'(SEQID N0:2) Renin S: 5'-TCTTCGATGCTTCGGATTCC-3'(SEQID N0:3) A: 5'-GAATTCTCGGAATCTCTGTTG-3'(SEQID N0:4) Reticulon S: 5'- TTACAAGCAGTGCAGAAAACC-3'(SEQID N0:5) A: 5'- AGTAAACATTGAAACCACAGCC-3'(SEQID N0:6) Interleukin-8 S 5'- TCTGCAGCTCTGTGTGAAGG-3'( ID NO
: SEQ :

) A: 5'-CTTCAP.AAACTTCTCCACAACC-(SEQID N0:8) 3' Chemokine (CXC) S 5 - CCCACGCCACGCTCTCC-3 ( ID NO
ligand 3 : ' ' SEQ :

) A: 5'-TCCTGTCAGTTGGTGCTCC (SEQID N0:10) -3' Immunofluorescence. Postpartum-derived cells were fixed with cold 4% (w/v) paraformaldehyde (Sigma-Aldrich, St. Louis, MO) for 10 minutes at room temperature. One isolate each of umbilical cord- and placenta-derived cells at passage 0 (PO) (directly after isolation) and passage 11 (P11) (two isolates of Placenta-derived, two isolates of LTmbilical 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-derived 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 R1; 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 ~'.-100; Sigma, St. Louis, MO) for 30 minutes to access intracellular antigens. Where the epitope of interest was located on the cell surface (CD34, ox-LDL R1),
12 PCT/US2004/020958 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 the process.
Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. The primary antibody solutions were removed and the cultures were washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG - Texas Red (1:250; Molecular Probes, Eugene, OR) and/or goat anti-rabbit IgG - Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG - FITC (1:150, Santa Cruz Biotech). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
Following immunostaining, fluorescence was visualized using an appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY).
In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution (no 1' control). Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, CA). For triple-stained samples, each image was taken using only one emission filter at a time.
Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, CA).
Preparation of cells for FAGS analysis. Adherent cells in flasks were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, CAl 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~/milliliter. One hundred microliter aliquots were delivered to conical tubes. Cells stained for intracellular antigens were permeabilized with Peim/Wash buffer (BD Pharmingen, San Diego, CA). Antibody was added to aliquots as per manufacturer's 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 microliter of 3% FBS.
Secondary antibody was added as per manufacturer's 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 milliliterPBS
and analyzed by flow cytometry. The following antibodies were used: oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROa (555042; BD Pharmingen, Bedford, MA), Mouse IgGl kappa, (P-4685 and M-5284; Sigma), and Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.).

FRCS analysis. Flow cytometry analysis was performed with FACScalibur (Becton Dickinson 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 analyzed by the O~CT 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-det~ived cells and controls (data not shown). CXC-ligand 3 was expressed at very low levels. GCP-2 was expressed at levels comparable to human adult and neonatal fibroblasts. 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-derived cells and controls using conventional PCR CXC ligand 3 primers listed in Table 9-1.
The expression of the cytokine IL-8 in postpartum-derived cells is elevated in both Growth medium-cultured and serum-starved postpartum-derived cells. All real-time PCR data was validated with conventional PCR and by sequencing PCR products.
When supernatants of cells grown in serum-free medium were examined for the presence of IL-8, the highest amounts were detected in media derived from umbilical cord-derived cells and some isolates of placenta-derived cells (Table 9-2). No 1L-8 was detected in medium derived from human dermal fibroblasts.
Table 9-2: 1L-8 protein expression measured by ELISA
Cell t a IL-8 ~ _ Human fibroblasts ND _ _ Placenta Isolate 1 ND

UMBC Isolate 1 205S.42+144.67 Placenta Isolate 2 ND

L1MBC Isolate 2 2365.86_+22.73 Placenta Isolate3 17.27+8.63 (normal OZ) Placenta Isolate 3 264.929.SS
(low0~, 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 resented here are icogran~/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.
Placenta-derived cells were also tested for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells derived from the human placenta were fixed with 4~1o paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Cells stained positive for both alpha-smooth muscle actin and vimentin. This pattern was preserved through passage 11. Only a few cells (<5%) at passage 0 stained positive for cytokeratin 18.
Cells derived from the human umbilical cord at passage 0 were probed for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin..
Umbilical cord-derived cells were positive for alpha-smooth muscle actin and vimentin, with the staining pattern consistent through passage 11.
Placenta-derived cells at passage 11 were also investigated by immunocytochemistry for the production of GROalpha and GCP-2. Placenta-derived cells were GCP-2 positive, but GROalpha production was not detected by this method.
The production of GROalpha, GCP-2, oxidized LDL receptor 1 and reticulon (NOGO-A) in umbilical cord-derived cells at passage 11 was investigated by immunocytochemistry.
Umbilical cord-derived cells were GCP-2 positive, but GRO alpha production was not detected by this method. Furthermore, cells were NOGO-A positive.
Summary. Accordance 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-derived cells, with IL-S also differentially regulated at the protein level. The presence of oxidized LDL receptor was not detected at the protein level by FACS analysis in cells derived from the placenta. Differential expression of GCP-2 and CXC
ligand 3 was not confirmed at the mRNA level; however, GCP-2 was detected at the protein level by FACS analysis in the placenta-derived cells. Although this result does not support data originally obtained from the microarray experiment, this may be due to a difference 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 verifies the data obtained from the microarray experiments.

Immunohistochemical Characterization of PPDC phenotype The phenotypes of cells found within human postpartum tissues, namely umbilical cord and placenta, were analyzed by immunohistochemistry.
Mate~zals & Methods Tissue Preparation. Human umbilical cord and placenta tissue were harvested and immersion fixed in 4% (w/v) paraformaldehyde overnight at 4°C.
Irmnunohistochemistry was penfonned using antibodies directed against the following epitopes (see Table 10-1): 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 Cmz, CA), anti-human oxidized LDL
receptor 1 (ox-LDL R1; 1:100; Santa Cruz Biotech), and anti-human NOGO-A
(1:100; Santa Cruz Biotech). Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, CA) on a dry ice bath containing ethanol.
Frozen blocks were then sectioned (10 micron thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
Table 10-1: Summary of Primary Antibodies Used Antibody Concentration Vendor Vimentin 1:500 Sigma, St. Louis, MO
Desmin (rb) 1:150 Sigma Desmin (m) 1:300 Chemicon, Temecula, CA

alpha-smooth muscle 1:400 Sigma actin (SMA) Cytokeratin 18 (CK18)1:400 Sigma von Willebrand factor1:200 Sigma (vWF) CD34 III 1:100 DakoCytomation, Carpinteria, CA

GROalpha-PE 1:100 BD, Franklin Lakes, NJ

GCP-2 1:100 Santa Cruz Biotech Ox-LDL R1 1:100 Santa Cmz Biotech NOGO-A 1:100 Santa Cruz Biotech Immunohistochemistry. Immunohistochemistry 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 blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3°Io (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 suuace (CD34, ox-LDL R1), triton was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL R1, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout the procedure. Primary antibodies, diluted in blocking solution, were then applied to the sections for a period of 4 hours at room temperature.
Primary antibody solutions were removed, and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG - Texas Red (1:250; Molecular Probes, Eugene, OR) and/or goat anti-rabbit IgG - Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG - FITC (1:150; Santa Cruz Biotech).
Cultures were washed, and 10 micromolar DAPI (Molecular Probes) was applied for 10 minutes to visualize cell nuclei.
Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY).
Positive staining was represented by fluorescence sib ial above control staining. 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).
Resrcdts Umbilical Cord Characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord (data not shown). In particular, vWF and CD34 expression were restricted to blood vessels contained within the cord.
CD34+ cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery & vein, but not contained with the vessels themselves.
CK18 and desrnin were observed within the vessels only, desmin being restricted to the middle and outer layers.
Placenta Characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 were all observed within the placenta and regionally specific.
GROalpha, GCP-2, ox-LDL R1, and NOGO-A Tissue Expression. None of these markers were observed within umbilical cord or placental tissue (data not shown).
Summary. Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD 34 are expressed in cells within human umbilical cord and placenta.
Based on izz vitro characterization studies showing that only vimentin and alpha-smooth muscle actin are expressed, the data suggests that the current process of postpartum-derived cell isolation harvests a subpopulation of cells or that the cells isolated change expression of markers to express vimentin and alpha-smooth muscle actin.
EXAMPLE 11:
In Vitro Immunology of Postpartum -derived cells Postpartum-derived cell lines were evaluated i~a vitro for their immunological characteristics in an effort to predict the immunological response, if any, these cells would elicit upon azz vivo transplantation. Postpartum-derived 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 a~ MOLECULAR IMNIUNOLOGIr, 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 A~ MOLECULAR IMIv~TNOLOGY, 5th Ed.
(2003) Saunders, Philadelphia, p. 171), CD 17S (Coumans, et.al., (1999) Jounzal of Inzrnzmologlcal Methods 224, 185-196), and PD-L2 (Abbas & Lichtman, CELLULAR
AND

MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171; Brown, et. al. (2003) Tlce Joiirzzal of Im~zufiology 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 i~a utero. To predict the extent to which postpartum placenta- and umbilical cord-derived cell lines elicit an immune response in nino, the cell lines were tested in a one-way mixed lymphocyte reaction (MLR).
Materials and Methods Cell culture. Cells were cultured in Growth medium (DMEM-low glucose (Gibco, Carlsbad, CA), 15% (v/v) fetal bovine serum (FBS); (Hyclone, Logan, UT), 0.001% (vlv) betamercaptoethanol (Sigma, St. Louis, MO), 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco, Carlsbad, GA)) until confluent in T75 flasks (Corning, Corning, NY) coated with 2% gelatin (Sigma, St. Louis, MO).
Antibody Staining. Cells were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, CA) and detached with Trypsin/EDTA (Gibco, Carlsbad, CA). Cells were harvested, centrifuged, and re-suspended in 3% (v/v) FBS in PBS at a cell concentration of 1x10 per milliliter. Antibody (Table 11-1) was added to one hundred microliters of cell suspension as per manufacturer's specifications and incubated in the dark for 30 minutes at 4°C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were re-suspended in five hundred microlite,rs of PBS and analyzed by flow cytometry using a FACSCalibur instrument (Becton Dickinson, San Jose, CA).
Table 11-1. Antibodies Antibod Manufacturer Catalo Number HLA-DRDPDQ BD Pharmingen (San 555558 Diego, CA) CD80 BD Pharmingen (San 557227 Diego, CA) CD86 BD Phaimingen (San 555665 Diego, CA) B7-H2 BD Pharmingen (San 552502 Diego, CA) HLA-G Abcam (Cambrid eshire,ab 7904-100 UK) CD 178 Santa Cruz (San Cruz,sc-19681 CA) PD-L2 BD Pharmingen (San 557846 Diego, CA) Mouse IgG2a Siama (St. Louis, F-6522 MO) Mouse I Glka a Si ma (St. Louis, P-4685 MO) 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-derived 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-derived cells was calculated as the mean proliferation of the receiver plus mitomycin C-treated postpartum-derived 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-derived 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 11-2). 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 11-3).
Table 11-2. Mixed Lymphocyte Reaction Data - Cell Line B (Placenta) DPM for Proliferation Assay ~~ _-~~

Plate ID:
Platel _-_--_--_ .
-~

AnalyticalCulture Repllcaies numberSystem 1 I 2 ~ MeanSD CV

ProIIlerallon _79 ; 138 112.030.1226.9 baseline 119 of receiver IM03-7769Control 241 ! 175 229.349.5421.6 of 272 ~
autostirrulation (fvilorrycin C
treated autologous cells) tvtR 23971 20921 22414.71525.976.8 allogenlc ! 22352 donor IfvD3-7768 (Mtorrycin C
treated) tvLR 664 559 1090 _771.0281.2136.5 with cell Ilne (Mlomycin C
treated cell type B) SI l ; 200 (donor) SI ~ ------~ ~ -7 (cell i ;
line) Prolileratlon 206 134 262 200.764.1732.0 baseline ~J
of reoeNer IM03-7770Control 1091 524 _739.0307.3341.6 of 602 autostirrulation (fvilorrycin Gtreated aulologcus cells) _tvLR 45005 44071 44268.3660.491.5 allogenic i 43729 donor (Mtorrycin C
treated) NLR 533 25822376 _1830.31128.2481.6 wRh cell line (Mtorrycin C
healed cell type e) SI _ 221 (donor) __ SI ' ; ~ _ cell - 9 line ( ) , Proliferation __ 157 128 124.035.1728.4 baseline _ 87 of receiver _ IM03-7771Cntrol 293 ' 508 _313.0185.81_ of 138 59.4 autostirtulation ' (Mtortycin -Gtreated autologous cells) tNti 24497 31388 30077.75054.5316.8 allogenic '; 34348 donor ItuD3-7768 (Mtorrycln C
treatecn NLR 601 843 a 622.029.704.8 w ilh cell line (Mtorrycin C
healed cell type B) (donor) (cell line) ' ProIHeralion 56 98 51 68.325.8137.8 baseline ~
of receiver IM03-7772Conhol 133 120 213_ __155_.3_50.3632.4 o_f _ T
aulostlrtulalion (Mnorrycin Ctrealed autologous cells) -MJ3 14222 22168 18822.04118.7521.9 allogenic ! 20076 donor (Mtomycin C
treated NLR a a a a a _ w - a flh cell Ilne (Mtorrycin C
treated cell type B) SI ___ _ 275 (donor) __ SI a -(cell line) ' IM03-7768 _84 242 208 178.083.1 46.
_ I 6 7 ___ _Prol'rferation baseline of receiver __ (allogenic _ _ _ _ _ donor) 361 617 __ __ _ _ Control i 304 427.3166.7139.0 of aufostlrrulation (Mlortycin treated autologous cells) i ~

Cell --- ____126_,__1241_43_ __131_.010.44___8.0 line Proliferation 5 -- _-type baseline B of r_eceN_er -Control 822 107 487 794.7294.9537.1 of atriostlrrulation (Mtortycin treated autclogous cells) Plate ID: Plate 2 AnalyticalGulture Replicates numberS stem 1 2 3 MeanSD CV

Prcliferauon baseline 908 181 330 473.0384.0281.2 of receiver IM03-7773Control of autostimulauon 269 405 572 415.3151.7636.5 (Mhomycln C heated autologous cells) MLR ellogentc donor 29151 28691 28719.0418.701.5 IM03~7768 (Mttomyctn 28315 C heated) MLR with cell Ilne (Milomycin 567 732 905 734.7169.0223.0 C heated cell type B) (donor) (cell line) Proliferation baseline 893 1376 185 818.0599.0373.2 of receiver IM03-7774Control of autoshmulatlon 261 381 568 403.3154.7138.4 (Mitomycm C heated autolagous cells) MLR ellogentc donor 53101 42839 48074.35134.1810.7 IMO3-77se (Mnomyctn 48283 C heated) MLR with cell Ilne (Mitomyctn 515 789 294 532.7247.9746.6 C heated cell type e) (donor) (cell Ilne) Prouferauon baseline 1272 300 544 705.3505.6971.7 of receiver IM03-7775Control of eutostlmulatlon 232 199 484 305.0155.8951.1 (Milomycln C treated autologous cells) MLR allogenlc donor 23554 10523 21014.09479.7445.1 IMO3~7768 (Mhomycin 28965 C treated) MLR with cell Ilne (Mltomycln 768 924 563 751.7181.0524.1 C treated cell type e) (donor) (cell Ilne) Prollterauon baseline 1530 137 1046 904.3707.2278.2 of receiver IM03-7776Control of sutosllmulatlon 420 218 394 344.0109.8931.9 (MUOmycln C heated sutologous cells) MLR allogenlc donor 28893 32493 32044.02952.229.2 IM03-7768 (Mitomycln 34746 C Irealed) MLR with cell line (Mhomycln a a a a a a C heated cell type 8) (donor) SI a (cell line) _9g_ Table 11-3. 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 4-6) Mixed Lymphocyte Reaction- Umbilical cord. 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 umbilical cord-derived 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 11-4). 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 11-5).

Table 11-4. Mixed Lymphocyte Reaction Data- Cell Line A (Umbilical cord) DPM for Proliferation Assay Plate ID: Platet AnalyticalCulture Replicates number S stem 1 2 3 Mean SD CV

Prolileradon baseline 1074 4D6 391 623.7390.0762.5 of receiver IM04-2478Control of auloslimulatlon 672 510 1402 861.3475.1955.2 (Mnomycin C treated autologous teas) MAR allogenlc donor 43777 48391 43466.35D87.1211,7 IMO4-2477 (Milomycln 38231 C treatey MLR with cell line 2914 5622 4881.71721.3635.3 (Mltomycin C treated 6109 cell type A) SI (donor) 70 SI (cell 8 line) Prollleration baseline 530 5D8 527 521.711.932.3 of receiver IM04-2479Control of autostimulatlon 701 567 1111 793.0283.4335.7 (Mltomycln C treated autologous cells) MLR allogenic donor 25593 24732 24344.01481.616.1 IMO4-2477 (Mitomycin 22707 C treated) MLR with cell Ilne 5086 3932 3505.01832.2162.3 (Mitomycin C treated 1497 cell type A) SI (donor) 47 SI (cell 7 line) Prolllerallon baseline 1192 854 1330 1125.3244.9021.8 of receiver Control of autostlmulalion 2963 993 2197 2051.0993.0848.4 (Mltomycin C treated autologous cells) IM04-2480MLR ellogenic donor 25416 29721 26298.03D78.2711.7 IMO4-2477 (Mnomycln 23757 C treated) MLR with cell Ilne 2596 5076 3699.31262.3934.1 (Mnomycin C treated 3426 cell Npe A) SI (donor) 23 SI (cell 3 line) Prolilerafon baseline 695 451 555 567.0122.4421.6 of recewer IM04-2481Control of sutostimulalion 738 1252 464 818.0400.0448.9 (Mitomycin C treated autologous cells) MLR allogenic donor 13177 24885 17835.36209.5234.8 IM04-2477 (Mitomycln 15444 C treated) MAR wgh cell line (Mitomycin 4495 3671 4280.0534.9512.5 C heated cell type 4674 A) SI (donor) 31 SI cell 8 line Place ID: Plate 2 AnalyticalCulture RepIIcates number S stem 1 2 3 Mean SD CV

Prolileralion baseline 432 533 274 413.0130.5431.6 of receiver IMD4-2482Control of autostlmulation 1459 633 598 896.7487.3154.3 (Mltomycin C treated 24286 30823 28818.33933.8213.7 autologous cells) 31346 MLR anogenic donor IMO4-2477 (Milomycln C treated) MLR with cell Ilne 2762 1502 3662.32724.4674.4 (Mitomycin C heated 6723 cell Npe A) SI (donor) 70 SI (cell 9 line) IM04-2477Prolileragon baseline 312 419 349 360.054.3415.1 of receiver (allogenicConhol of autosumulaticn 567 6D4 374 515.0123.5024.0 donor) (Mitomycin heated autologous cells) Cell Prollleradon baseline 5101 3735 3936.31078.1927.4 line of receiver 2973 2882.31466.0450.9 type Control of autostlmulatlon 1924 4570 A (Mitomycln treated 2153 aulologous cells) Table 11-5. Average stimulation index of umbilical cord-derived cells and an allogeneic donor in a mixed lymphocyte reaction with five individual allogeneic receivers.
Average Stimulation Index Recipient Umbilical Cord Plate 1 (receivers 42.75 6.5 1-4) Plate 2 (receiver 70 9 5) Antigen Presenting Cell Markers - Placenta. Histograms of placenta-derived cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CDS6, and B7-H2, as noted by fluorescence value consistent with the IgG control, indicating that placenta-derived cell lines lack the cell surface molecules required to directly stimulate allogeneic PBMCs (e.g., CD4+ T cells).
Immuno-modulating Markers - Placenta-derived cells. Histograms of placenta-derived 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 (data not shown).
Antigen Presenting Cell Markers - Umbilical cord-derived cells. Histograms of umbilical cord-derived 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 cord-derived cell lines lack the cell surface molecules required to directly stimulate allogeneic PBMCs (e.g., CD4+ T cells).
Immuno-modulating Markers- Umbilical cord-derived cells. Histograms of umbilical cord-derived 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.
Summary. In the mixed lymphocyte reactions conducted with placenta-derived cell lines, the average stimulation index ranged from 1.3 to 3, and that of the allogeneic positive controls ranged from 46.25 to 27q. In the mixed lymphocyte reactions conducted with umbilical cord-derived 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. Placenta- and umbilical cord-derived cell lines were negative for the expression of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CDS6, and B7-H2, as measured by flow cytometry. Placenta- and umbilical cord-derived 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-DP, DR, DQ, CD80, CD86, and B7-H2, thereby allowing for the stimulation of allogeneic PBMCs (e.g., 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 allogeneic PBMCs (e.g., 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.

References Bruder SP et. al. USP 6,355,239 B1 (2002) Abbas, AIs, Lichtman, AH Cellular and Molecular Immunology 5th Ed. (2003) Saunders, Philadelphia, p. 171 Bouteiller P.Le et. al., (2003) Placenta 24;510-S15 Coumans B et.al., (1999) Journal of Immunological Methods 224, 185-196]
Brown, Julia et. al. (2003) The Journal of Immunology 170, 1257-1266 Secretion of trophic factors by postpartum-derived cells 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. S~~nzp. 212:215-26), monocyte chemotactic protein 1 (MCP-1) (Salcedo et al. (2000) Blood 96;34-40), interleukin-8 (IL-8) ( Li et al. (2003) J. Inznzuzzol. 170:3369-76), keratinocyte growth factor (IiGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) (Hughes et al.
(2004) Ann.
Tlzor-ac. Sung. 77:512-8), tissue inhibitor of matrix metalloproteinase 1 (TIMP1), angiopoietin 2 (ANG2), platelet derived growth factor (PDGF-bb), thrombopoie.tin (TPO), heparin-binding epidermal growth factor (HB-EGF), stromal-derived factor la (SDF-la)), neurotrophic/neuroprotective activity (brain-derived neurotrophic factor (BDNF) (Cheng et al.
(2003) Dev. Biol. 258;319-33), interleukin-6 (IL-6), granulocyte chemotactic protein-2 (GCP-2), transforming growth factor beta2 (TGFbeta2)), or chemokine activity (macrophage inflammatory protein la (MIPla), macrophage inflammatory protein lbeta (MIPIb), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), I309, thymus and activation-regulated chemokine (TARC), Eotaxin, macrophage-derived chemokine (MDC), IL-8).
Methods & Materials Cell culture. PPDCs derived from placenta and umbilical cord as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth medium (DMEM-low glucose (Gibco, Carlsbad, CA), 15% (v/v) fetal bovine serum (SH30070.03;
Hyclone, Logan, UT), 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco)) 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 milliliter 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 milliliter Growth medium, and cells were counted. Cells were seeded at 5,000 cells/cm2 on a T75 flask containing 15 milliliter 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 semm albumin (Sigma), 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (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 -0°C. To estimate the number of cells in each flask, cells were washed with phosphate-buffered saline (PBS) and detached using 2 milliliter trypsin/BDTA (Gibco). Trypsin activity was inhibited by addition of 8 milliliter 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-1,1L-6, VEGF, SDF-1a , 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 picogram/milliliter/rnillion cells (n=2, sem).
Searchlight Multiplexed ELISA assay. Chemokines (MIPIa, MIPlb, MCP-1, Rantes, I309, TARO, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, T)IvvIPl, ANG2, PDGF-bb, TPO, HB-EGF were measured using Searchlight Proteome Arrays (Pierce Biotechnology Inc.). The Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to 16 proteins per well. The arrays are produced by spotting a 2 x 2, 3 x 3, or 4 x 4 pattern of four to 16 different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture chemiluminescent signal generated at each spot within each well of the plate.
The amount of signal generated in each spot is proportional to the amount of target protein in the original standard or sample.
Resrclts ELISA assay. MCP-1 and IL-6 were secreted by placenta- and umbilical cord-derived PPDCs and dermal fibroblasts (Table 12-1). Umbilical cord-derived cells secreted at least 10-fold higher amounts of MCP-1 and IL6 than other cell populations. GCP-2 and IL-8 were highly expressed by umbilical-derived PPDCs. TGF-beta2 was not detectable.
VEGF was detected in fibroblast medium.
The amount of HGF, FGF, and BDNF secreted from umbilical cord-derived cells were noticeably higher than fibroblasts and placenta-derived cells (Tables 12-2 and 12-3). Similarly, TnVIPl, TPO, HBEGF, MCP-1, TARC, and IL-8 were higher in umbilical cord-derived cells than other cell populations (Table 12-3). No ANG2 or PDGF-bb were detected.
Table 12-1. ELISA assay results MCP-1 1L-6 VEGF SDF-la GGP-2 IL-8 TGF-beta2 Fibroblast17+1 61+3 29+2 19+1 21+1 ND ND

Placenta60+3 41+2 ND ND ND ND ND

(042303) Umbilical1150_+744234_+289ND ND 160_+11 205S++145ND

(022803) Placenta125_+16 10+1 ND ND ND ND ND

(0710031 Umbilical2794_+841356_+43ND ND 2184_+982369_+23ND

(071003) Placenta21+10 67+3 ND ND 44+9 17+9 ND

(101503) BME

Placenta77+16 339_+21 ND 1149_+13754++~ 265_+10 ND

(101503) % O~, W/O

BME

itey: rVU: mot uetected.
Table 12-2. Searchlight Multiplexed ELISA assay results TIIuVIPIANG2 PDGFbb TPO IiGF HGF FGF VEGF HBEGF BDNF

hFB 19306.3ND ND 230.55.0 ND ND 27.9 1.3 ND

P1 24299.5ND ND 546.6S.8 16.4 ND ND 3.51.3ND

U1 5771S.4ND ND 1240.05.S 559.3 145.7ND 9.3 165.7 P3 14176.8ND ND 565.75.2 10.2 ND ND 1.9 33.6 U3 21850.0ND ND 1134.59.0 195.6 30.5 ND 5.4 358.6 lCey: tuts (human nbroblasts), Y1 (placenta-denved YY1J(: (U4L:iU:i)), U1 (umbilical cord-derived YYDC (U22503)), P3 (placenta-derived PPDC (071003)), U3 (umbilical cord-derived PPDC
(071003)). ND: Not Detected.
Table 12-3. Searchlight Multiplexed ELISA assay results MIPla MIPlb MCP1 RANTES I309 TARC EotaxinMDC ILS

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 1S.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 Key: hrl3 (human tUbroblasts), Yl (placenta-aenvea rrL~ ~u4~sus)), a 1 (umblncal cord-nenvea rrlW (VLZxUS)), P3 (placenta-derived PPDC (071003)), U3 (umbilical cord-derived PPDC
(071003)). ND: Not Detected.
Summary. Umbilical cord-cells secreted significantly higher amount of trophic factors than placenta-derived cells and fibroblasts. Some of these trophic factors, such as HGF, bFGF, MCP-1 and 1L-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL,-6, have important roles in neural regeneration. Under these conditions, the expression of some factors was confined to umbilical cord-derived cells, such as MlPlb, Rantes, I309, and FGF.
Re ere~zces Le Belle JE, Svendsen CN.(2002) Stem cells for neurodegenerative disorders:
where can we go from here? BioDrugs.l6;389-401 Rosen EM, Lamszus K, Laterra J, Polverini PJ, Rubin JS, Goldberg ID. (1997) HGF/SF
in angiogenesis. Ciba Found Symp. 212;215-26,.
Salcedo R, Ponce ML, Young HA, Wasserman K, Ward JM, Kleinman HK, Oppenheim JJ, Murphy WJ. (2000) Human endothelial cells express CCR2 and respond to MCP-l: direct role of MCP-1 in angiogenesis and tumor progression. Blood. 96;34-40.
Li A, Dubey S, Val-ney ML, Dave BJ, Singh RK (2003) IL-8 directly enhanced endothelial cell survival, proliferation, and matrix metalloproteinases production and regulated angiogenesis.J Immunol. 170;3369-76 Hughes GC, Biswas SS, Yin B, Coleman RE, DeGrado TR, Landolfo CK, Lowe JE, Annex BH, Landolfo KP. (2004) Therapeutic angiogenesis in chronically ischemic porcine myocardium: comparative effects of bFGF and VEGF. Ann Thorac Surg. 77;812-8.
Cheng A, Wang S, Cai J, Rao MS, Mattson MP (2003) Nitric oxide acts in a positive feedback loop with BDNF to regulate neural progenitor cell proliferation and differentiation in the mammalian brain. Dev Bio1.258;319-33..
Sebire G, Emilie D, Wallon C, Hery C, Devergne O, Delfraissy JF, Galanaud P, Tardieu M. (1993) I12 oitro production of IL-6, IL-1 beta, and tumor necrosis factor-alpha by human embryonic microglial and neural cells. J Immunol. 150;1517-23.
EXAMPLE 13:
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 vi.vo.
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. Nephr-ol. 10:299-306). To determine the potential for PPDCs to initiate clotting, umbilical cord- and placenta-derived PPDCs were evaluated for tissue factor expression and their ability to initiate plasma clotting.
Methods & Materials Human Tissue factor. Human tissue factor SBvIPLASTIN (Organon Tekailca Corporation, Durham, NC), was reconstituted with 20 milliliter 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 microliter phosphate buffered saline (PBS), nucroliter diluted Simplastin~ (except a blank well), 30 microliter 0.1 molar calcium chloride, and 100 microliter of normal human plasma. The plate was immediately placed in a temperature-controlled nucroplate reader and absorbance measured at 405 nanometer at 40 second intervals for 30 nunutes.
J-82 and postpartum-derived cells. J-82 cells (ATCC, MD) were grown in Iscove's modified Dulbecco's medium (IIvvIDM; Gibco, Carlsbad, CA) containing 10% (v/v) fetal bovine serum (FBS; Hyclone, Logan U'T), 1 millimolar sodium pyruvate (Sigma Chemical, St. Louis, MO), 2 millimolar L-Glutamin (Mediatech Herndon, VA), 1 x non-essential amino acids (Mediatech Hendon, VA). At 70% confluence, cells were transferred to wells of 96-well plate at 100,000, 50,000, and ?5,000 cells/well. Postpartum cells derived from placenta and umbilical cord were cultured in Growth Medium (DMEM-low glucose (Gibco), 15°l0 (v/v) FBS, 50 Units/milliliter penicillin, 50 micrograms/milliliter streptomycin (Gibco), and 0.001 °70 betamercaptoethanol (Sigma)) in gelatin-coated T75 flasks (Corning, Corning, NY). Placenta-derived cells at passage 5 and umbilical cord-derived cells at passages 5 and 11 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 microgram/milliliter ONTO 859 (Centocor, Malvern, PA) for 30 minutes. Calcium chloride (30 microliter) was added to each well. The plate was immediately placed in a temperature-controlled microplate reader and absorbance measured at 405 nanometers at 40 second intervals for 30 minutes.
Antibody Staining. Cells were washed in PBS and detached from the flask with Trypsin/EDTA (Gibco Carlsbad, CA). Cells were harvested, centrifuged, and re-suspended 3%
(v/v) FBS in PBS at a cell concentration of 1x107 per milliliter. Antibody was added to 100 microliter cell suspension as per the manufacturer's specifications, and the cells were incubated in the dark for 30 minutes at 4°C. After incubation, cells were washed with PBS and centrifuged at 150 x g for 5 minutes to remove unbound antibody. Cells were re-suspended in 100 nucroliter of 3% FBS and secondary antibody added as per the manufacturer's instructions.
Cells were incubated in the dark for 30 minutes at 4°C. After incubation, cells were washed with PBS and centrifuged to remove unbound secondary antibody. Washed cells were re-suspended in 500 microliter 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 I/z to max; Table 13-1).
Clotting was observed with both early (P5) and late (P18) cells. The T'/a 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 13-1. The effect of human tissue factor (SnVIPLAST1N), 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.
Sim lastin~ Dilution T'/z to max (seconds) 1:2 61 _ 107 1:4 _ 1:8 147 _ 1:16 174 _ 1:32 266 1:64 317 1:128 37S

0 (negative control) 11SS ,_ -_ J-82 cells 100,000 122 50,000 172 25,000 275 Pla P5 50,000 757 Umb PS

50,000 S33 Umb P 1 S

50,000 443 Summary. Placenta- and umbilical cord-derived PPDCs express tissue factor, which can induce clotting. The addition of an antibody to tissue factor can inhibit tissue factor. Tissue factor is normally found on cells in a conformation that is inactive but is activated by mechanical or chemical (e.g., LPS) stress (Sakariassen et al. (2001) Tlr.ronzb. Res.
104:149-74; Engstad et al.
(2002) ITZt. Ifnnaacnopharnaacol. 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-deuved PPDCs are transplanted in tissue but should be inhibited when PPDCs are injected intravenously.
References Doshi and Marmur, Critical Care llTed., 30:5241-5250 (2002) Moll and Ortel, Afzn. In.tef~a. Med., 127:177-1S5 (1997) Differentiation of PPDCs to an Osteogenic Phenotype Mesenchymal stem cells (MSCs) derived from bone maiTOw can differentiate into osteoblast-like cells that mineralize and express alkaline phosphatase.
Additional markers expressed by osteoblasts, such as osteocalcin and bone sialoprotein, have also been used to demonstrate differentiation into an osteoblast-like cell. A determination was made as to whether postpartum-derived cells can also differentiate into an osteogenic phenotype by culturing in an - lOS -osteogenic medium and in the presence of bone morphogenic proteins (BMP) -2 (Rickard et a.1., 1994) or -4, and transforming growth factor betal.
Methods & Materials Culture of cells. Prior to initiation of osteogenesis, Mesenchymal Stem Cells (MSC) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM, Cambrex, Walkerville, MD). Other cells were cultured in Growth medium (DMEM-low glucose (Gibco, Carlsbad, CA), 15°10 (v/v) fetal bovine serum (SH30070.03; Hyclone, Logan, UTI, 0.001% (v/v) betamercaptoethanol (Sigma, St. Louis, MO), penieillin/streptomycin (Gibco)), in a gelatin-coated T75 flask were washed with phosphate buffered saline (PBS).
Osteoblasts (9F1721; Cambrex) were grown in osteoblast growth medium (Cambrex) and RNA was extracted as described below.
Osteogenesis Protocol I. Placenta-derived cells, isolate 1, P3, placenta-derived cells, isolate 2, P4 (previously karyotyped and shown to be predominantly neonatal-derived cells), umbilical cord-derived cells isolate 1, P4, and MSC at P3 were seeded at 5 x 103 cells/cm2 in 24 well plates and 6-well dishes in Growth medium and incubated overnight. The medium was removed and replaced with Osteogenic medium (DMEM-low glucose, 10% (v/v) fetal bovine serum, 10 millimolar betaglycerophosphate (Sigma), 100 nanomolar dexamethasone (Sigma, St. Louis, MO), 50 micromolar ascorbate phosphate salt (Sigma), fungizone (Gibco), penicillin and streptomycin (Gibco)). Osteogenic medium was supplemented with 20 nanograms/milliliter hTGF-betal (Sigma), 40 nanograms/milliliter hrBMP-2 (Sigma), or 40 nanograms/milliliter hrBMP-4 (Sigma). Cultures were treated for a total of 14, 21 and 28 days, with media changes every 3-4 days.
Protocol 2. Postpartum-derived cells were tested for the ability to differentiate into an osteogenic phenotype. Umbilical cord-derived cells (isolate 1, P3 & isolate 2, P4) and placenta-derived cells (isolate 1; P4 & isolate 2, P4) were seeded at 30,000 cellslwell in 6-well, gelatin-coated plates in Growth medium. Mesenchymal stem cells (MSC) (isolate 1; P3 &
isolate 2;
P4), fibroblasts (1F1853, P11), and ileac crest bone marrow cells (070203; P3;
W02003025149) were also seeded at 30,000 cells/well in 6-well, gelatin-coated plates (gelatin-coated) in mesenchymal stem cell growth medium (MSCGM, Cambrex) and Growth medium, respectively.
Osteogenic induction was initiated by removing the initial seeding media (24 h) and replacing it with osteogenic induction medium (DMEM-low glucose, 10% fetal bovine serum, 10 millimolar betaglycerophosphate (Sigma), 100 nanomolar dexamethasone (Sigma), micromolar ascorbate phosphate salt (Sigma), penicillin and streptomycin (Gibco)). In some cases, osteogenic medium was supplemented with either hrBMP-2 (20 nanograms/milliliter) (Sigma), hrBMP-4 (Sigma), or with both hrBMP-2 (20 nanograms/milliliter) and hrBMP-4 (20 nanograms/milliliter) (Sigma). Cultures were treated for a total of 28 days, with media changes every 3-4 days.
RNA extraction and Reverse Transcription. Cells were lysed with 350 microliter buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, MO) according to the manufacturer's instructions (RNeasy Mini kit, Qiagen, Valencia, CA) and stored at -80°C. Cell lysates were thawed and RNA extracted according to the manufacturer's instructions (RNeasy Mini kit, Qiagen, Valencia, CA) with a 2.7 U/sample DNase treatment (Sigma St.
Louis, MO).
RNA was eluted with 50 microliter 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.
Polymerase Chain Reaction. PCR was performed on cDNA samples using Assays-on-DemandTM gene expression products bone sialoprotein (Hs00173720), osteocalcin (Hs00609452) GAPDH (Applied Biosystems, Foster Gity, CA), 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 min and 95°C for 10 min followed by 40 cycles of 95°C for 15 sec and 60°C for 1 min.
von Kossa Staining. Cells were fixed with 10% (v/v) neutral buffered formalin (Richard-Allan, Kalamazoo, MI). After fixation, the cells were washed in deionized water and incubated in 5% (wlv) silver nitrate (Aldrich Chemical Company, Milwaukee, WI) for one hour in direct sunlight. Cells were then washed in DI water and incubated in 5%
(w/v) sodium thiosulfate (EM Sciences, Gibbstown, NJ) for five minutes. Cells were washed in distilled water and examined by light microscopy.
Results Protocol 1. RNA extracted from osteoblasts was used as a positive control for the real-time gene expression of osteocalcin and bone sialoprotein. Osteoblast expression levels relative to placenta-derived cells grown in growth medium of osteocalcin and BSP was 2.5- and 8000-fold, respectively. MSCs grown in the osteogenic medium for 28 days mineralized and were positive for von Kossa staining. Extensive mineralization was observed in one placenta isolate -llo-that had predominantly neonatal-derived cells. Also, one placenta isolate show induction of BSP
expression levels in osteogenic media and low levels of osteocalcin induction.
MSC expression of osteocalcin and BSP was significantly increased in osteogenic medium at 21 days. The addition of BMP-2 and -4 enhanced BSP expression but had no effect on osteocalcin expression. TGF-betal did not augment the effect of osteogenesis medium.
BMP-4 and TGF-betal both increased osteocalcin expression by a placenta isolate.
Protocol 2. Osteogenic differentiation, as shown by positive von Kossa staining for mineralization, was observed with placenta-derived cells P4 and ICBM (070203), P3 incubated with osteogenic medium supplemented with BMP2 or 4, and MSCs (092903) P3 incubated with osteogenic medium supplemented with BMP 4 (Table 14-1). None of the other cells differentiated into the osteogenic phenotype and stained by von Kossa. To ensure that von Kossa staining was related to the cell and not to the extracellular matrix, cells were counterstained with nuclear fast red. Large lipid clioplets were observed in some MSCs consistent with an adipocyte phenotype. This suggests that MSCs do not differentiate specifically into an osteogenic phenotype in these conditions. Furthermore, adipogenesis increased when MSCs were incubated in osteogenic medium supplemented with either BMP2 or BMP4.
Table 14-1: Results of osteogenic differentiation using von Kossa staining for Protocol 2.
Umbilical cord-derived cells (Umb), placenta-derived cells (Pla), mesenchymal stem cells (MSC), fibroblasts (Fib), and ileac crest bone marrow cells (ICBM) cells were cultured in osteogenic medium (OM) atone or supplemented with BMP2 or BMP2 and BMP4.
Number Cell Line Conditions Von Kossa Comments 1 Umb 071003 Osteogenic Neg O1P3 medium (OM) 2 Umb 071003 OM, BMP2 Neg 3 Umb 071003 OM, BMP4 Neg 4 ICBM 070203 Osteogenic Neg Normal 01P3 medium (OM) ICBM 070203 OM, BMP2 Pos Normal 6 ICBM 070203 OM, BMP4 Pos Normal 7 MSC 092903 Osteogenic Neg lots of fat medium (OM) S MSC 092903 OM, BMP2 Neg lots of fat 9 MSC 092903 OM, BMP4 Pos lots of fat Pla lOlE03 Osteogenic Neg medium (OM) 11 ~ Pla 101603 OM, BMP2 Pos _ 12 Pla 101603 OM, BMP4 Pos
13 MSC 012104 Osteogenic Neg Fat 01P4 medium (OM)
14 MSC 012104 OM, BMP2 Neg Fat
15 MSC 012104 OM, BMP2, Neg Fat
16 Umb 022503 Osteogenic Neg O1P4 medium (OM)
17 Umb 022503 OM, BMP2 Neg 15 Umb 022503 OM, BMP2, Neg 19 Pla 100703 Osteogenic Neg medium (OM) 20 Pla 100703 OM, BMP2 Nea 21 Pla 100703 OM, BMP2, Neg 22 Fib 1F1853 Osteogenic Neg 01P11 medium (OM) ~

23 Fib 1F1S53 OM, BMP3 Neg 24 Fib 1F1853 OM, BMP2, Neg Summary. Bone marrow-derived MSCs (Kadiyala et al., 1997) as well as cells derived from other tissue such adipose (Halvorsen et al., 2001) have been shown to differentiate into osteoblast-like cells. MSCs have also been shown to differentiate into adipocytes or osteoblasts in response to BMPs (Chen et al., 1998) due to different roles for bone morphogenic protein (BMP) receptor type IB and IA.
Neonatal-derived placenta-derived cells and MSCs showed mineralization as well as induction of osteocalcin and bone sialoprotein. Under the conditions used, umbilical-derived cells did not show mineralization or induction of osteoblast genes. Maternal placenta-derived cells may require addition of BMP-4 or TGF to the osteogenic medium for mineralization to occur. The gestational age of the sample may also be a factor in the ability of cells derived from postpartum tissues to differentiate.
References:
Kadiyala S, Young RG, Thiede MA, Bruder SP. (1997) Culture expanded canine mesenchymal stem cells possess osteochondrogenic potential in vivo and in.
vitro. Cell Transplant.6:125-34.

Chen D, Ji X, Harns MA, Feng JQ, Karsenty G, Celeste AJ, Rosen V, Mundy GR, Harris SE. (1998) Differential roles for bone morphogenic protein (BMP) receptor type IB and IA in differentiation and specification of mesenchymal precursor cells to osteoblast and adipocyte lineages. J Cell Biol. 142:295-305 Halvorsen YD, Franklin D, Bond AL, Hitt DC, Auchter C, Boskey AL, Paschalis EP, Wilkison WO, Gimble JM (2001) Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells. Tissue Eng. 7:729-41.
Richard DJ et. al., (1994) Induction of rapid osteoblast differentiation in rat bone marrow stromal cell cultures by dexamethasone and BMP-2. Dev Biol 161,:218-228 W02003025149 A2 HO, Tony, W. ;KOPEN, Gene, C. ;RIGHTER, William, F.
;RUTKOWSKI, J., Lynn ;HERRING, W., Joseph ;RAGAGLIA, Vanessa ;WAGNER, Joseph CELL POPULATIONS WHICH CO-EXPRESS CD49C AND CD90, NEURONYX, INC.
Application No. US0229971 LTS, Filed 20020920, A2 Published 20030327 ,A3 Published Chondrogenic Differentiation of Postpartum-derived Cells Cartilage damage and defects lead to approximately 600,000 surgical procedures each year in the United States alone (1). A number of strategies have been developed to treat these conditions but these have had limited success. One approach, Cartecel (Genzyme), uses autologous chondrocytes that are collected from a patient and expanded in vitro and then implanted into the patient (1). This approach has the disadvantage of collecting healthy cartilage and requiring a second procedure to implant the cultured cells. One novel possibility is a stem cell-based therapy in which cells are placed at or near the defect site to directly replace the damaged tissue. Cells may be differentiated into chondrocytes prior to the application or progenitor cells that can differentiate r.'f~ situ may be used. Such transplanted cells would replace the chondrocytes lost in the defect.
Candidate cells for this indication should be evaluated for their ability to differentiate into chondrocytes ifi vitro. A number of protocols have been developed for testing the ability of cells to differentiate and express chondrocyte marker genes. Postpartum-derived cells were tested for their ability to differentiate into chondrocytes in vitro in two different assay systems: the pellet assay culture system and collagen gel cultures. The pellet culture system has been used successfully with selected lots of human mesenchymal stem cells (MSC). MSCs grown in this assay and treated with transforming growth factor-beta3 have been shown to differentiate into chondrocytes (2). The collagen gel system has been used to culture chondrocytes in vitro (3).
Chondrocytes grown under these conditions form a cartilage-like structure.
Materials afZd Metlaods Cell Culture Postpar-tuna tissue-derioed cells. Human umbilical cords and placenta were received and cells were isolated as described above. Cells were cultured in Growth medium (Dulbecco's Modified Essential Media (DMEM) with 15% (vlv) fetal bovine serum (Hyclone, Logan UT), penicillin/streptomycin (Invitrogen, Carlsbad, CA), and 0,001% (v/v) 2-mercaptoethanol (Sigma, St. Louis, MO)) on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C with 5 % COZ. For use in experiments, cells were between passages 4 and 12.
Human czrtica~lar chondrocytes. Human articular chondrocytes were purchased from Cambrex (Walkersville, MD) and cultured in the same media as the postpartum-derived cells.
Twenty-four hours before the experiment, the culture media was changed to a media containing.
1% FBS.
Huuzafz mesefaclaywal stern cells (h.MSC). MSCs were purchased from Cambrex (Walkersville, MD) and cultured in MSCGM (Cambrex). Cells used for experiments were between passages 2 and 4.
Collagen gel assays. Cultured cells were trypsinized to remove from culture plate. Cells were washed with centrifugation twice at 300 x g for 5 min in DMEM without serum and counted. Cells were mixed with the following components at the final concentrations listed. Rat tail collagen (1 milligram/milliliter , BD DiscoveryLabware, Bedford, MA), 0.01 N NaOH and Chondrogenic medium (DMEM, 100 U/100 microgram Penicillin/Streptomycin, 2 millimolar L-Glutamine, 1 millimolar Sodium Pyruvate, 0.35 millimolar L-Proline, 100 nanomolar dexamethasone, 0.17 millimolar L-Ascorbic Acid, 1 % (v/v) ITS (insulin, transfen~in, selenium) (All components from Sigma Chemical Company)). The cells were gently mixed with the medium the samples were aliquoted into individual wells of a 24 well ultra-low cluster plate (Corning, Corning, NI') at a concentration of either 2 x 105 per well or 5 x 105 per well.
Cultures were placed in an incubator and left undisturbed for 24-48 hours.
Medium was replaced with fresh chondrogenic medium supplemented with appropriate growth factor every 24-48 hours. Samples were allowed to culture for up to 28 days at which time they were removed and fixed in 10% (v/v) formalin (VWR Scientific, West Chester, PA) and processed for histological examination. Samples were stained with Safranin O or hematoxylin/eosin for evaluation.

Pellet culture assays. Cultured cells were trypsinized to remove from culture plate.
Cells were washed with centrifugation twice at 300 x g for 5 minutes in DMEM
without serum and counted. Cells were resuspended in fresh chondrogenic medium (described above) at a concentration of 5 x 105 cells per milliliter. Cells were aliquoted into new polypropylene tubes at 2.5 x 105 cells per tube. The appropriate samples were then treated with TGF-beta3 (10 nanograms/milliliter, Sigma) or GDF-5 (100 nanograms/milliliter; R&D Systems, Minneapolis, MN). Cells were then centrifuged at 150 x g for 3 minutes. Tubes were then transferred to the incubator at and left undisturbed for 24-48 hours at 37 °C and 5 % CO~.
Media was replaced with fresh chondrocyte cell media and growth factor, where appropriate, every 2-3 days.
Samples were allowed to culture for up to 28 days at which time they were removed and fixed and stained as described above.
Resrslts Pellets were prepared and cultured and described in Methods. Pellets were grown in media (Control) or supplemented with TGF-beta3 (10 nanograms/milliliter) or GDF-5 (100 nanograms/milliliter) that was replaced every 2-3 days. Pellets collected after 21 days of culture and stained by Safranin O to test for the presence of glycosoaminoglycans. The pellets treated with TGFbeta3 and GDF-5 showed some positive Safranin O staining as compared to control cells. The morphology of the umbilical cord cells showed some limited chondrocyte-like morphology.
Safranin O stains of cell pellets from placenta cells showed similar glycosoaminoglycan expression as compared to the umbilical cord cells. The morphology of the cells also showed some limited chondrocyte-like morphology.
Summary. The results of the present study show that the postpartum-derived cells partially differentiated into chondrocytes in. vitro in the pellet culture and the collagen gel assay systems. The postpartum-derived cells showed some indications of glycosaminoglycan expression by the cells. Morphology showed limited similarity to cartilage tissue.
REFERENCES
1. U.S. Markets for Current and Emerging Orthopedic Biomaterials Products and Technologies. Medtech Insight L.L.C. 2002 2. Johnstone, B, T. M. Hering, A.I. Caplan, V.M. Goldberg and J.U. Yoo. ht Vitro Chondrogenesis of Bone-Marrow-Derived Mesenchymal Stem Cells. 1998. Exp Cell Res 238:265-272.

3. Gosiewska, A., A. Rezania, S. Dhanaraj, M. Vyakarnam, J. Zhou, D. Burns, L.
Brown, W. bong, M. Zimmerman and J. Geesin. Development of a Three-Dimensional Transmigration Assay for Testing Cell-Polymer Interactions for Tissue Engineering Applications. 2001 Tissue Eng. 7:267-277.

Evaluation of Chondrogenic Potential of Cells Derived from Postpartum Tissue in an Irr Vitro Pellet Culture Based Assay This example describes evaluation of the chondrogenic potential of cells derived from placental or umbilical tissue using in vitro pellet culture based assays.
Cells from umbilical cord and placenta at early passage (P3) and late passage (P12) were used. The chondrogenic potential of the cells was assessed in pellet culture assays, under chondrogenic induction conditions, in medium supplemented with transforming growth factor beta-3 (TGFbeta-3), GDF-5 (recombinant human growth and differentiation factor 5), or a combination of both.
lVlaterzals & Methods Reagents. Dulbecco's Modified Essential Media (DMEM), Penicillin and Streptomycin, were obtained from Invitrogen, Carlsbad, CA. Fetal calf serum (FCS) was obtained from HyClone (Logan, UT). Mesenchymal stem cell growth medium (MSCGM) and hMSC
chondrogenic differentiation bullet kit were obtained from Biowhittaker, Walkersville, MD.
TGFbeta-3 was obtained from Oncogene research products, San Diego, CA. GDF-5 was obtained from Biopharm, Heidelberg, Germany (W09601316 A1, US5994094 A).
Cells. Human mesenchymal stem cells (Lot# 2F1656) were obtained from Biowhittaker, Walkersville, MD and were cultured in MSCGM according to manufacturer's instructions. This lot has been tested previously, and was shown to be positive in the chondrogenesis assays.
Human adult and neonatal fibroblasts were obtained from American Type Culture Collection (ATCC), Manassas, VA and cultured in growth medium (Dulbecco's Modified Essential supplemented with 15% (v/v) fetal bovine serum, penicillin/streptomycin (100 U/100 milligram, respectively) and 0.001 % (v/v) 2-mercaptoethanol (Sigma, St. Louis, MO) on gelatin-coated tissue culture plastic flasks. Postpartum tissue-derived cells, isolated from human umbilical cords (Lot# 022703Umb) and placenta (Lot# 071003P1ac) as described in previous examples, were utilized. Cells were cultured in Growth medium similar to fibroblasts.
The cell cultures were incubated at 37 °C with 5 % CO~. Cells used for experiments were at passages 3 and 12.

Pellet culture assay. For pellet cultures, 0.25x106 cells were placed in a 15 milliliter conical tube and centrifuged at 150xg for 5 minutes at room temperature to form a spherical pellet according to protocol for chondrogenic assay from Biowhittaker. Pellets were cultured in chondrogenic induction medium containing TGFbeta-3 (10 nanograms/milliliter), GDF-5 (500 nanograms/milliliter), or a combination of TGFbeta-3 (10 nanograms/milliliter), and GDF-5 (500 nanograms/milliliter) for three weeks. Untreated controls were cultured in growth medium.
During culture, pellets were re-fed with fresh medium every other day.
Treatment groups included the following:
Treatment Groun A. Placenta-derived cells early passage (P EP) + GDF-5 B. Placenta-derived cells late passage (P LP) + GDF-5 C. Umbilical cord derived cells early passage (U EP) + GDF-5 D. Umbilical cord derived cells late passage (U LP) + GDF-5, n=2 E. Human Mesenchymal Stem cells (HMSC) + GDF-5 F. I-~uman adult fibroblast cells (HAF) + GDF-5 G. Placenta-derived cells early passage (P EP) + TGFbeta-3 H. Placenta-derived cells late passage (P LP) + TGFbeta-3 I. Umbilical cord derived cells early passage (U EP) + TGFbeta-3 J. Umbilical cord derived cells late passage (U LP) + TGFbeta-3, n=2 K. Human Mesenchymal Stem cells (HMSC) + TGFbeta-3 L. Human adult fibroblast cells (HAF) + TGFbeta-3 M. Placenta-derived cells early passage (P EP) + GDF-5+ TGFbeta-3, n=1 N. Placenta-derived cells late passage (P LP) + GDF-5+ TGFbeta-3 O. Umbilical cord derived cells early passage (U EP) + GDF-5 + TGFbeta-3 P. Umbilical cord derived cells late passage (U LP) + GDF-5 + TGFbeta-3, n=2 Q. Human Mesenchymal Stem cells (HMSC) + GDF-5+ TGFbeta-3 R. Human adult fibroblast cells (HAF) + GDF-5+ TGFbeta-3 S. Human neonatal fibroblast cells (HNF) + GDF-5+ TGFbeta-3 T. Placenta-derived cells early passage (P EP) U. Placenta-derived cells late passage (P LP) V. Umbilical cord derived cells early passage (U EP) W. Umbilical cord derived cells late passage (LT LP) X. Human Mesenchymal Stem cells (HMSC) Y. Human adult fibroblast cells (HAF) Histology of in vitro samples. At the end of the culture period pellets were fixed in 10%
buffered formalin and sent to MPI Research (Mattawan, MI) for paraffin embedding, sectioning, and staining with Hematoxylin /Eosin (H/E) and Safranin O (SO) staining.
Results Placenta- and umbilical cord-derived cells, MSCs, and fibroblasts formed cell pellets in chondrogenic induction medium with the different growth factors. The size of the pellets at the end of culture period varied among the different cell types. Pellets formed with placenta-derived cells were similar in size, or slightly larger than, those formed by MSCs and fibroblasts. Pellets formed with the umbilical cord-derived cells tended to be larger and looser than the other groups.
Pellets fomned with all cell types and cultured in control medium were smaller than pellets cultured in chondrogenic induction medium.
Examination of cross sections of pellets stained with hematoxylin/eosin and Safranin-O
showed that umbilical cord-derived cells at early passage had the potential to undergo chondrogenic differentiation. Chondrogenesis as assessed by cell condensation, cell morphology and Safranin O positive staining of matrix was observed in the umbilical cell pellets cultured in chondrogenic induction medium supplemented with TGFbeta-3, GDF-5, or both.
Chondrogenesis in pellets was similar for TGFbeta-3, GDF-5, and the combined treatments.
Control pellets cultured in growth medium showed no evidence of chondrogenesis.
Chondrogenic potential of the umbilical cord derived cells was marginally lower than that observed with the MSCs obtained from Biowhittaker.
Umbilical cord derived cells at late passage and placenta-derived cells did not demonstrate as distinct a chondrogenic potential as did early passage umbilical cord-derived cells. However, this may be due to the fact that chondrogenic induction conditions were optimized for MSCs, not for postpartum-derived cells. Nonetheless, distinct cell populations were observed in placenta-derived cells at both passages located apically or centrally. Some cell condensation was observed with fibroblast, but it was not associated with Safranin O staining.

Endothelial Network Formation Assay 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-derived cells in ita vitro assays. The study followed a well-established method of seeding endothelial cells onto a culture plate coated with MATRIGEL (BD
Discovery Labware, Bedford, MA), a basement membrane extract (Nicosia and Ottinetti (1990) 1st Vitro Cell Dev.
Biol. 26(2):119-28). Treating endothelial cells on MATRIGEL (BD Discovery Labware, Bedford, MA) with angiogenic factors will stimulate the cells to form a network that is similar to capillaries. This is a common i.~t vr.'tro assay for testing stimulators and inhibitors of blood vessel formation (Ito et al. (1996) Int. J. Cancer- 67(1):148-52). The present studies made use of a co-culture system with the postpartum-derived cells seeded onto culture well inserts. These permeable inserts allow for the passive exchange of media components between the endothelial and the postpartum-derived cell culture media.
Material c~ Meth.ods Cell Culture.
Postpartur~t. tissue-derived cells. Human umbilical cords and placenta were received and cells were isolated as previously described (Example 1). Cells were cultured in Growth medium (Dulbecco's Modified Essential Media (DMEM; Invitrogen, Carlsbad, CA), 15%
(vlv) fetal bovine serum (Hyclone, Logan UT), 100 Units/milliliter penicillin, 100 microgramlmilliliter streptomycin (Invitrogen), 0.001 % (v/v) 2-mercaptoethanol (Sigma, St. Louis, MO)) on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37 °C with 5 % COZ. Cells used for experiments were between passages 4 and 12.
Actively growing postpartum cells were trypsinized, counted, and seeded onto COSTAR
TRANSWELL 6.5 millimeter 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
under standard growth conditions.
Hur~tan ntesertchymal stem cells (ItMSC). hMSCs were purchased from Cambrex (Walkersville, MD) and cultured in MSCGM (Cambrex). The cultures were incubated under standard growth conditions.
Actively growing MSCs were trypsinized and counted and seeded onto COSTAR
TRANSWELL 6.5 millimeter 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 under standard growth conditions.
Htvmata umbilical veita endothelial cells (HUVEC). HUVEC 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 under standard growth conditions. Cells used in the assay were between passages 4 and 10.
Hufzzafz corofza~y aWery endothelial cells (HCAEC). HCAEG were purchased from Cambrex Incorporated (Walkersville, MD). These cells were also maintained in separate cultures in either the EBM or EGM media formulations. Cells were grown on standard tissue cultured plastic under standard growth conditions. Cells used for experiments were between passages 4 and 8.
Endothelial Network Formation (MATRIGEL) assays. Culture plates were coated with MATRIGEL (BD Discovery Labware, Bedford, MA) according to manufacturer's specifications. Briefly, MATRIGELTM (BD Discovery Labware, Bedford, MA) was thawed at 4°C and approximately 250 microliter 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 medium 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 milliliter Growth medium with 2% (vlv) FBS. Cells were then incubated for approximately 30 minutes to allow cells to settle.
Endothelial cell cultures were then treated with either 10 nanomolar 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-derived cells were added to appropriate wells with Growth medium with 2% FBS
in the insert chamber. Cultures were incubated at 37°C with 5 %CO~ 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 (data not shown). HUVEC cells fomn limited cell networks in co-culture experiments with hMSC and with 10 nanomolar bFGF (data not shown). I-ifJVEC cells without any treatment showed very little or no network formation (data not shown). These results suggest that the postpartum-derived cells release angiogenic factors that stimulate the HLTVEC.
In a co-culture system with placenta-derived cells or with umbilical cord-derived cells, CAECs form cell networks (data not shown).

Table 17-1 shows levels of known angiogenic factors released by the postpartum-derived cells in Growth medium. Postpartum-derived 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 was removed, immediately frozen and stored at -SO°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-derived 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 17-1. Potential angiogenic factors released from postpartum-derived cells.
Postpartum-derived cells were cultured in 24 hours in media with 2% FBS in atmospheric oxygen. Media was 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.

( /ml) ( /ml)( /ml) ( /ml)( /ml)( /ml)( /ml)( /ml) ( /ml) Plac (P4) 91655.3 175.5 <2.0 275.5 3.0 58.3 7.5 644.6 <1.2 Plac (P11) 1592832.428.1 <2.0 1273.1193.3 5960.334.8 12361.11.7 Umb 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 Plac: placenta derived cells; Umb cord: Umbilical cord derived cells Table 17-2 shows levels of known angiogenic factors released by the postpartum-derived cells. Postpartum-derived 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% O~ incubation for 24 hours. Media was 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-derived 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 (TIIvVIP-1), angiopoietin 2 (ANG2), thrombopoietin (TPO), keratinocyte a ~owth factor (KGF), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEGF).
Table 17-2. Potential angiogenic factors released from postpartum-derived cells.
Postpartum-derived cells were cultured in 24 hours in media with 2% FBS in 5%
oxygen. Media was 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 milliter of culture media.
PDGF- HB-( ~~) ( ~~) ( ~~) (p~~) ( ~) (p~~) ( F~~)( ~W ( ml) Plac (P4) 72972.5 253.6 <2.0 743.1 2.5 30.2 15.1 1495.1 <1.2 Plac (P11) 458023.155.1 <2.0 2562.2 114.2 213S.0295.1 7521.3 1.8 Umb 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 Plac: placenta derived cells; Umb cord: Umbilical cord derived cells Summary. The results of the present study show that postpartum-derived cells can stimulate both human umbilical vein and coronary artery endothelial cells to form networks in an i~2 vitro MATRIGELTM (BD Discovery Labwwe, Bedford, MA) assay. This effect is similar to that seen with known angiogenic factors in this assay system. These results suggest that the postpartum-derived cells are useful for stimulating angiogenesis ifi vivo.

Transplantion of PPDCs Cells derived from the postpartum umbilical cord and placenta are useful for regenerative therapies. The tissue produced by postpartum-derived cells transplanted into SC)D mice with a _122_ biodegradable material was evaluated. The materials evaluated were VICRYL non-woven, 35/65 PCL/FGA foam, and RAD 16 self-assembling peptide hydrogel.
Metlaods & Materials Cell Culture. Placenta-derived cells and umbilical cord derived cells were grown in Growth medium (DMEM-low glucose (Gibco, Carlsbad CA), 15% (v/v) fetal bovine serum (Cat.
#SH30070.03; Hyclone, Logan, UT), 0.001°Io (v/v) betamercaptoethanol (Sigma, St. Louis, MO), 50 Units/milliliter penicillin, 50 microgram/milliliter streptomycin (Gibco)) in a gelatin-coated flasks.
Matrix Preparation. A nonwoven scaffold was prepared using a traditional needle punching technique as described below. Fibers, comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the tradename VICRYL were obtained from Ethicon, Inc. (Somerville, NJ). The fibers were filaments of approximately 20 microns in diameter. The fibers were then cut and crimped into uniform 2-inch lengths to form 2-inch staple fiber. A dry lay needle-punched nonwoven matrix was then prepared utilizing the VICRYL staple fibers. The staple fibers were opened and carded on standard nonwoven machinery. The resulting mat was in the form of webbed staple fibers. The webbed staple fibers were needle-punched to form the dry lay needle-punched nonwoven scaffold. The nonwoven scaffold was rinsed in water followed by another incubation in ethanol to remove any residual chemicals or processing aids used during the manufacturing process.
Foams, composed of 35/65 poly(epsilon-caprolactone)/poly(glycolic acid) (35/65 PCL/PGA) copolymer, werer formed by the process of lyophilized, as discussed in U.S. Patent No. 6,355,699.
Sample Preparation. One million viable cells were seeded in 15 microliter Growth medium onto 5 millimeter diameter, 2.25 millimeter thick VICRYL non-woven scaffolds (64.33 milligram/cubic centimeters; Lot#3547-47-1) or 5 millimeter diameter 35/65 PCL/PGA foam (Lot# 3415-53). Cells were allowed to attach for two hours before adding more Growth medium to cover the scaffolds. Cells were grown on scaffolds overnight. Scaffolds without cells were also incubated in medium.
RAD16 self-assembling peptides (3D Matrix, Cambridge, MA under a material transfer agreement) was obtained as a sterile 1 ~/o (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 microliter.

TEST 14~IATERIAL (N=4/Rx) 1. VICRYL non-woven + 1 x 106 umbilical cord-derived cells 2. 35/65 PCLIPGA foam + 1 x 106 umbilical cord-derived cells 3. RAD 16 self-assembling peptide + 1 x 106 umbilical cord-derived cells 4. VICRYL non-woven + 1 x 106 placenta-derived cells 5. 35/65 PCL/PGA foam + 1 x 106 placenta-derived cells 6. RAD 16 self-assembling peptide + 1 x 106 placenta-derived cells 7. 35/65 PCL/PGA foam 8. 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 Musculacs)lFox Chase SCIDlMale (Harla~z Sprague Dawley, Inc., l~2dia~aapolis, In~liarza), S weeks of age. All handling of the SC1D mice took place under a hood.
The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligram/kilogram KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligram/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 scmbbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine. Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period.
Subcutaneous Implantation Technique. Four skin incisions, each approximately 1.0 cm in length, were made on the dorsum of the mice. Two cranial sites were located transversely over the dorsal lateral thoracic region, about 5-mm caudal to the palpated inferior edge of the scapula, with one to the left and one to the right of the vertebral column.
Another two were placed transversely over the gluteal muscle area at the caudal sacro-lumbar level, about 5-mm caudal to the palpated iliac crest, with one on either side of the midline.
Implants were randomly placed in these sites. The skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected for RAD16) about 1-cm caudal to the incision.
- 1?4 -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 nlicroisolator 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 libitanra.
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.
Resr~lts There was minimal ingrowth of tissue into foams implanted subcutaneously in SCID
mice after 30 days (data not shown). In contrast there was extensive tissue fill in foams implanted with umbilical-derived cells or placenta-derived cells (data not shown).
There was some tissue in growth in VICRYL non-woven scaffolds. Non-woven scaffolds seeded with umbilical cord- or placenta-derived cells showed increased matrix deposition and mature blood vessels (data not shown).
Summary. The purpose of this study was to determine the type of tissue formed by cells derived from human umbilical cord or placenta in scaffolds in immune deficient mice. Synthetic absorbable non-woven/foam discs (5.0 millimeter diameter x 1.0 millimeter thick) or self-assembling peptide hydrogel were seeded with either cells derived from human umbilical cord or placenta and implanted subcutaneously bilaterally in the dorsal spine region of SC1D mice. The present study demonstrates that postpartum-derived cells can dramatically increase good quality tissue formation in biodegradable scaffolds.

Chondro~enic and Osteo~enic Potential of Postpartum-Derived Cells on Implantation in SCID mice The chondrogenic potential of cells derived from umbilical cord or placenta tissue was evaluated following seeding on bioresorbable growth factor-loaded scaffolds and implantation into SC)D mice.
Materials & Methods Reagents. Dulbecco's Modified Essential Media (DMEM), Penicillin and Streptomycin, were obtained from Invitrogen, Carlsbad, CA. Fetal calf serum (FCS) was obtained from HyClone (Logan, UT). Mesenchymal stem cell growth medium (MSCGM) was obtained from Biowhittaker, Walkersville, MD. TGFbeta-3 was obtained from Oncogene research products, San Diego, CA. GDF-5 was obtained from Biopharm, Heidelberg, Germany (International PCT
Publication No. W096/01316 A1, U.S. Patent No. 5,994,094A). Chondrocyte growth medium comprised DMEM-High glucose supplemented with 10% fetal calf serum (FCS), 10 milliMolar HEPES, 0.1 milliMolar nonessential amino acids, 20 microgram/milliliter L-proline, 50 microgram/milliliter ascorbic acid, 100 Unit/milliliter penicillin, 100 microgram/milliliter streptomycin, and 0.25 microgram/milliliter arnphotericin B. Bovine fibrinogen was obtained from Calbiochem.
Cells. Human mesenchymal stem cells (hMSC, Lot# 2F1656) were obtained from Biowhittaker, Walkersville, MD and were cultured in MSCGM according to the manufacturer's instructions. This lot was tested in the laboratory previously in iii vitro experiments and was shown to be positive in the chondrogenesis assays. Human adult fibroblasts were obtained from American Type Culture Collection (ATCC), Manassas, VA and cultured in Growth Medium on gelatin-coated tissue culture plastic flasks. Postpartum-derived cells isolated from human umbilical cords (Lot# 022703Umb) and placenta (Lot# 071003P1ac) were prepared as previously described (Example 1). Cells were cultured in Growth medium on gelatin-coated tissue culture plastic flasks. The cell cultures were incubated in standard growth conditions. Cells used for experiments were at passages 5 and 14.
Scaffold. 65/35 Polyglycolic acid (PGA)/ Polycaprolactone (PCL) foam scaffolds [4x5 centimeters, lmillimeter thick, Ethylene Oxide (ETO) sterilized] reinforced with Polydioxanone (PDS) mesh (PGAIPCL foam-PDS mesh) were obtained from Center for Biomaterials and Advanced Technologies (CBAT, Somerville, NJ). Punches (3.5 millimeters) made from scaffolds were loaded with either GDF-5 (3.4 micrograms/scaffold), TGFbeta-3 (10 nanograms/scaffold), a combination of GDF-5 and TGFbeta-3, or control medium, and lyophilized.

Cell seeding on scaffolds. Placenta- and umbilical cord-derived cells were treated with trypsin, and cell number and viability was determined. 0.75x106 cells were resuspended in 15 microliter of Growth Medium and seeded onto 3.5 millimeter scaffold punches in a cell culture dish. The cell-seeded scaffold was incubated in a cell culture incubator in standard air with 5%
C02 at 37°C for 2 hours after which they were placed within cartilage explant rings.
Bovine Cartilage Explants. Cartilage explants 5 millimeter in diameter were made from cartilage obtained from young bovine shoulder. Punches (3 millimeter) were excised from the center of the explant and replaced with cells seeded 3.5 millimeter resorbable scaffold. Scaffolds with cells were retained within the explants using fibrin glue (60 microliter of bovine fibrinogen, 3 milligram/milliliter). Samples were maintained in chondrocyte growth medium overnight, rinsed in Phosphate Buffered Saline the following day, and implanted into SC1D
mice.
Animals. SCID mice ((Nlus fnusca~lc~s)lFox Chase SCID/Male), 5 weeks of age, were obtained from Harlan Sprague Dawley, Inc. (Indianapolis, Indiana) and Charles River Laboratories (Portage, Michigan). Animals used in the study were selected without any apparent systematic bias. A tag was placed on each individual animal cage listing the accession number, implantation technique, animal number, species/strain, surgery date, i~a vivo period, and date of euthanasia. The animals were identified by sequential numbers marked on the ear with an indelible ink marker.
Experimental Design. A total of 42 mice were tested. Two scaffolds were implanted subcutaneously in each mouse as described below; 42 mice for subcutaneous implantation; 28 treatments with n-value of 3 per treatment. The study corresponds to IACUC
Approval Number:
Skillman IACUC O1-037., The study lasted six weeks.
SLID Implantation.
A. Body Weights Each animal was weighed prior to being anesthetized and at necropsy.
B. Anesthesia and Sacrgical Preparatio~a:
All handling of the SLID mice occurred under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of KETASET (ketamine hydrochloride [60 milligram/kilogram]), ROMPUN (xylazine [10 milligrarn/kilogram]), 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 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 _127_ to prevent drying of the tissue during the anesthetic period. The anesthetized and surgically prepared animal was placed in the desired recumbent position.
C. Subcutaneous Irzaplarztatiorz Technique:
An approximate 2-cm skin incision was made just lateral to the thoracic spine parallel to the vertebral column. The skin was separated from the underlying connective tissue via blunt dissection. Each SC1D mouse received 2 treatments that were placed in subcutaneous pockets created by blunt dissection in each hemithorax through one skin incision.
Tacking sutures of 5-0 ETHIBOND EXCEL (polyester) were used to tack the skin to musculature around each scaffold to prevent subcutaneous migration. Scaffolds were implanted for 6 weeks and then harvested.
The experimental design is outlined in Table 19-1.
Table 19-1. Experimental Design:Treatment (N=3 per treatment) A. 65/35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, EP, TGFbeta3 B. 65!35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, EP, rhGDF-5 C. 65/35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, EP, rhGDF-5+TGFbeta3 D. 65/35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, EP, control E. 65/35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, LP, TGFbeta3 F. 65/35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, LP, rhGDF-5 G. 65/35 PGA/PCL Foam + PDS mesh cultured with Placenta-derived cells, LP, rhGDF
5+TGFbeta3 H. 65/35 PGA1PCL Foam + PDS mesh cultured with Placenta-derived cells, LP, control I. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, EP, TGFbeta3 J. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, EP, rhGDF-5 K. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, EP, rhGDF
5+TGFbeta3 L. 65/35 PGAIPCL Foam + PDS mesh cultured with Umbilical cord-derived cells, EP, control M. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, LP, TGFbeta3 N. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, LP, rhGDF-5 O. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, LP, rhGDF-5+TGFbeta3 P. 65/35 PGA/PCL Foam + PDS mesh cultured with Umbilical cord-derived cells, LP, control Q. 65/35 PGAIPCL Foam + PDS mesh cultured with hMSC, TGFbeta3 R. 65/35 PGAIPCL Foam + PDS mesh cultured with hMSC, rhGDF-5 S. 65/35 PGA/PCL Foam + PDS mesh cultured with hMSC, rhGDF-5+TGFbeta3 T. 65/35 PGA/PCL Foam + PDS mesh cultured with hMSC, control U. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult TGFbeta3 V. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult rhGDF-5 W. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult rhGDF-5+TGFbeta3 X. 65/35 PGA/PCL Foam + PDS mesh cultured with fibroblasts, Adult control Y. 65/35 PGA/PCL Foam + PDS mesh, TGFbeta3 Z. 65/35 PGA/PCL Foam + PDS mesh, rhGDF-5 AA. 65/35 PGA/PCL Foam + PDS mesh, rhGDF-5+TGFbeta3 BB. 65/35 PGA/PCL Foam + PDS mesh, control D. Necrops~~ a~zd Hi.stologic Pnepaf~atio~a Gross examination was performed on any animals that died during the course of the study or were euthanized in moribund condition. Selected tissues were saved at the discretion of the study director and/or pathologist.
Mice were euthanized by COZ inhalation at their designated intervals. Gross observations of the implanted sites were recorded. Samples of the subcutaneous implantation sites with their overlying skin were excised and fixed in 10% buffered formalin. Each implant was bisected into halves, and one half was sent to MPI Research (Mattawan, MI) for paraffin embedding, sectioning, and staining with Hematoxylin & Eosin (H&E) and Safranin O (SO).
The data obtained from this study were not statistically analyzed.
Resiclts New cartilage and bone formation was observed in the majority of the samples including growth factor-loaded, cell-seeded scaffolds, cell-seeded control scaffolds, and scaffolds loaded with growth factor alone. The extent of new cartilage and bone formation varied within the treatment and control groups.
Early and Late passage placenta-derived cell seeded scaffolds showed new cartilage and bone formation within the scaffolds. No obvious differences in new cauilage and bone formation was observed between the different growth factor-loaded, cell-seeded scaffolds and scaffolds seeded with cells alone. Compared to control scaffolds (without growth factors and without cells), it appeared that there was greater extent of new cartilage formation in cell-seeded scaffolds both with and without growth factors and in growth factor-loaded scaffolds alone.
New cartilage formation with placenta-derived cell-seeded scaffolds was similar to MSC- and fibroblast-seeded scaffolds.

In growth factor-treated and control scaffolds seeded with umbilical cord-derived cells at early and late passage, new cartilage and bone formation were observed. The extent of cartilage formation appeared to be less than that seen with placenta-derived cells. No one sample showed extensive cartilage formation as seen with the placenta-derived cells. Bone formation appeared to be higher in scaffolds seeded with umbilical cord-derived cells on scaffolds containing both TGFbeta-3 and rhGDF-5.
hMSC-loaded scaffolds also showed new cartilage and bone formation. The extent of new cartilage and bone formation was similar for all the hMSC treatment groups. Human adult fibroblast seeded scaffolds also demonstrated new cartilage and bone formation. Results were similar to those obtained with placenta-derived cells and hMSCs In the control group, in which growth factor-loaded scaffolds or scaffold alone were placed in cartilage rings and implanted, new cartilage and bone formation were also observed.
Not surprisingly, the extent of new cartilage formation was greater in scaffolds with growth factor than in scaffolds without growth factor. Increased bone formation was present in the control with the combination of the two tested growth factors.
New cartilage formation was observed adjacent to the cartilage explant rings as well as within the scaffolds. New cartilage formation within the scaffolds adjacent to the cartilage rings could be a result of chondrocyte migration. Cartilage formation seen as islands within the scaffolds may be a result of either migration of chondrocytes within the scaffolds, differentiation of seeded cells or differentiation of endogenous mouse progenitor cells. This observation stems from the fact that in control growth factor-loaded scaffolds with no seeded cells, islands of chondrogenic differentiation were observed. New bone formation was observed within the scaffolds independently and also associated with chondrocytes. Bone formation may have arisen from osteoblast differentiation as well as endochondral ossification.
It is difficult to separate new cartilage and bone formation associated with chondrocytes that migrated versus that from any chondrogenic and osteogenic differentiation of seeded cells that may have occurred. Staining of sections with specific human antibodies may distinguish the contribution of the seeded cells to the observed chondrogenesis and osteogenesis. It is also possible that placenta-derived cells and umbilical cord-derived cells stimulated chondrocyte migration.
Abundant new blood vessels were observed with the scaffolds loaded with placenta-derived cells and umbilical cord-derived cells. Blood vessels were abundant in areas of bone formation. New blood vessels were also observed within the hMSC- and fibroblast-seeded scaffolds associated with new bone formation.

Systemic effects of the adjacent scaffold (with growth factor (GF)) on the control scaffolds (no GF, no cells) on promoting new cartilage and bone formation cannot be ruled out.
Analysis of new cartilage and bone formation in scaffolds, taking into consideration the scaffolds implanted adjacent to it in SCID mice, showed no clear pattern of systemic effect of growth factor from the adjacent scaffold.
Summary. Results showed that new cartilage and bone formation were observed in growth factor and control scaffolds seeded with placenta- and umbilical cord-derived cells.
Results with placenta-derived cells were similar to that seen with human mesenchymal stem cells, while the extent of new cartilage like tissue formation was slightly less pronounced in umbilical cord-derived cells. Growth factor-loaded scaffolds implanted without cells also demonstrated new cartilage and bone formation. These data indicate that new cartilage formation within the scaffolds may arise from chondrocytes that migrated from the bovine explants, from chondrogenic differentiation of endogenous progenitor cells, and from chondrogenic differentiation of seeded cells.
These results suggest that placenta- and umbilical cord-derived cells undergo chondrogenic and osteogenic differentiation. These results also suggest that placenta- and umbilical cord-derived cells may promote migration of chondrocytes from the cartilage explant into the scaffolds. Abundant new blood vessels were also observed in the scaffolds especially associated with new bone formation.
While the present invention has been particularly sho~m and described with reference to the presently preferred embodiments, it is understood that the invention is not linuted to the embodiments specifically disclosed and exemplified herein. Numerous changes and modifications may be made to the preferred embodiment of the invention, and such changes and modifications may be made without departing from the scope and spirit of the invention as set forth in the appended claims.

CBATOD19.ST25.txt SEQUENCE LISTING
<110> Ethicon, Incorporated Kihm, Anthony J.
Seyda, Agnieszka Dhanaraj, Sridevi Wang, ziwei Harmon, Alexander M.
Harris, Ian Ross Messing, Darin J.
Mistry, Sanjay Yi, Chin-Feng Gosiewska, Anna <120> CARTILAGE AND BONE REPAIR AND REGENERATION USING
POSTPARTUM-DERIVED CELLS
<130> CBAT-0019 <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 <2l0> 4 <211> 21 <212> DNA
<213> Artificial Sequence <220>
<223> Synthetic Construct CBAT0019.ST25.txt <400> 4 gaattctcgg aatctctgtt g 21 <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 17 <210> 10 <211> 19 <212> DNA
<213> Artificial Sequence <220>

<223> Synthetic Construct CBAT0019.ST25.txt <400> 10 tcctgtcagt tggtgctcc 19

Claims (105)

What is Claimed:
1. A postpartum-derived cell comprising a cell derived from human postpartum tissue substantially free of blood, wherein said cell is capable of self-renewal and expansion in culture and has the potential to differentiate into a cell of an osteogenic or chondrogenic phenotype;
wherein said cell requires L-valine for growth; wherein said cell is capable of growth in about 5% to about 20% oxygen; wherein said cell further comprises at least one of the following characteristics:
(a) production of at least one of granulocyte chemotactic protein 2 (GCP-2), reticulon 1, tissue factor, vimentin, and alpha-smooth muscle actin;
(b) lack of production of at least one of GRO-alpha or oxidized low density lipoprotein receptor, as detected by flow cytometry;
(c) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
(d) 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;
(e) expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of interleukin 8;
reticulon 1; chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha);
chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 2);
chemokine (C-X-C
motif) ligand 3; and tumor necrosis factor, alpha-induced protein 3 or expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of C-type lectin superfamily member A2, Wilms tumor 1, aldehyde dehydrogenase 1 family member A2, renin, oxidized low density lipoprotein receptor 1, protein kinase C zeta, clone IMAGE:4179671, hypothetical protein DKFZp564F013, downregulated in ovarian cancer 1, and clone DKFZp547K1113;
(f) 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; cDNA DKFZp586M2022 (from clone DKFZp586M2022);

mesenchyme homeobox 2; sine oculis homeobox homolog 1; crystallin, alpha B;
dishevelled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1;
tetranectin; 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; hypothetical gene BC008967; collagen, type VIII, alpha 1;
tenascin C;
iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2;
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; 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); cDNA clone EUROIMAGE 1968422; EphA3;
KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C
(atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; cDNA DKFZp564B222 (from clone DKFZp564B222); vesicle-associated membrane protein 5; 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;
(g) secretion of at least one of monocyte chemotactic protein-1, interleukin(IL)-6, IL-8, granulocyte chemotactic protein-2, hepatocyte growth factor, keratinocyte growth factor, fibroblast growth factor, heparin binding-epidermal growth factor, brain derived neurotrophic factor, thrombopoietin, macrophage inflammatory protein (MIP)-1a, RANTES, and tissue inhibitor of matrix metalloprotease 1;
(h) lack of secretion of at least one of transforming growth factor-beta2, angiopoetin-2, platelet derived growth factor-bb, MIP1b, I309, macrophage-derived chemokine, and vascular endothelial growth factor, as detected by ELISA; and (i) the ability to undergo at least 40 population doublings in culture.
2. The cell of claim 1 which has been isolated from a post-partum placenta or fragment thereof by enzymatic dissociation with at least one of a matrix metalloprotease, a neutral protease, and a mucolytic enzyme that digests hyaluronic acid.
3. A method of inducing differentiation of a postpartum-derived cell of claim 1 to a chondrogenic phenotype comprising exposing said cell to one or more chondrogenic differentiation-inducing agents.
4. The method of claim 3 wherein said chondrogenic differentiation-inducing agent comprises at least one of transforming growth factor-beta3 (TGFbeta3) and growth and differentiation factor-5(GDF-5).
5. The method of claim 3 further comprising culturing the cell in chondrogenic medium.
6. The method of claim 5 wherein said chondrogenic medium comprises Dulbecco's modified Eagle's medium, L-glutamine, sodium pyruvate, L-praline, dexamethasone, L-ascorbic acid, insulin, transferrin, selenium, and an antibiotic agent.
7. The method of claim 6 wherein said chondrogenic medium further comprises at least one of collagen and sodium hydroxide.
8. The method of claim 3 further comprising evaluating differentiation of said cell by a pellet culture assay.
9. The method of claim 3 further comprising evaluating differentiation of said cell by detecting the presence of a glycosaminoglycan or collagen.
10. The method of claim 9 wherein said step of evaluating comprises staining said cell with Safranin-O or hematoxylin/eosin.
11. The cell produced by the method of claim 3.
12. A method of inducing differentiation of a postpartum-derived cell of claim 1 to an osteogenic phenotype comprising exposing said cell to one or more osteogenic differentiation-inducing agents.
13. The method of claim 12 wherein said differentiation-inducing agent comprises at least one of bone morphogenic protein (BMP)-2, BMP-4, and transforming growth factor-betal.
14. The method of claim 12 further comprising culturing said cell in osteogenic medium.
15. The method of claim 14 wherein said osteogenic medium comprises Dulbecco's modified Eagle's medium-low glucose, serum, beta-glycerophosphate, dexamethasone, ascorbic phosphate salt, and at least one antibiotic or antimycotic agent.
16. The method of claim 12 further comprising evaluating said differentiation by detecting an osteogenic lineage-specific marker.
17. The method of claim 16 wherein said marker is osteocalcin, bone sialoprotein, or alkaline phosphatase.
18. The method of claim 12 further comprising detecting said differentiation by measuring mineralization.
19. The method of claim 18 wherein said step of detecting comprises von Kossa staining.
20. The cell produced according to the method of claim 12.
21. A cell population comprising the postpartum-derived cell of claim 1.
22. The cell population of claim 21 wherein said cell population is substantially homogeneous.
23. The cell population of claim 21 wherein said cell population is heterogeneous.
24. The cell population of claim 23 further comprising at least one cell type of bone marrow cells, chondrocytes, chondroblasts, chondrocyte progenitor cells, or stem cells.
25. A cell population comprising the postpartum-derived cell of claim 12.
26. The cell population of claim 25 wherein said cell population is substantially homogeneous.
27. The cell population of claim 25 wherein said cell population is heterogeneous.
28. The cell population of claim 27 further comprising at least one cell type of bone marrow cells, chondrocytes, chondroblasts, chondrocyte progenitor cells, stem cells, or other pluripotent or multipotent cell.
29. A cell population comprising the postpartum-derived cell of claim 20.
30. The cell population of claim 29 wherein said cell population is substantially homogeneous.
31. The cell population of claim 29 wherein said cell population is heterogeneous.
32. The cell population of claim 31 further comprising at least one cell type of bone marrow cells, osteocytes, osteoblasts, osteoclasts, bone lining cells, stem cells or other pluripotent or multipotent cell.
33. A cell lysate prepared from the cell population of claim 21.
34. A soluble cell fraction prepared from the cell lysate of claim 33.
35. A cell lysate prepared from the cell population of claim 25.
36. A soluble cell fraction prepared from the cell lysate of claim 35.
37. A cell lysate prepared from the cell population of claim 29.
38. A soluble cell fraction prepared from the cell lysate of claim 37.
39. An extracellular matrix of the cell population of claim 21.
40. An extracellular matrix of the cell population of claim 25.
41. An extracellular matrix of the cell population of claim 29.
42. A composition comprising the cell population of claim 21 and one or more bioactive factors.
43. A composition comprising the cell population of claim 25 and one or more bioactive factors.
44. A composition comprising the cell population of claim 29 and one or more bioactive factors.
45. The composition of claim 42 wherein said bioactive factor is a chondrogenic differentiation-inducing factor.
46. The composition of claim 42 wherein said bioactive factor is an osteogenic differentiation-inducing factor.
47. A pharmaceutical composition comprising a cell of claim 1 and a pharmaceutically acceptable carrier.
48. A pharmaceutical composition comprising a cell of claim 11 and a pharmaceutically acceptable carrier.
49. A pharmaceutical composition comprising a cell of claim 20 and a pharmaceutically acceptable carrier.
50. A pharmaceutical composition comprising the extracellular matrix of claim 39 and a pharmaceutically acceptable carrier.
51. A pharmaceutical composition comprising the extracellular matrix of claim 40 and a pharmaceutically acceptable carrier.
52. A pharmaceutical composition comprising the extracellular matrix of claim 41 and a pharmaceutically acceptable earner.
53. A pharmaceutical composition comprising the lysate of claim 33 and a pharmaceutically acceptable carrier.
54. A pharmaceutical composition comprising the lysate of claim 35 and a pharmaceutically acceptable carrier.
55. A pharmaceutical composition comprising the lysate of claim 37 and a pharmaceutically acceptable carrier.
56. A cell culture comprising at least one cell of claim 1 in a culture medium.
57. The cell culture of claim 56 wherein said culture medium comprises chondrogenic medium or osteogenic medium.
58. The cell culture of claim 56 further comprising at least one chondrogenic differentiation-inducing agent.
59. The cell culture of claim 58 wherein said chondrogenic differentiation-inducing agent is at least one of transforming growth factor-beta3 or growth and differentiation factor-5.
60. The cell culture of claim 56 further comprising at least one osteogenic differentiation-inducing agent.
61. The cell culture of claim 60 wherein said osteogenic differentiation-inducing agent is at least one of transforming growth factor-beta 1, bone morphogenic protein (BMP)-2, or BMP4.
62. A matrix comprising a cell population of claim 21.
63. A matrix comprising a cell population of claim 25.
64. A matrix comprising a cell population of claim 29.
65. The matrix of claim 62 wherein said matrix comprises a three-dimensional scaffold.
66. The matrix of claim 63 wherein said matrix comprises a three-dimensional scaffold.
67. The matrix of claim 64 wherein said matrix comprises a three-dimensional scaffold.
68. A method of treating a condition in a patient comprising administering to said patient one or more postpartum-derived cells of claim 1.
69. The method of claim 68 wherein said condition is a bone or cartilage condition.
70. The method of claim 69 wherein said bone or cartilage condition is a congenital bone or cartilage defect, meniscal injury or defect, bone/spinal deformation, osteosarcoma, myeloma, bone dysplasia or scoliosis, osteoporosis, periodontal disease, dental bone loss, osteomalacia, rickets, fibrous osteitis, renal bone dystrophy, spinal fusion, spinal disc reconstruction or removal, Paget's disease of bone, rheumatoid arthritis, osteoarthritis, or a traumatic or surgical injury.
71. The method of claim 68 wherein said postpartum-derived cells are administered with at least one other cell type.
72. The method of claim 71 wherein said at least one other cell type is bone marrow cells, chondrocytes, chondroblasts, chondrocyte progenitor cells, osteocytes, osteoblasts, osteoclasts, bone lining cells, stem cells, or other pluripotent or multipotent cell.
73. The method of claim 68 wherein said postpartum-derived cells are inoculated on a matrix.
74. The method of claim 73 wherein said matrix is implanted into said patient.
75. The method of claim 68 wherein said postpartum-derived cells are induced to differentiate to a chondrogenic or osteogenic phenotype prior to said step of administering.
76. The method of claim 68 wherein said postpartum-derived cells are co-administered with at least one bioactive factor.
77. The method of claim 68 wherein said cells are administered to a bone of said patient.
78. The method of claim 68 wherein said cells are administered to cartilage of said patient.
79. A method of regenerating a tissue in a patient in need thereof comprising administering the cell population of claim 21 to said patient.
80. The method of claim 79 wherein said tissue is bone or cartilage.
81. A method of regenerating a tissue in a patient in need thereof comprising administering the cell population of claim 25 to said patient.
82. The method of claim 81 wherein said tissue is bone or cartilage.
83. A method of regenerating a tissue in a patient in need thereof comprising administering the cell population of claim 29 to said patient.
84. The method of claim 83 wherein said tissue is bone or cartilage.
85. The method of claim 68 wherein said cells are implanted into said patient.
86. A conditioned medium generated by the growth of the culture of claim 56.
87. A method for identifying a compound that stimulates chondrogenesis of a postpartum-derived cell comprising contacting a cell of claim 1 with said compound and monitoring said cell for a marker of chondrogenesis.
88. A method for identifying a compound that stimulates osteogenesis of a postpartum-derived cell comprising contacting a cell of claim 1 with said compound and monitoring said cell for a marker of osteogenesis.
89. A method for identifying a compound that is toxic to a postpartum-derived cell of claim 1 comprising contacting said cell with said compound and monitoring survival of said cell.
90. A kit composing at least one cell of claim 1 and at least one additional component of a matrix, a hydrating agent, a cell culture substrate, a differentiation-inducing agent, and cell culture media.
91. The kit of claim 90 wherein said matrix is a three-dimensional scaffold.
92. The kit of claim 91 wherein said cell is seeded on said scaffold.
93. The kit of claim 90 wherein said differentiation-inducing agent is an osteogenic differentiation-inducing agent or a chondrogenic differentiation-inducing agent.
94. A method of treating a patient having a bone or cartilage condition comprising administering to said patient the extracellular matrix of a cell of claim 1.
95. A method of heating a patient having a bone or cartilage condition comprising administering to said patient the cell lysate of claim 33.
96. A method of treating a patient having a bone or cartilage condition comprising administering to said patient the conditioned medium of claim 86.
97. The pharmaceutical composition of claim 47 wherein said composition comprises an effective amount of said cells to treat a bone or cartilage condition.
98. The pharmaceutical composition of claim 48 wherein said composition comprises an effective amount of said cells to treat a bone or cartilage condition.
99. The pharmaceutical composition of claim 49 wherein said composition comprises an effective amount of said cells to treat a bone or cartilage condition.
100. The pharmaceutical composition of claim 50 wherein said composition comprises an effective amount of said extracellular matrix to treat a bone or cartilage condition.
101. The pharmaceutical composition of claim 51 wherein said composition comprises an effective amount of said extracellular matrix to treat a bone or cartilage condition.
102. The pharmaceutical composition of claim 52 wherein said composition comprises an effective amount of said extracellular matrix to treat a bone or cartilage condition.
103. The pharmaceutical composition of claim 53 wherein said composition comprises an effective amount of said lysate to treat a bone or cartilage condition.
104. The pharmaceutical composition of claim 54 wherein said composition comprises an effective amount of said lysate to treat a bone or cartilage condition.
105. The pharmaceutical composition of claim 55 wherein said composition comprises an effective amount of said lysate to treat a bone or cartilage condition.
106. The pharmaceutical composition of claim 47 further comprising at least one other cell type of stem cells, bone marrow cells, chondrocytes, chondroblasts, osteocytes, osteoblasts, osteoclasts, bone lining cells, and other bone or cartilage progenitor cells.
107. The pharmaceutical composition of claim 48 further comprising at least one other cell type of stem cells, bone marrow cells, chondrocytes, chondroblasts, osteocytes, osteoblasts, osteoclasts, bone lining cells, and other bone or cartilage progenitor cells.
105. The pharmaceutical composition of claim 49 further comprising at least one other cell type of stem cells, bone marrow cells, chondrocytes, chondroblasts, osteocytes, osteoblasts, osteoclasts, bone lining cells, and other bone or cartilage progenitor cells.
CA2530412A 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells Expired - Fee Related CA2530412C (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/US2004/020958 WO2005038012A2 (en) 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells

Publications (2)

Publication Number Publication Date
CA2530412A1 true CA2530412A1 (en) 2005-04-28
CA2530412C CA2530412C (en) 2016-02-09

Family

ID=38456561

Family Applications (7)

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
CA2530732A Expired - Fee Related CA2530732C (en) 2003-06-27 2004-06-25 Regeneration and repair of neural 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
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
CA2530255A Expired - Fee Related CA2530255C (en) 2003-06-27 2004-06-25 Soft tissue repair and regeneration 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
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 (6)

Application Number Title Priority Date Filing Date
CA2530732A Expired - Fee Related CA2530732C (en) 2003-06-27 2004-06-25 Regeneration and repair of neural 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
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
CA2530255A Expired - Fee Related CA2530255C (en) 2003-06-27 2004-06-25 Soft tissue repair and regeneration 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
CA2530421A Expired - Fee Related CA2530421C (en) 2003-06-27 2004-06-25 Repair and regeneration of ocular tissue using postpartum-derived cells

Country Status (8)

Country Link
US (28) US8703121B2 (en)
EP (14) EP1641917B1 (en)
JP (7) JP4950659B2 (en)
AU (7) AU2004252568B2 (en)
CA (7) CA2530412C (en)
ES (14) ES2564044T3 (en)
PL (14) PL2338982T3 (en)
WO (7) WO2005001077A2 (en)

Families Citing this family (536)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US20080077251A1 (en) * 1999-06-07 2008-03-27 Chen Silvia S Cleaning and devitalization of cartilage
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
IL156303A0 (en) * 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
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
EP2316463B1 (en) 2001-02-14 2017-04-12 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making them
EP2314673B1 (en) * 2001-02-14 2013-07-24 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
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
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
US20040078090A1 (en) 2002-10-18 2004-04-22 Francois Binette Biocompatible scaffolds with tissue fragments
US7824701B2 (en) * 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
KR20100125479A (en) * 2002-11-26 2010-11-30 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US20090169597A1 (en) * 2007-12-27 2009-07-02 Ethicon, Incorporated Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
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
BRPI0407221A (en) 2003-02-11 2006-01-31 John E Davies Wharton Jelly Extract, Method for Obtaining a Human Parent Cell, Methods for Producing a Cell Population, Cell Populations, and Method for Producing Bone Tissue
US20040219136A1 (en) * 2003-02-13 2004-11-04 Hariri Robert J Use of umbilical cord blood to treat individuals having a disease, disorder or condition
US8197837B2 (en) 2003-03-07 2012-06-12 Depuy Mitek, Inc. Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
JP2006524092A (en) * 2003-04-24 2006-10-26 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Non-invasive left ventricular volume determination
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US8491883B2 (en) * 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US8703121B2 (en) * 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
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
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
US7485310B2 (en) * 2003-08-12 2009-02-03 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
AU2004296765B2 (en) * 2003-12-02 2011-03-24 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
AU2005227295A1 (en) 2004-03-22 2005-10-06 Osiris Therapeutics, Inc. 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
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
CN101080486B (en) * 2004-04-23 2012-05-16 佰欧益股份有限公司 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
US20060083732A1 (en) * 2004-06-30 2006-04-20 Arlene Gwon Hyaluronic acid in the enhancement of lens regeneration
DK1789534T3 (en) 2004-08-16 2014-11-17 Cellres Corp Pte Ltd ISOLATION OF stem / progenitor cells FOSTER HINDE FROM an 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
US7799754B2 (en) * 2004-10-14 2010-09-21 Biomimetic Therapeutics, Inc. Compositions and methods for treating bone
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
US20060171930A1 (en) * 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
WO2006083394A2 (en) * 2004-12-21 2006-08-10 Ethicon, Inc. Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
WO2006071773A2 (en) * 2004-12-23 2006-07-06 Ethicon Incoporated Treatment of osteochondral diseases using postpartum-derived cells and products thereof
JP5340599B2 (en) * 2004-12-23 2013-11-13 エシコン・インコーポレイテッド Umbilical tissue-derived postpartum cells and methods for producing and using the same
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
CN103356705B (en) 2005-03-31 2015-09-30 斯丹姆涅恩有限公司 Prepare the method for the medicine in order to treat burn wound
US20060222634A1 (en) 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US8153430B2 (en) * 2005-03-31 2012-04-10 Stemnion, Inc. Methods related to surgery
WO2006110806A2 (en) * 2005-04-12 2006-10-19 The Trustees Of The University Of Pennsylvania Multipotent adult stem cells
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
DK1879606T3 (en) * 2005-04-25 2013-09-23 Massachusetts Inst Technology Self-organizing peptides to promote hemostasis
AU2006202209B2 (en) * 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
ES2400916T3 (en) * 2005-06-08 2013-04-15 Janssen Biotech, 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
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
WO2007033180A1 (en) 2005-09-12 2007-03-22 Abela Pharmaceuticals, Inc. Materials for facilitating administration of dimethyl sulfoxide (dmso) and related compounds
WO2007033082A2 (en) 2005-09-12 2007-03-22 Abela Pharmaceuticals, Inc. Compositions comprising dimethyl sulfoxide (dmso)
JP2007106760A (en) * 2005-09-16 2007-04-26 Kenji Yoshida Hematopoietic stem cell proliferator
US8182841B2 (en) 2005-09-27 2012-05-22 Tissue Tech, Inc. Amniotic membrane preparations and purified compositions and anti-inflammation methods
MX343814B (en) 2005-10-13 2016-11-24 Anthrogenesis Corp Immunomodulation using placental stem cells.
KR20080056302A (en) * 2005-10-13 2008-06-20 안트로제네시스 코포레이션 Production of oligodendrocytes from placenta-derived stem cells
JP4921767B2 (en) * 2005-10-14 2012-04-25 株式会社カネカ Cell differentiation induction method
EP1945794A2 (en) * 2005-11-09 2008-07-23 Chemimage Corporation System and method for cytological analysis by raman spectroscopic imaging
US10117900B2 (en) * 2005-11-09 2018-11-06 Athersys, Inc. MAPC treatment of brain injuries and diseases
US11000546B2 (en) 2005-11-09 2021-05-11 Athersys, Inc. Immunomodulatory properties of MAPCs and uses thereof
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
BRPI0618794A2 (en) * 2005-11-17 2011-09-13 Biomimetic Therapeutics Inc use of a biocompatible matrix, kit and composition for bone augmentation, especially for maxillofacial bone augmentation
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
ES2391034T3 (en) * 2005-12-19 2012-11-20 Ethicon, Inc. In vitro expansion of postpartum derived cells in rotary bottles
CA2634820C (en) 2005-12-22 2021-05-25 Jane Ennis Viable cells from frozen umbilical cord tissue
US20070160588A1 (en) * 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
CN105106239A (en) * 2005-12-28 2015-12-02 伊西康公司 Treatment of peripheral vascular disease using postpartum-derived cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
CN101374941A (en) * 2005-12-29 2009-02-25 人类起源公司 Improved composition for collecting and preserving placental stem cells and methods of using the composition
PT2471905T (en) 2005-12-29 2019-01-11 Celularity Inc Placental stem cell populations
US8241902B2 (en) 2006-01-11 2012-08-14 Technion Research & Development Foundation Ltd. Preparation of adult stem cell-derived connective tissue progenitors
US20070253931A1 (en) * 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
US20080286249A1 (en) * 2006-01-12 2008-11-20 Varney Timothy R Use of mesenchymal stem cells for treating genetic diseases and disorders
GB0600972D0 (en) * 2006-01-18 2006-03-01 Univ Leeds Enrichment of cells
US9944900B2 (en) * 2006-01-18 2018-04-17 Hemacell Perfusion Pulsatile perfusion extraction method for non-embryonic pluripotent stem 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
CA3028279C (en) 2006-01-23 2021-08-03 Athersys, Inc. 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
CA2644508A1 (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
US20090232777A1 (en) * 2006-03-01 2009-09-17 Evy Lundgren-Akerlund Expansion and Differentiation of Mesenchymal Stem Cells
ES2537641T3 (en) * 2006-03-23 2015-06-10 Pluristem Ltd. Methods of cell expansion and uses of cells and conditioned media produced in this way for therapy
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
EP2019858B1 (en) * 2006-04-17 2012-06-13 BioE LLC Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US20090208466A1 (en) * 2006-04-21 2009-08-20 James Yoo 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
EP2010100B1 (en) 2006-04-27 2018-11-28 Johnson & Johnson Surgical Vision, Inc. Materials comprising polysiloxane polymers for enhancement of lens regeneration
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
BRPI0711599B8 (en) * 2006-05-11 2021-07-27 Hli Cellular Therapeutics Llc methods for collecting and using stem cells from placental umbilical cord blood
US20080064098A1 (en) * 2006-06-05 2008-03-13 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
KR20090031895A (en) * 2006-06-09 2009-03-30 안트로제네시스 코포레이션 Placental niche and use thereof to culture stem cells
EP2035093A4 (en) * 2006-06-14 2010-02-17 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
EP2079829A2 (en) * 2006-06-28 2009-07-22 The University Of Kansas Differentiation of stem cells from umbilical cord matrix into hepatocyte lineage cells
WO2008005427A2 (en) 2006-06-30 2008-01-10 Biomimetic Therapeutics, Inc. Pdgf-biomatrix compositions and methods for treating rotator cuff injuries
US9161967B2 (en) 2006-06-30 2015-10-20 Biomimetic Therapeutics, Llc Compositions and methods for treating the vertebral column
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
US7736485B2 (en) * 2006-08-11 2010-06-15 Aqua Resources Corporation Nanoplatelet magnesium 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
CA2673521C (en) 2006-10-12 2017-01-17 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
WO2008051568A2 (en) * 2006-10-23 2008-05-02 Anthrogenesis Corporation 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
ES2524443T3 (en) * 2006-11-13 2014-12-09 DePuy Synthes Products, LLC In vitro expansion of postpartum 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
KR20090086260A (en) * 2006-12-07 2009-08-11 테바 파마슈티컬 인더스트리즈 리미티드 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
WO2008083223A1 (en) * 2006-12-28 2008-07-10 Isolagen Technologies, Inc. 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
EP2129775A1 (en) * 2007-02-12 2009-12-09 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
CN103356711A (en) 2007-02-12 2013-10-23 人类起源公司 Immunomodulation using placental stem cells
AU2008218763B2 (en) * 2007-02-20 2013-10-24 Biomimetic Therapeutics, Llc. Prevention and treatment for osteonecrosis and osteoradionecrosis of the jaw using PDGF and a bone matrix
JP2010530214A (en) * 2007-03-01 2010-09-09 クライオ−セル インターナショナル インコーポレイテッド Acquisition, isolation and cryopreservation of endometrial / menstrual cells
WO2008109816A1 (en) * 2007-03-08 2008-09-12 Hemacell Perfusion, Inc. Method for isolation of afterbirth derived cells
US20100172830A1 (en) * 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
EP2626416A3 (en) * 2007-04-07 2013-12-18 The Whitehead Institute for Biomedical Research Reprogramming of somatic cells
WO2009008928A2 (en) * 2007-04-13 2009-01-15 Stemnion, Inc. Methods for treating nervous system injury and disease
EP2137300B1 (en) 2007-04-26 2011-10-26 Ramot at Tel-Aviv University Ltd. Pluripotent autologous stem cells from oral or gastrointestinal mucosa
US8574567B2 (en) 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
EP2607477B1 (en) 2007-05-03 2020-09-23 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
CN101842105B (en) 2007-05-21 2013-06-19 韦克福里斯特大学健康科学院 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
EP1997457B1 (en) * 2007-06-01 2010-11-10 Allergan, Inc. Biological tissue growth support through 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
CA2688504A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US20090004253A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Composite device for the repair or regeneration of tissue
US20090004271A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Morselized foam for wound treatment
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US9095562B2 (en) 2007-07-05 2015-08-04 Regenerative Sciences, Inc. Methods and compositions for optimized expansion and implantation of mesenchymal stem cells
DE102007034679A1 (en) * 2007-07-25 2009-01-29 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Material compositions containing adult stem cells derived from exocrine glandular tissue, especially for use in regenerative medicine, e.g. to restore injured or damaged myocardial tissue
US9744043B2 (en) 2007-07-16 2017-08-29 Lifenet Health Crafting of cartilage
WO2009015343A2 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of multi-lineage progenitor cells to chondrocytes
ES2451441T3 (en) 2007-07-27 2014-03-27 Humacyte, Inc. Compositions comprising human collagen and human elastin and uses thereof
CA2695225C (en) 2007-07-31 2021-06-01 Lifescan, Inc. Differentiation of human embryonic stem cells to pancreatic endocrine
CA2695863A1 (en) * 2007-08-08 2009-02-12 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
EP3689421A1 (en) 2007-09-07 2020-08-05 MiMedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
RU2010109698A (en) * 2007-09-19 2011-09-27 Плуристем Лтд. (Il) ADHESIVE CELLS OF FATTY TISSUE OR PLACENTA AND THEIR USE FOR MEDICAL PURPOSES
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
ES2719931T3 (en) 2007-09-28 2019-07-16 Celularity Inc Tumor suppression using human placental perfusate and intermediate natural killer cells that come from human placenta
CN102036688B (en) 2007-10-05 2014-07-02 伊西康公司 Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
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
US20100297234A1 (en) * 2007-10-19 2010-11-25 Ilene Sugino Method of using an extracellular matrix to enhance cell transplant survival and differentiation
KR20170116221A (en) * 2007-11-07 2017-10-18 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
WO2009064958A1 (en) * 2007-11-14 2009-05-22 Osteosphere, Llc Ex-vivo production of human demineralized bone matrix
US9062290B2 (en) 2007-11-27 2015-06-23 Lifescan, Inc. Differentiation of human embryonic stem cells
US20090163990A1 (en) * 2007-12-19 2009-06-25 Chunlin Yang Decellularized omentum matrix and uses thereof
WO2009085969A2 (en) * 2007-12-19 2009-07-09 Regenerative Sciences, Llc 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
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
AU2009212151C1 (en) 2008-02-07 2015-09-17 Biomimetic Therapeutics, Llc Compositions and methods for distraction osteogenesis
EP2254608B1 (en) 2008-02-07 2016-05-04 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
BR122017025207B1 (en) 2008-02-21 2021-03-16 Centocor Ortho Biotech Inc surface that is part of a container or matrix intended for use in a cell culture or analysis, devoid of a layer of feeder cells and devoid of an adsorbent layer
US8318485B2 (en) * 2008-02-25 2012-11-27 Natalie Gavrilova Stem cell therapy for the treatment of diabetic retinopathy and diabetic optic neuropathy
US20090220995A1 (en) 2008-02-28 2009-09-03 Sachs David H Multiple administrations of umbilicus derived cells
US20090232782A1 (en) * 2008-03-14 2009-09-17 Yu-Show Fu Method for treating brain ischemic injury through transplantation of human umbilical mesenchymal stem cells
CN102026546A (en) * 2008-03-14 2011-04-20 再生科学有限责任公司 Compositions and methods for cartilage repair
KR20170005148A (en) 2008-04-21 2017-01-11 티슈 리제너레이션 쎄라퓨틱스, 인코포레이티드 Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US9480549B2 (en) 2008-04-25 2016-11-01 Allosource Multi-layer tissue patches
US9358320B2 (en) 2008-04-25 2016-06-07 Allosource Multi-layer tissue patches
JP2011519616A (en) * 2008-04-30 2011-07-14 エシコン・インコーポレイテッド Blood vessels by tissue engineering
US20110143429A1 (en) * 2008-04-30 2011-06-16 Iksoo Chun Tissue engineered blood vessels
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
US8748181B2 (en) * 2008-05-23 2014-06-10 President And Fellows Of Harvard College Methods of generating patterned soft substrates and uses thereof
US20090297485A1 (en) * 2008-05-28 2009-12-03 Allan Mishra Compositions and methods for treating psychiatric and neurodegenerative disorders
AU2009257400B2 (en) * 2008-06-11 2014-05-01 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
US9497943B2 (en) 2008-06-13 2016-11-22 Whitehead Institute For Biomedical Research Nucleic acid constructs encoding reprogramming factors linked by self-cleaving peptides
MX2010014304A (en) * 2008-06-26 2011-02-15 Kci Licensing Inc Stimulation of cartilage formation using reduced pressure treatment and chondrocytes.
JP5294041B2 (en) * 2008-06-27 2013-09-18 独立行政法人産業技術総合研究所 Pancreatic cell regenerative transplant kit for pancreatic disease or diabetes
EP2318516A1 (en) * 2008-06-30 2011-05-11 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
CA2734237C (en) * 2008-08-20 2019-07-02 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
WO2010021714A2 (en) 2008-08-20 2010-02-25 Anthrogenesis Corporation Improved cell composition and methods of making the same
MX2011001992A (en) 2008-08-22 2011-03-29 Anthrogenesis Corp Methods and compositions for treatment of bone defects with placental cell populations.
EP3115451A1 (en) 2008-09-02 2017-01-11 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
US20100069827A1 (en) * 2008-09-12 2010-03-18 Barry Neil Silberg Pre-Surgical Prophylactic Administration of Antibiotics and Therapeutic Agents
US9446227B2 (en) 2008-09-12 2016-09-20 Sonescence, Inc. Ultrasonic dispersion of compositions in tissue
EP3238749B1 (en) 2008-10-06 2018-09-19 3-D Matrix Ltd. Tissue plug
US20110300203A1 (en) * 2008-10-22 2011-12-08 Trustees Of Columbia University In The City Of New York Cartilage regeneration without cell transplantation
CN107435038B (en) 2008-10-31 2021-07-09 詹森生物科技公司 Differentiation of human embryonic stem cells into the pancreatic endocrine lineage
CN107904201B (en) 2008-10-31 2021-11-16 詹森生物科技公司 Differentiation of human embryonic stem cells into the pancreatic endocrine lineage
CA2741426C (en) 2008-10-31 2020-04-21 Meredith Hans Method and device 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.
RU2015130665A (en) 2008-11-19 2018-12-24 Антродженезис Корпорейшн AMNIOTIC ADHESIVE CELLS
KR101837080B1 (en) 2008-11-20 2018-03-09 얀센 바이오테크 인코포레이티드 Pluripotent stem cell culture on micro-carriers
BRPI0921996A2 (en) 2008-11-20 2015-08-18 Centocor Ortho Biotech Inc Methods and compositions for culturing and binding cells on flat substrates.
CA2740803A1 (en) * 2008-11-20 2010-05-27 Lifecell Corporation Method for treatment and prevention of parastomal hernias
NZ592839A (en) * 2008-11-21 2012-10-26 Anthrogenesis Corp Treatment of diseases, disorders or conditions of the lung using placental cells
US20110245804A1 (en) 2008-12-05 2011-10-06 Regenerative Sciences, Llc Methods and Compositions to Facilitate Repair of Avascular Tissue
WO2010065239A1 (en) * 2008-12-05 2010-06-10 Wake Forest University Health Sciences Stem cells from urine and methods for using the same
WO2010064702A1 (en) * 2008-12-05 2010-06-10 国立大学法人 東京大学 Biomarker for predicting prognosis of cancer
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
DK2379087T3 (en) 2008-12-19 2014-11-10 Depuy Synthes Products Llc Cells derived from umbilical cord tissue for the treatment of neuropathic pain and spasticity
AU2009327384B2 (en) * 2008-12-19 2014-07-10 DePuy Synthes Products, LLC Treatment of lung and pulmonary diseases and disorders
US20130302283A1 (en) 2012-05-14 2013-11-14 Advanced Technologies And Regenerative Medicine, Llc hUTC MODULATION OF PRO-INFLAMMATORY MEDIATORS OF LUNG AND PULMONARY DISEASES AND DISORDERS
US10179900B2 (en) * 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
AU2009327383B2 (en) * 2008-12-19 2014-08-28 DePuy Synthes Products, LLC Regeneration and repair of neural tissue following 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
CA2746515A1 (en) * 2008-12-30 2010-07-08 Kci Licensing, Inc. Reduced pressure augmentation of microfracture procedures for cartilage repair
TWI440719B (en) * 2009-01-23 2014-06-11 Univ Tokyo Method for evaluation of cultured cells
US9247943B1 (en) 2009-02-06 2016-02-02 Kleiner Intellectual Property, Llc Devices and methods for preparing an intervertebral workspace
US20120142603A1 (en) * 2009-02-12 2012-06-07 University Of Southern California Bioadhesive patch for sutureless closure of soft tissue
KR101422690B1 (en) * 2009-02-27 2014-07-23 (주)차바이오앤디오스텍 Composition for skin regeneration by using medium or secretion of embryonic stem cell derived endothelial progenitor cells and use thereof
EP2411504B1 (en) * 2009-03-26 2017-05-10 DePuy Synthes Products, Inc. Human umbilical cord tissue cells as therapy for alzheimer's disease
EP2411505A4 (en) * 2009-03-26 2013-01-30 Univ California Mesenchymal stem cells producing inhibitory rna for disease modification
US20120039857A1 (en) * 2009-04-06 2012-02-16 Capricor, Inc. Systems and methods for cardiac tissue repair
MY153917A (en) * 2009-05-13 2015-04-15 Medipost Co Ltd Tsp-1, tsp-2, il-17br and hb-egf associated with stem cell activities and applications thereof
EP2432495B1 (en) * 2009-05-20 2017-03-22 Humacyte, Inc. Elastin for soft tissue augmentation
US20120128639A1 (en) * 2009-05-28 2012-05-24 University Of Central Florida Research Foundation, Inc. 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
WO2011011302A2 (en) 2009-07-20 2011-01-27 Centocor Ortho Biotech Inc. Differentiation of human embryonic stem cells
EP2456862A4 (en) 2009-07-20 2013-02-27 Janssen Biotech Inc Differentiation of human embryonic stem cells
JP5819826B2 (en) 2009-07-20 2015-11-24 ヤンセン バイオテツク,インコーポレーテツド Differentiation of human embryonic stem cells
US20110020293A1 (en) * 2009-07-21 2011-01-27 Abt Holding Company Use of Stem Cells to Reduce Leukocyte Extravasation
ES2360434B1 (en) * 2009-07-21 2012-04-12 Universitat Internacional De Catalunya PLURIPOTENTIAL MOTHER CELLS OBTAINED FROM THE DENTAL PULP.
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
US10973656B2 (en) 2009-09-18 2021-04-13 Spinal Surgical Strategies, Inc. Bone graft delivery system and method for using same
US20170238984A1 (en) 2009-09-18 2017-08-24 Spinal Surgical Strategies, Llc Bone graft delivery device with positioning handle
US9629729B2 (en) 2009-09-18 2017-04-25 Spinal Surgical Strategies, Llc Biological delivery system with adaptable fusion cage interface
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
USD723682S1 (en) 2013-05-03 2015-03-03 Spinal Surgical Strategies, Llc Bone graft delivery tool
US9173694B2 (en) 2009-09-18 2015-11-03 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US9186193B2 (en) 2009-09-18 2015-11-17 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US8906028B2 (en) 2009-09-18 2014-12-09 Spinal Surgical Strategies, Llc Bone graft delivery device and method of using the same
USD750249S1 (en) 2014-10-20 2016-02-23 Spinal Surgical Strategies, Llc Expandable fusion cage
CA3049393C (en) * 2009-09-23 2020-12-29 Restem Llc Umbilical cord lining stem cells and methods and material for isolating and culturing same
JP2013508013A (en) 2009-10-16 2013-03-07 ザ ユニバーシティ オブ メディスン アンド デンティストリー オブ ニュー ジャージー Method of treating chronic neural tissue injury using cell therapy strategy
MX350966B (en) * 2009-10-29 2017-09-26 Janssen Biotech Inc Pluripotent stem cells.
EP2493314B1 (en) 2009-10-30 2020-04-08 Abela Pharmaceuticals, Inc. Dimethyl sulfoxide (dmso) and methylsulfonylmethane (msm) formulations to treat osteoarthritis
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
CN102711799A (en) * 2009-11-10 2012-10-03 纽约市哥伦比亚大学理事会 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
KR101923537B1 (en) * 2009-12-23 2018-11-29 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
KR101867369B1 (en) 2009-12-23 2018-06-14 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
KR20120115602A (en) * 2010-01-26 2012-10-18 안트로제네시스 코포레이션 Treatment of bone-related cancers using placental stem cells
EA201290577A1 (en) 2010-01-26 2013-06-28 Стем Селз Спин С.А. CELL HOMOGENATES FROM STEM CELLS OBTAINED FROM GROWING REIT HORNS, METHOD FOR ITS OBTAINING AND APPLICATION
AR080218A1 (en) 2010-02-18 2012-03-21 Osiris Therapeutics Inc IMMUNOCOMPATIBLE CORIONIC MEMBRANE PRODUCTS
AU2011218066B2 (en) * 2010-02-19 2015-04-23 Lifecell Corporation Abdominal wall treatment devices
AU2011217784B2 (en) 2010-02-22 2014-10-09 Biomimetic Therapeutics, Llc. Platelet-derived growth factor compositions and methods for the treatment of tendinopathies
JP6013196B2 (en) 2010-03-01 2016-10-25 ヤンセン バイオテツク,インコーポレーテツド Method for purifying cells derived from pluripotent stem cells
TWI756797B (en) 2010-04-07 2022-03-01 美商瑟魯勒瑞堤股份有限公司 Angiogenesis using placental stem cells
CN102933221A (en) 2010-04-08 2013-02-13 人类起源公司 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
DK2569419T3 (en) 2010-05-12 2019-05-13 Janssen Biotech Inc DIFFERENTIAL HUMAN EMBRYONIC STAMPS
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US20130209415A1 (en) * 2010-06-03 2013-08-15 The Board Of Trustees Of The Leland Stanford Junior University Purified compositions of cardiovascular progenitor cells
PT2588083T (en) 2010-07-02 2017-06-26 Univ North Carolina Chapel Hill Biomatrix scaffolds
EP3549617B1 (en) 2010-07-12 2023-09-13 University of Southern California Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
CN103097520B (en) 2010-07-13 2017-12-05 人类起源公司 The method for producing NK
US8825388B2 (en) 2010-07-13 2014-09-02 Qualcomm Incorporated Indoor likelihood heatmap
JP2012031127A (en) * 2010-08-03 2012-02-16 Nagoya Univ Composition including umbilical cord matrix stromal cell
US20130203146A1 (en) * 2010-08-03 2013-08-08 Jackie Y. Ying Microfabricated scaffold structures
IL207586A0 (en) 2010-08-12 2010-12-30 Omrix Biopharmaceuticals Ltd A fibrin based therapeutic preparation and use thereof
WO2012021885A1 (en) * 2010-08-13 2012-02-16 The Trustees Of Columbia University In The City Of New York Three-dimensional tissue engineering devices and uses thereof
SG187947A1 (en) 2010-08-31 2013-03-28 Janssen Biotech Inc Differentiation of pluripotent stem cells
CN103180435A (en) * 2010-08-31 2013-06-26 库克通用生物技术有限责任公司 Systemic, allogenic stem cell therapies for treatment of diseases in animals
SG187946A1 (en) 2010-08-31 2013-03-28 Janssen Biotech Inc Differentiation of human embryonic stem cells
SG10201506852VA (en) 2010-08-31 2015-10-29 Janssen Biotech Inc Differentiation of human embryonic stem cells
US9725689B2 (en) 2010-10-08 2017-08-08 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US8546338B2 (en) 2010-12-08 2013-10-01 Johnson & Johnson Consumer Companies, Inc. Self-assembling hydrogels based on dicephalic peptide amphiphiles
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
WO2012092485A1 (en) 2010-12-31 2012-07-05 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
JP5388233B2 (en) * 2011-01-19 2014-01-15 富士ソフト株式会社 Method for evaluating the cartilage characteristics of regenerated cartilage
US8945536B2 (en) * 2011-01-26 2015-02-03 The Chinese University Of Hong Kong Stem cell sheet for tissue repair
US9044430B2 (en) 2011-03-18 2015-06-02 Microvascular Tissues, Inc. Allogeneic microvascular tissue for soft tissue treatments
KR101835917B1 (en) 2011-03-22 2018-03-07 플루리스템 리미티드 Methods for treating radiation or chemical injury
US9758395B2 (en) 2011-04-28 2017-09-12 Aquero Company, Llc Lysine-based polymer coagulants for use in clarification of process waters
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
US10478206B2 (en) 2011-04-29 2019-11-19 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
EP2714111B1 (en) * 2011-05-31 2021-03-17 LifeCell Corporation Adipose tissue matrices
MX357749B (en) 2011-06-01 2018-07-23 Anthrogenesis Corp Treatment of pain using placental stem cells.
EP2717888B1 (en) 2011-06-10 2020-09-09 Tissuetech, Inc. Methods of processing fetal support tissues
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
US9133438B2 (en) 2011-06-29 2015-09-15 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
US20130005829A1 (en) 2011-06-30 2013-01-03 Advanced Technologies And Regenerative Medicine, Llc. Segmented, epsilon-Caprolactone-Rich, Poly(epsilon-Caprolactone-co-p-Dioxanone) Copolymers for Medical Applications and Devices Therefrom
US10865383B2 (en) 2011-07-12 2020-12-15 Lineage Cell Therapeutics, Inc. Methods and formulations for orthopedic cell therapy
AU2011374879C1 (en) * 2011-08-10 2018-06-14 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
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
CA2859657C (en) 2011-12-20 2021-03-02 Lifecell Corporation Acellular tissue matrix particles and flowable products comprising them
CA2860850C (en) 2011-12-20 2019-05-21 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
KR102090751B1 (en) 2011-12-22 2020-03-19 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US9611513B2 (en) * 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
SI3321355T1 (en) 2011-12-30 2021-11-30 Amit Patel Methods and compositions for the clinical derivation of an allogenic cell and therapeutic uses
ES2705823T3 (en) 2012-01-24 2019-03-26 Lifecell Corp Matrices of elongated tissues
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
SG11201405052RA (en) 2012-03-07 2014-10-30 Janssen Biotech Inc Defined media for expansion and maintenance of pluripotent stem cells
WO2013158664A2 (en) 2012-04-17 2013-10-24 Kythera Biopharmaceuticals, Inc. Use of engineered viruses to specifically kill senescent cells
ES2749178T3 (en) 2012-04-24 2020-03-19 Lifecell Corp Functionalized tissue matrices
US9884076B2 (en) 2012-06-05 2018-02-06 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
CA2875786C (en) 2012-06-08 2022-12-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
JP6389170B2 (en) * 2012-07-06 2018-09-12 スリーディー マトリックス, エルティーディー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
EP2872191B1 (en) 2012-07-13 2019-08-07 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
JP6433896B2 (en) 2012-08-13 2018-12-05 シーダーズ−サイナイ・メディカル・センターCedars−Sinai Medical Center Exosomes and microribonucleic 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
US11819522B2 (en) 2012-09-19 2023-11-21 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
DK2898064T3 (en) 2012-09-19 2019-03-04 Microvascular Tissues Inc COMPOSITIONS FOR TREATMENT AND PREVENTION OF TISSUE DAMAGE AND DISEASE
US10596202B2 (en) 2012-09-19 2020-03-24 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US9872937B2 (en) 2012-09-19 2018-01-23 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
EP3332816B1 (en) 2012-09-26 2020-11-04 LifeCell Corporation Processed adipose tissue
WO2014051398A1 (en) * 2012-09-28 2014-04-03 한국생명공학연구원 Pharmaceutical composition comprising acecainide or derivative thereof for preventing or treating diseases associated with muscular weakness
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
US10279018B2 (en) 2012-12-03 2019-05-07 Unity Biotechnology, 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
BR112015015770A2 (en) 2012-12-31 2017-07-11 Janssen Biotech Inc Human embryonic stem cell culture at air-liquid interface for differentiation in endocrine pancreatic cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
WO2014106141A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Suspension and clustering of human pluripotent cells for differentiation into pancreatic endocrine cells
WO2014105546A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
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
AU2014215458A1 (en) 2013-02-05 2015-08-13 Anthrogenesis Corporation Natural killer cells from placenta
WO2014164815A2 (en) 2013-03-12 2014-10-09 Allergan, Inc. Adipose tissue combinations, devices, and uses thereof
KR102277830B1 (en) 2013-03-13 2021-07-14 알로소스 Fascia fibrous compositions and methods for their use and manufacture
JP2016512433A (en) * 2013-03-14 2016-04-28 アントフロゲネシス コーポレーション Enhanced placental stem cells and uses thereof
WO2014150784A1 (en) 2013-03-15 2014-09-25 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
US11229789B2 (en) 2013-05-30 2022-01-25 Neurostim Oab, Inc. Neuro activator with controller
CN105307719B (en) 2013-05-30 2018-05-29 格雷厄姆·H.·克雷西 Local nerve stimulation instrument
WO2015023901A1 (en) 2013-08-15 2015-02-19 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
WO2015085232A1 (en) 2013-12-06 2015-06-11 Allosource Method of drying sheets of tissue
WO2015095380A1 (en) * 2013-12-17 2015-06-25 Kassab Ghassan S Devices, systems and methods for tissue engineering of luminal grafts
US10328058B2 (en) 2014-01-28 2019-06-25 Mayo Foundation For Medical Education And Research Treating atherosclerosis by removing senescent foam cell macrophages from atherosclerotic plaques
CA3100140C (en) * 2014-01-28 2023-10-24 Buck Institute For Research On Aging Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders
WO2015116735A1 (en) 2014-01-28 2015-08-06 Mayo Foundation For Medical Education And Research Methods and combinations for killing senescent cells and for treating senescence-associated diseases and disorders
ES2871138T3 (en) 2014-02-12 2021-10-28 Gyroscope Therapeutics Ltd Apparatus for Suprachoroidal Administration of Therapeutic Agent
EP3628337A1 (en) 2014-03-10 2020-04-01 3-D Matrix, Ltd. Sterilization and filtration of peptide compositions
RU2016139369A (en) 2014-03-10 2018-04-10 3-Д Матрикс, Лтд. COMPOSITIONS OF SELF-ORGANIZING PEPTIDES
EP3122866B1 (en) 2014-03-25 2019-11-20 Terumo BCT, Inc. Passive replacement of media
EP3139935A4 (en) * 2014-05-07 2017-11-15 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
US10006006B2 (en) 2014-05-16 2018-06-26 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
US9925088B2 (en) 2014-06-06 2018-03-27 Janssen Biotech, Inc. Sub-retinal tangential needle catheter guide and introducer
US9949874B2 (en) 2014-06-06 2018-04-24 Janssen Biotech, Inc. Therapeutic agent delivery device with convergent lumen
US20160000550A1 (en) * 2014-07-05 2016-01-07 Deborah Nagle Methods for treating diseases of the colon
EP3168297B1 (en) * 2014-07-11 2020-09-16 Metcela Inc. Cardiac cell culture material
US10064752B2 (en) 2014-09-11 2018-09-04 Orbit Biomedical Limited Motorized suprachoroidal injection of therapeutic agent
US10322028B2 (en) 2014-09-11 2019-06-18 Orbit Biomedical Limited Method and apparatus for sensing position between layers of an eye
US10219936B2 (en) 2014-09-11 2019-03-05 Orbit Biomedical Limited Therapeutic agent delivery device with advanceable cannula and needle
US10258502B2 (en) 2014-09-18 2019-04-16 Orbit Biomedical Limited Therapeutic agent delivery device
US9504905B2 (en) 2014-09-19 2016-11-29 Bally Gaming, Inc. Card shuffling device and calibration method
KR101613478B1 (en) * 2014-09-22 2016-04-19 (주)안트로젠 Composition comprising mesenchymal stem cell-hydrogel and preparation method thereof
EP3198006B1 (en) 2014-09-26 2021-03-24 Terumo BCT, Inc. Scheduled feed
WO2016054591A1 (en) 2014-10-03 2016-04-07 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
MX2017004520A (en) 2014-10-14 2018-03-15 Lynch Samuel Compositions for treating wounds.
US20180280447A1 (en) * 2014-10-29 2018-10-04 Sungkwang Medical Foundation Placenta-derived cells excreting c3 or c1r complement and composition containing same
CA2966431C (en) * 2014-10-31 2023-02-14 The Administrators Of The Tulane Educational Fund Surgical grafts for replacing the nipple and areola or damaged epidermis
ES2831298T3 (en) 2014-11-07 2021-06-08 Exocobio Inc Composition that includes exosomes derived from stem cells to whiten the skin or improve wrinkles
BR112017011513A2 (en) * 2014-12-05 2018-07-10 Janssen Biotech Inc treatment of eye conditions using progenitor cells
WO2016086403A1 (en) * 2014-12-05 2016-06-09 Meridigen Biotech Co., Ltd. Method of distinguishing mesenchymal stem cells
CN107249608A (en) * 2014-12-16 2017-10-13 詹森生物科技公司 Retinosis is treated using progenitor cells
US20170080033A1 (en) * 2014-12-16 2017-03-23 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
AU2015363008B2 (en) 2014-12-19 2021-12-09 Janssen Biotech, Inc. Suspension culturing of pluripotent stem cells
WO2016111899A1 (en) 2015-01-05 2016-07-14 Petrucci Gary M 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
TW201642914A (en) 2015-02-23 2016-12-16 組織科技股份有限公司 Apparatuses and methods for treating ophthalmic diseases and disorders
US10433928B2 (en) 2015-03-10 2019-10-08 Allergan Pharmaceuticals Holdings (Ireland) Unlimited Company Multiple needle injector
WO2016141460A1 (en) 2015-03-11 2016-09-15 The University Of British Columbia Pancreatic endocrine progenitor cell therapies for the treatment of obesity and type 2 diabetes (t2d)
AU2016243612A1 (en) * 2015-03-31 2017-10-12 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
WO2016187555A1 (en) 2015-05-20 2016-11-24 Tissuetech, Inc. Compositions and methods for preventing the proliferation and epithelial-mesenchymal transition of epithelial cells
CA2986702C (en) 2015-05-21 2023-04-04 David Wang 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
EP3342858B1 (en) 2015-08-28 2023-10-04 Rohto Pharmaceutical Co., Ltd. Ror1-positive mesenchymal stem cells and method for preparing same, pharmaceutical composition containing ror1-positive mesenchymal stem cells and method for preparing same, and method for preventing or treating diseases by using ror1-positive mesenchymal stem cells
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
AU2016365312A1 (en) * 2015-12-04 2018-05-24 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
KR20170076484A (en) * 2015-12-24 2017-07-04 삼성전자주식회사 Method of isolating senescent cell with high expression of Protocadherin gene
US10814038B2 (en) 2016-01-06 2020-10-27 3-D Matrix, Ltd. Combination compositions
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US10791730B2 (en) 2016-01-14 2020-10-06 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
AU2017240591B2 (en) 2016-03-30 2023-08-24 Asterias Biotherapeutics, Inc. Oligodendrocyte progenitor cell compositions
KR102288170B1 (en) 2016-04-08 2021-08-09 알레간 인코포레이티드 Aspiration and injection device
MA45479A (en) 2016-04-14 2019-02-20 Janssen Biotech Inc DIFFERENTIATION OF PLURIPOTENT STEM CELLS IN ENDODERMAL CELLS OF MIDDLE INTESTINE
JP7108537B2 (en) 2016-04-27 2022-07-28 ロート製薬株式会社 Mesenchymal stem cells expressing at least one cell surface marker selected from the group consisting of CD201, CD46, CD56, CD147 and CD165, a method for preparing the same, and a pharmaceutical composition containing the mesenchymal stem cells and a method for preparing the same
WO2017210652A1 (en) 2016-06-03 2017-12-07 Cedars-Sinai Medical Center Cdc-derived exosomes for treatment of ventricular tachyarrythmias
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US10646374B2 (en) 2016-06-17 2020-05-12 Orbit Biomedical Limited Apparatus and method to form entry bleb for subretinal delivery of therapeutic agent
US10806629B2 (en) 2016-06-17 2020-10-20 Gyroscope Therapeutics Limited Injection device for subretinal delivery of therapeutic agent
US11000410B2 (en) 2016-06-17 2021-05-11 Gyroscope Therapeutics Limited Guide apparatus for tangential entry into suprachoroidal space
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
EP3481446B1 (en) 2016-07-05 2020-09-30 LifeCell Corporation Tissue matrices incorporating multiple tissue types
MA45623A (en) * 2016-07-05 2019-05-15 Janssen Biotech Inc TREATMENT OF RETINAL VASCULAR DISEASE USING PROGENITOR CELLS
KR20190055088A (en) * 2016-08-19 2019-05-22 리젠티스 코퍼레이션 Extracellular matrix for tissue remodeling 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
NZ753034A (en) * 2016-11-02 2023-04-28 Axogen Corp Amnion tissue grafts and methods of preparing and using same
RU2644306C1 (en) * 2016-11-22 2018-02-08 Общество с ограниченной ответственностью "ДЖИ-Групп" Method of recovery of defects of covering tissues
WO2018125851A1 (en) * 2016-12-26 2018-07-05 Michael Moeller Systems and methods to isolate and expand stem cells from urine
US11273072B2 (en) 2017-01-13 2022-03-15 Gyroscope Therapeutics Limited Suprachoroidal injection device
US10772986B2 (en) 2017-01-26 2020-09-15 Allosource Fascia fibrous compositions and methods for their use and manufacture
EP3593809A4 (en) 2017-03-08 2021-01-06 Rohto Pharmaceutical Co., Ltd. Ror1-positive mesenchymal stem cell-containing pharmaceutical composition for preventing or treating disease associated with fibrosis, method for preparing same, and method for preventing or treating disease associated with fibrosis using ror1-positive mesenchymal stem cells
US11076984B2 (en) 2017-03-13 2021-08-03 Gyroscope Therapeutics Limited Method of performing subretinal drainage and agent delivery
US11278597B2 (en) 2017-03-17 2022-03-22 Rutgers, The State University Of New Jersey Compositions and methods for wound healing
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor
JP2020516654A (en) 2017-04-13 2020-06-11 センティ バイオサイエンシズ インコーポレイテッド Combination cancer immunotherapy
AU2018255346A1 (en) 2017-04-19 2019-11-07 Capricor, Inc. 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
WO2019018545A1 (en) 2017-07-18 2019-01-24 The Research Foundation For The State University Of New York Biomarkers for intracranial aneurysm
US10251917B1 (en) 2017-09-19 2019-04-09 Gary M. Petrucci Methods and materials for treating tumors
CN111201046B (en) 2017-10-18 2022-06-28 生命细胞公司 Adipose tissue products and methods of production
US11123375B2 (en) 2017-10-18 2021-09-21 Lifecell Corporation Methods of treating tissue voids following removal of implantable infusion ports using adipose tissue products
US11246994B2 (en) 2017-10-19 2022-02-15 Lifecell Corporation Methods for introduction of flowable acellular tissue matrix products into a hand
US11826488B2 (en) 2017-10-19 2023-11-28 Lifecell Corporation Flowable acellular tissue matrix products and methods of production
WO2019079681A1 (en) 2017-10-20 2019-04-25 President And Fellows Of Harvard College Methods for producing mature adipocytes and methods of use thereof
US20210038650A1 (en) * 2017-10-23 2021-02-11 Cell Medicine, Inc. Mesenchymal stem cell therapy of leigh syndrome
TW201932126A (en) * 2017-11-03 2019-08-16 美商健生生物科技公司 Method of inhibiting angiogenesis
US10953225B2 (en) 2017-11-07 2021-03-23 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
WO2019139137A1 (en) 2018-01-12 2019-07-18 国立大学法人大阪大学 Stratified squamous epithelial cell normal differentiation and maturation promoting agent, epithelial disease therapeutic agent, and stratified squamous epithelial cell normal differentiation and maturation promoting method
US20190218521A1 (en) * 2018-01-18 2019-07-18 Lorenzo Bracco Culture medium of viruses for human vaccines have to consist of human cells from placenta and/or from umbilical cord of a fetus of the blood type 0 Rh- and of the mother of the blood type 0 Rh- (both, fetus and mother, must be of the blood type 0 Rh-)
WO2019152483A1 (en) * 2018-01-30 2019-08-08 University Of Georgia Research Foundation, Inc. Methods for vascular construction and products therefrom
CN108384752A (en) * 2018-02-13 2018-08-10 中国人民解放军第四五五医院 Application at Cartilage culture base and its in micro- splitting technique in knee cartilage differentiation
AU2019276713A1 (en) * 2018-05-30 2021-01-21 Direct Biologics Llc A growth factor and extracellular vesicle frozen or powdered additive comprising a mesenchymal stem cell (MSC) preparation and methods of use
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
KR20210094534A (en) 2018-10-17 2021-07-29 센티 바이오사이언시스, 인코포레이티드 Combination Cancer Immunotherapy
KR20210121112A (en) 2019-01-23 2021-10-07 아스테리아스 바이오세라퓨틱스, 인크. 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
JP2022534110A (en) 2019-05-30 2022-07-27 ライフセル コーポレーション biological breast implants
KR20220025834A (en) 2019-06-26 2022-03-03 뉴로스팀 테크놀로지스 엘엘씨 Non-invasive neural activators with adaptive circuits
WO2021039943A1 (en) * 2019-08-29 2021-03-04 味の素株式会社 Method for producing mesenchymal stem cells from living body-derived cell sample containing mesenchymal stem cells
WO2021050907A1 (en) * 2019-09-13 2021-03-18 The University Of North Carolina At Chapel Hill Method of making human mouse xenografts
US20240052313A1 (en) * 2019-10-07 2024-02-15 University Of Utah Research Foundation Chondrogenic human mesenchymal stem cell (msc) sheets
EP4041147A1 (en) 2019-10-11 2022-08-17 Gyroscope Therapeutics Limited Dose clip assembly for syringe
WO2021126921A1 (en) 2019-12-16 2021-06-24 Neurostim Solutions, Llc Non-invasive nerve activator with boosted 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
WO2022136913A1 (en) 2020-12-22 2022-06-30 Gyroscope Therapeutics Limited Ocular cannula guide
WO2022154474A1 (en) * 2021-01-12 2022-07-21 (주)셀라토즈테라퓨틱스 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
US5266480A (en) * 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
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
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US4925667A (en) * 1986-05-27 1990-05-15 Qmax Technology Group, Inc. Substrate with particulate cosmetic
JPS642643A (en) 1987-06-26 1989-01-06 Bio Material Yunibaasu:Kk Artificial skin
NZ226750A (en) 1987-10-29 1990-09-26 Amrad Corp Ltd Immortalisation of neural precursor cells by introducing a retrovirus vector containing a myc-oncogene
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5162405A (en) * 1987-12-24 1992-11-10 Elf Atochem North America, Inc. Single-functional and mixtures of multi-functional oligomeric performance additive compositions and their uses
GB8803697D0 (en) 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
US4963439A (en) 1988-04-19 1990-10-16 Ube Industries, Ltd. Continuous fiber-reinforced Al-Co alloy matrix composite
US20030032178A1 (en) 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
US5618670A (en) * 1988-08-26 1997-04-08 The United States Of America As Represented By The Department Of Health & Human Services Detection method for c-raf-1 genes
US4925677A (en) 1988-08-31 1990-05-15 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
JPH06104061B2 (en) 1989-02-10 1994-12-21 花王株式会社 Cell culture support material
US5284766A (en) 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
EP0385733B1 (en) * 1989-02-27 1994-06-01 Takasago International Corporation Process for preparing optically active 6-t-butoxy-3,5-dihydroxyhexanoic esters
FR2646438B1 (en) 1989-03-20 2007-11-02 Pasteur Institut A METHOD FOR SPECIFIC REPLACEMENT OF A COPY OF A GENE PRESENT IN THE RECEIVER GENOME BY INTEGRATION OF A GENE DIFFERENT FROM THAT OR INTEGRATION
US5437994A (en) 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5574205A (en) 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
US5840580A (en) 1990-05-01 1998-11-24 Becton Dickinson And Company Phenotypic characterization of the hematopoietic stem cell
US5145770A (en) * 1990-06-04 1992-09-08 Biosurface Technology, Inc. Cryopreservation of cultured epithelial sheets
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.
SK120193A3 (en) 1991-05-02 1994-07-06 Yeda Res & Dev Pharmaceutical composition for the prevention and/or treatment of pathological processes
US6399369B1 (en) 1991-07-08 2002-06-04 Neurospheres Holdings Ltd. Multipotent neural stem cell cDNA libraries
WO1993003139A1 (en) 1991-08-08 1993-02-18 Kao Corporation Cell culture support, production thereof, and production of cell cluster using same
EP0529751A1 (en) 1991-08-09 1993-03-03 W.R. Grace & Co.-Conn. Cell culture substrate, test material for cell culture and preparations thereof
WO1993004169A1 (en) 1991-08-20 1993-03-04 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
AU2694592A (en) * 1991-09-30 1993-05-03 Walser, Mackenzie 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)
AU673865B2 (en) 1992-10-29 1996-11-28 Australian National University, The Angiogenesis inhibitory antibodies
US5955343A (en) 1992-12-28 1999-09-21 Massachusetts Institute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5670483A (en) 1992-12-28 1997-09-23 Massachusetts Insititute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5707643A (en) 1993-02-26 1998-01-13 Santen Pharmaceutical Co., Ltd. Biodegradable scleral plug
US5494899A (en) * 1993-04-07 1996-02-27 Oklahoma Medical Research Foundation Selective regulation of B lymphocyte precursors by hormones
WO1994025584A1 (en) 1993-04-28 1994-11-10 Johns Hopkins University School Of Medicine Chronic endothelial cell culture under flow
IL110589A0 (en) 1993-08-10 1994-11-11 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF- beta family
JP3680114B2 (en) 1993-09-17 2005-08-10 敏一 中村 Cranial nerve disorder treatment
US6686198B1 (en) 1993-10-14 2004-02-03 President And Fellows Of Harvard College Method of inducing and maintaining neuronal cells
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US5456835A (en) * 1993-11-08 1995-10-10 Hemasure, Inc. Device and process for removing free hemoglobin from blood
DE4406073A1 (en) 1994-02-24 1995-08-31 Univ Ludwigs Albert Process for the production of human, clonogenic fibroblasts, process for gene transfection of fibroblasts and fibroblasts obtained in this way
US5698518A (en) 1994-03-30 1997-12-16 Oklahoma Medical Research Foundation Method for regulating inflammation and tumor growth with calmodulin, calmodulin analogues or calmodulin antagonists
US5466233A (en) 1994-04-25 1995-11-14 Escalon Ophthalmics, Inc. Tack for intraocular drug delivery and method for inserting and removing same
US6703017B1 (en) 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US5834308A (en) 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
JPH10505250A (en) 1994-06-06 1998-05-26 ケース ウエスターン リザーブ ユニバーシティ Biomatrix for tissue regeneration
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
ATE259419T1 (en) 1996-03-15 2004-02-15 Munin Corp HUMAN CYR61, AN EXTRACELLULAR MATRIX SIGNALING MOLECULE
US6223188B1 (en) * 1996-04-10 2001-04-24 Sun Microsystems, Inc. Presentation of link information as an aid to hypermedia navigation
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
US6541028B1 (en) 1997-01-17 2003-04-01 Celadon Science, Llc Methods for promoting healing of corneal resurfacing wounds
AU6144698A (en) 1997-02-05 1998-08-25 Case Western Reserve University Stimulatory effects of bfgf and bmp-2 on osteogenic differentiation of mesenchymal stem cells
WO1998051317A1 (en) 1997-05-13 1998-11-19 Osiris Therapeutics, Inc. Osteoarthritis cartilage regeneration using human mesenchymal stem cells
EP0983062A4 (en) 1997-05-21 2002-10-23 Sloan Kettering Inst Cancer Method for increasing the concentration of ascorbic acid in brain tissues of a subject
JP4132089B2 (en) * 1997-05-30 2008-08-13 オステオバイオロジックス,インコーポレイテッド Fiber reinforced porous biodegradable implantation device
ES2251773T5 (en) * 1997-07-14 2009-05-12 Osiris Therapeutics, Inc. REGENERATION OF THE CARDIAC MUSCLE USING MESENQUIMATOSE PRECURSOR CELLS.
US5902598A (en) 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
CA2312847C (en) 1997-12-02 2014-09-30 Zen Bio, 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
JP4441115B2 (en) 1998-03-13 2010-03-31 オシリス セラピューティクス,インコーポレイテッド Methods and uses of human non-self mesenchymal stem cells
AU755273B2 (en) 1998-03-16 2002-12-05 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
JP2002513545A (en) 1998-05-07 2002-05-14 ザ ユニヴァーシティー オブ サウス フロリダ Bone marrow cells as a source of neurons for brain and spinal cord repair
DE69936720T2 (en) 1998-05-29 2008-04-30 Osiris Therapeutics, Inc. HUMAN CD45 + AND / OR FIBROBLASTES + MESENCHYMAL STEM CELLS
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
ES2335066T3 (en) 1998-08-10 2010-03-18 The Government Of The Usa, As Represented By The Secretary, Department Of Health And Human Services DIFFERENTIATION OF NON-INSULIN CELL PRODUCTS IN GLP-1 OR EXENDIN-4 INSULIN CELLS AND USING THEMSELVES.
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
AU781750B2 (en) 1999-02-04 2005-06-09 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
KR100968165B1 (en) 1999-03-10 2010-07-06 더 리전츠 오브 더 유니버시티 오브 캘리포니아 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
CA2370330C (en) 1999-04-16 2009-01-20 Wm. Marsh Rice University Biodegradable poly(propylene fumarate) networks cross linked with poly(propylene fumarate)-diacrylate macromers
US6261600B1 (en) * 1999-04-30 2001-07-17 Drugtech Corporation Folic acid supplement
US7371400B2 (en) 2001-01-02 2008-05-13 The General Hospital Corporation Multilayer device for tissue engineering
US6287340B1 (en) 1999-05-14 2001-09-11 Trustees Of Tufts College Bioengineered anterior cruciate ligament
US6372494B1 (en) 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
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
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
US6333029B1 (en) 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
EP2348104A1 (en) 1999-08-05 2011-07-27 Mcl Llc Multipotent adult stem cells and methods for isolation
WO2002064748A2 (en) 2001-02-14 2002-08-22 Furcht Leo T Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
US6555374B1 (en) 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
JP2003509374A (en) 1999-09-14 2003-03-11 チルドレンズ メディカル センター コーポレーション Method for treating muscular dystrophy using bone marrow cells
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
US20030129745A1 (en) 1999-10-28 2003-07-10 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
EP1099754A1 (en) 1999-11-10 2001-05-16 Universiteit Leiden Mesenchymal stem cells and/or progenitor cells, their isolation and use
US20020164307A1 (en) 1999-12-06 2002-11-07 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
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
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
US7785882B2 (en) 2000-01-18 2010-08-31 Cornell Research Foundation, Inc. Neuronal progenitor cells from hippocampal tissue and a method for isolating and purifying them
US7544509B2 (en) 2000-01-24 2009-06-09 Mcgill University Method for preparing stem cell preparations
US6610535B1 (en) 2000-02-10 2003-08-26 Es Cell International Pte Ltd. Progenitor cells and methods and uses related thereto
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
CA2399434C (en) 2000-02-11 2010-10-19 The Schepens Eye Research Institute, Inc. Isolation and transplantation of retinal stem cells
IL151650A0 (en) 2000-03-09 2003-04-10 Saneron Ccel Therapeutics Inc Human cord blood as a source of neural tissue for repair of the brain and spinal cord
WO2001065934A2 (en) * 2000-03-09 2001-09-13 Lee Walters Applications of immune system tolerance to treatment of various diseases
US7405078B2 (en) 2000-03-16 2008-07-29 Cellseed Inc. Bed material for cell culture, method for co-culture of cell and co-cultured cell sheet obtainable therefrom
US6436704B1 (en) 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
US6673606B1 (en) 2000-04-12 2004-01-06 The Children's Hospital Of Philadelphia Therapeutic uses for mesenchymal stromal cells
US6375972B1 (en) 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
WO2001088103A2 (en) 2000-05-12 2001-11-22 University Of Utah Research Foundation Compositions and methods for tissue dedifferentiation and 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
EP2264147A1 (en) 2000-07-21 2010-12-22 Cellseed Inc. Epidermal cultured cell sheet, multi-layered cultured skin sheet and processes for producing those sheets
EP2275532B1 (en) 2000-07-21 2012-06-06 Cellseed Inc. Myocardium-like cell sheets and processes for producing them
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
DE10038814A1 (en) * 2000-08-09 2002-02-21 Abb Research Ltd High voltage direct current transformer
AU2001293586A1 (en) 2000-09-29 2002-04-08 Vincent Tropepe Primitive neural stem cells and method for differentiation of stem cells to neural cells
US6639470B1 (en) 2000-10-06 2003-10-28 Skyworks Solutions, Inc. Constant current biasing circuit for linear power amplifiers
US7560280B2 (en) 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
US20020098584A1 (en) 2000-11-06 2002-07-25 Palmer Theo D. Postmortem stem cells
KR20030088022A (en) 2000-11-30 2003-11-15 스템론 인크. 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
US6599323B2 (en) 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
WO2002059278A2 (en) 2001-01-24 2002-08-01 The Government Of The United States Of America, As Represented By The Secretary Of Department Of Health & 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
US7449180B2 (en) 2001-02-06 2008-11-11 John Kisiday Macroscopic scaffold containing amphiphilic peptides encapsulating cells
WO2002062961A2 (en) 2001-02-06 2002-08-15 Massachusetts Institute Of Technology Peptide scaffold encapsulation of tissue cells and uses thereof
EP2314673B1 (en) 2001-02-14 2013-07-24 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
EP2316463B1 (en) 2001-02-14 2017-04-12 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making them
CA2441986A1 (en) 2001-03-27 2002-10-03 Massachusetts Institute Of Technology Methods and products related to fgf dimerization
US7838292B1 (en) 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
CA2442177A1 (en) 2001-03-29 2002-10-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
US20050054102A1 (en) 2001-04-19 2005-03-10 Anna Wobus Method for differentiating stem cells into insulin-producing cells
KR100762261B1 (en) 2001-04-27 2007-10-04 (주)바이오니아 Process for preparation of full-length cDNA and anchor and primer used for the same
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
US20030022369A1 (en) 2001-05-18 2003-01-30 Helen Fillmore Differentiation of specialized dermal and epidermal cells into neuronal cells
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
EP1423504B1 (en) * 2001-08-08 2009-09-23 Levesque Biosciences, Inc. Compositions and methods for isolation, propagation, and differentiation of non-embryonic 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
EP1298201A1 (en) 2001-09-27 2003-04-02 Cardion AG Process for the production of cells exhibiting an islet-beta-cell-like state
US7072332B2 (en) * 2001-09-27 2006-07-04 Samsung Electronics Co., Ltd. Soft switch using distributed firewalls for load sharing voice-over-IP traffic in an IP network
CA2461859A1 (en) 2001-09-28 2003-04-10 Es Cell International Pte Ltd Methods of derivation and propagation of undifferentiated human embryonic stem (hes) cells on feeder-free matrices and human feeder layers
EP1444345A4 (en) 2001-10-18 2004-12-08 Ixion Biotechnology Inc Conversion of liver stem and progenitor cells to pancreatic functional cells
US7129034B2 (en) * 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
EP1453954A4 (en) 2001-11-09 2004-12-15 Artecel Sciences Inc Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
WO2003054146A2 (en) 2001-11-14 2003-07-03 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
WO2003045439A1 (en) 2001-11-28 2003-06-05 Anges Mg, Inc. Genetic remedies for neurodegenerative diseases
US6712850B2 (en) 2001-11-30 2004-03-30 Ethicon, Inc. Porous tissue scaffolds for the repair and regeneration of dermal tissue
CA2469209C (en) 2001-12-04 2012-04-24 Jianjian Jin Cultured cells from pancreatic islets
US20030109036A1 (en) 2001-12-06 2003-06-12 The Regents Of The University Of California Method for differentiating islet precursor cells into beta cells
AU2002354101A1 (en) * 2001-12-06 2003-06-17 Sankyo Company, Limited Medicinal compositions containing human amnion-origin cells
US7033831B2 (en) * 2001-12-07 2006-04-25 Geron Corporation Islet cells from human embryonic stem cells
WO2003050266A2 (en) * 2001-12-11 2003-06-19 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
AU2002350352A1 (en) * 2001-12-21 2003-07-15 Mount Sinai Hospital Cellular compositions and methods of making and using them
US7101546B2 (en) 2001-12-21 2006-09-05 Amcyte, Inc. In situ maturation of cultured pancreatic stem cells having a specified, intermediate stage of development
EP1461421A2 (en) 2001-12-28 2004-09-29 Cellartis AB 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
AU2003217357A1 (en) 2002-02-07 2003-09-02 The Research Foundation Of The State University Of New York Generation of new insulin cells from progenitor cells present in adult pancreatic islets
WO2003068937A2 (en) * 2002-02-13 2003-08-21 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
WO2003070189A2 (en) 2002-02-15 2003-08-28 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
AU2003215280A1 (en) 2002-02-15 2003-09-09 Northwestern University Self-assembly of peptide-amphiphile nanofibers under physiological conditions
KR100489248B1 (en) 2002-02-19 2005-05-11 메디포스트(주) Isolation and culture-expansion methods of mesenchymal stem/progenitor cells from umbilical cord blood, and differentiation method of umbilical cord blood-derived mesenchymal stem/progenitor cells into various mesenchymal tissues
WO2003072728A2 (en) 2002-02-22 2003-09-04 University Of Florida Cellular trans-differentiation
US20030161818A1 (en) * 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
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
US20050118715A1 (en) 2002-04-12 2005-06-02 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
JP4136434B2 (en) 2002-04-17 2008-08-20 進 清野 Induction of insulin-producing cells
AU2003239159A1 (en) 2002-04-19 2003-11-03 University Of Pittsburgh Of The Commonwealth System Of Higher Education Placental derived stem cells and uses thereof
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
CA2484223A1 (en) 2002-04-25 2003-11-06 Wisconsin Alumni Research Foundation Use of human neural stem cells secreting gdnf for treatment of parkinson's and other neurodegenerative diseases
US20040029269A1 (en) 2002-05-07 2004-02-12 Goldman Steven A Promoter-based isolation, purification, expansion, and transplantation of neuronal progenitor cells, oligodendrocyte progenitor cells, or neural stem cells from a population of embryonic stem cells
WO2003094965A2 (en) * 2002-05-08 2003-11-20 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
KR20050008757A (en) 2002-05-30 2005-01-21 셀진 코포레이션 Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
GB2405642B (en) 2002-06-07 2007-03-28 Es Cell Int Pte Ltd Methods of regulating differentiation in stem cells
US7410797B2 (en) 2002-06-11 2008-08-12 Ogle Roy C Meningeal-derived stem cells
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
EP1539930A4 (en) 2002-07-29 2006-08-09 Es Cell Int Pte Ltd Multi-step method for the differentiation of insulin positive, glucose
US7527968B2 (en) 2002-07-29 2009-05-05 Robarts Research Institute Regeneration initiating cells
WO2004016747A2 (en) 2002-08-14 2004-02-26 University Of Florida Bone marrow cell differentiation
AU2003265094A1 (en) * 2002-08-26 2004-03-11 Neuronova Ab Method for culturing stem cells
EP1543111A4 (en) * 2002-08-28 2006-09-13 Univ Florida Neurogenesis from hepatic stem cells
EP1539928A4 (en) 2002-09-06 2006-09-06 Amcyte 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
EP1583422B1 (en) 2002-12-05 2016-03-30 Case Western Reserve University Cell-based therapies for ischemia
US20090169597A1 (en) 2007-12-27 2009-07-02 Ethicon, Incorporated Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
JP4571387B2 (en) 2003-02-07 2010-10-27 宣男 櫻川 Human amnion-derived side population cells and uses thereof
BRPI0407221A (en) * 2003-02-11 2006-01-31 John E Davies Wharton Jelly Extract, Method for Obtaining a Human Parent Cell, Methods for Producing a Cell Population, Cell Populations, and Method for Producing Bone Tissue
EP1606065A4 (en) 2003-02-11 2010-09-29 Univ Northwestern Methods and materials for nanocrystalline surface coatings and attachment of peptide amphiphile nanofibers thereon
US7521481B2 (en) 2003-02-27 2009-04-21 Mclaurin Joanne Methods of preventing, treating and diagnosing disorders of protein aggregation
WO2006019366A1 (en) 2003-03-28 2006-02-23 Wisconsin Alumni Research Foundation Physiochemical culture conditions for embryonic stem cells
WO2005034624A2 (en) 2003-05-21 2005-04-21 The General Hospital Corporation Microfabricated compositions and processes for engineering tissues containing multiple cell types
US8703121B2 (en) * 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular 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
EP2228035A1 (en) * 2003-12-23 2010-09-15 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
US7598083B2 (en) 2004-10-29 2009-10-06 Centocor, Inc. Chemically defined media compositions
US7655678B2 (en) * 2004-11-30 2010-02-02 Council of Scientfic & Industrial Research Pharmaceutical composition for the management of tumors
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060171930A1 (en) 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
WO2006083394A2 (en) 2004-12-21 2006-08-10 Ethicon, Inc. Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
JP5340599B2 (en) * 2004-12-23 2013-11-13 エシコン・インコーポレイテッド Umbilical tissue-derived postpartum cells and methods for producing and using the same
WO2006071773A2 (en) 2004-12-23 2006-07-06 Ethicon Incoporated 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
CA2589063C (en) * 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
US7741311B2 (en) 2005-01-03 2010-06-22 Shaker Mousa Composition and method for treating occlusive vascular diseases, nerve regeneration, and wound healing
JP2008538276A (en) 2005-01-28 2008-10-23 ノヴァセラ・リミテッド Methods for embryonic stem cell culture
CN103356705B (en) 2005-03-31 2015-09-30 斯丹姆涅恩有限公司 Prepare the method for the medicine in order to treat burn wound
US7923007B2 (en) 2005-08-08 2011-04-12 Academia Sinica Brain tissue damage therapies
US9175261B2 (en) * 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
ES2391034T3 (en) 2005-12-19 2012-11-20 Ethicon, Inc. In vitro expansion of postpartum derived cells in rotary bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US20070160588A1 (en) 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
PT2471905T (en) 2005-12-29 2019-01-11 Celularity Inc Placental stem cell populations
EP1989318B1 (en) * 2006-01-06 2014-07-30 Agilent Technologies, Inc. Reaction buffer composition for nucleic acid replication with packed dna polymerases
ES2537641T3 (en) 2006-03-23 2015-06-10 Pluristem Ltd. Methods of cell expansion and uses of cells and conditioned media produced in this way for therapy
US20080064098A1 (en) * 2006-06-05 2008-03-13 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
CA2673521C (en) * 2006-10-12 2017-01-17 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
ES2524443T3 (en) * 2006-11-13 2014-12-09 DePuy Synthes Products, LLC In vitro expansion of postpartum cells using microcarriers
CN103356711A (en) * 2007-02-12 2013-10-23 人类起源公司 Immunomodulation 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
CN102036688B (en) 2007-10-05 2014-07-02 伊西康公司 Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
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
TWI363851B (en) * 2008-03-28 2012-05-11 Ind Tech Res Inst Lighting device and assembling method thereof
DK2379087T3 (en) 2008-12-19 2014-11-10 Depuy Synthes Products Llc Cells derived from umbilical cord tissue for the treatment of neuropathic pain and spasticity
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
AU2009327383B2 (en) 2008-12-19 2014-08-28 DePuy Synthes Products, LLC Regeneration and repair of neural tissue following injury
AU2009327384B2 (en) 2008-12-19 2014-07-10 DePuy Synthes Products, LLC Treatment of lung and pulmonary diseases and disorders
EP2411504B1 (en) 2009-03-26 2017-05-10 DePuy Synthes Products, Inc. Human umbilical cord tissue cells as therapy for alzheimer's disease
JP2014506716A (en) * 2011-01-19 2014-03-17 イー・アイ・デュポン・ドウ・ヌムール・アンド・カンパニー Lithium battery separator with shutdown function

Also Published As

Publication number Publication date
US10758576B2 (en) 2020-09-01
EP2399990A1 (en) 2011-12-28
EP2336298A3 (en) 2011-08-03
EP1641918B1 (en) 2016-01-06
US9579351B2 (en) 2017-02-28
AU2004252570B2 (en) 2011-09-15
US7524489B2 (en) 2009-04-28
JP2007528706A (en) 2007-10-18
JP4950660B2 (en) 2012-06-13
PL1641914T3 (en) 2017-01-31
PL1641915T3 (en) 2017-01-31
ES2541604T3 (en) 2015-07-22
JP5148873B2 (en) 2013-02-20
PL2341131T3 (en) 2016-01-29
PL2322599T3 (en) 2015-10-30
US20170143770A1 (en) 2017-05-25
ES2569780T3 (en) 2016-05-12
EP2338981B1 (en) 2015-04-22
ES2542070T3 (en) 2015-07-30
ES2597837T3 (en) 2017-01-23
PL2338980T3 (en) 2016-04-29
EP2341131B1 (en) 2015-08-05
US20050058631A1 (en) 2005-03-17
EP1641917A2 (en) 2006-04-05
US7560276B2 (en) 2009-07-14
CA2530416C (en) 2015-08-25
US10220059B2 (en) 2019-03-05
ES2564045T3 (en) 2016-03-17
US20070014771A1 (en) 2007-01-18
JP2007528705A (en) 2007-10-18
CA2530412C (en) 2016-02-09
PL1641917T3 (en) 2016-11-30
AU2004252570C1 (en) 2012-03-01
EP2338980B1 (en) 2015-04-22
EP1641914B1 (en) 2016-07-20
AU2004254616B2 (en) 2011-09-01
US20190192577A1 (en) 2019-06-27
AU2004252571B2 (en) 2011-09-15
US20070036767A1 (en) 2007-02-15
US10500234B2 (en) 2019-12-10
US9498501B2 (en) 2016-11-22
ES2565582T3 (en) 2016-04-05
JP4948166B2 (en) 2012-06-06
PL2336298T3 (en) 2016-08-31
PL2399990T3 (en) 2015-12-31
US20060234376A1 (en) 2006-10-19
US20170166863A1 (en) 2017-06-15
ES2554343T3 (en) 2015-12-18
PL1641918T3 (en) 2016-06-30
AU2004252567B2 (en) 2011-10-06
EP1641916A2 (en) 2006-04-05
EP2322599A3 (en) 2011-07-27
JP2007528703A (en) 2007-10-18
ES2542069T3 (en) 2015-07-30
JP2007521008A (en) 2007-08-02
US20060153818A1 (en) 2006-07-13
US11179422B2 (en) 2021-11-23
US20070009494A1 (en) 2007-01-11
AU2004281371B2 (en) 2012-05-10
EP1641915B1 (en) 2016-07-27
US20050032209A1 (en) 2005-02-10
JP2007521009A (en) 2007-08-02
US8318483B2 (en) 2012-11-27
AU2004252571A1 (en) 2005-01-06
EP2338982A3 (en) 2011-08-03
US20140045263A1 (en) 2014-02-13
CA2530732C (en) 2015-03-31
US20060154367A1 (en) 2006-07-13
WO2005003334A3 (en) 2005-04-07
AU2004281371C1 (en) 2012-10-11
JP2007521793A (en) 2007-08-09
AU2004252570A1 (en) 2005-01-06
EP2338981A2 (en) 2011-06-29
WO2005001076A2 (en) 2005-01-06
WO2005001080A2 (en) 2005-01-06
EP2338981A3 (en) 2011-08-03
CA2530533A1 (en) 2005-01-13
AU2004252566B2 (en) 2011-09-15
WO2005001079A2 (en) 2005-01-06
ES2552226T3 (en) 2015-11-26
ES2550267T3 (en) 2015-11-05
ES2568463T3 (en) 2016-04-29
US20050054098A1 (en) 2005-03-10
EP1641917B1 (en) 2016-04-27
JP4948165B2 (en) 2012-06-06
US20170166864A1 (en) 2017-06-15
WO2005001078A3 (en) 2005-04-21
US20100260843A1 (en) 2010-10-14
CA2530421A1 (en) 2005-01-06
WO2005038012A2 (en) 2005-04-28
JP2007528702A (en) 2007-10-18
EP1649013B1 (en) 2016-01-20
US20050058630A1 (en) 2005-03-17
US20130022585A1 (en) 2013-01-24
EP2322599B1 (en) 2015-04-15
US8277796B2 (en) 2012-10-02
US9717763B2 (en) 2017-08-01
CA2530255C (en) 2015-12-08
US20060153816A1 (en) 2006-07-13
EP2338982A2 (en) 2011-06-29
WO2005038012A3 (en) 2005-09-15
ES2600555T3 (en) 2017-02-09
EP1641913A2 (en) 2006-04-05
EP1641913B1 (en) 2016-01-06
EP1641915A2 (en) 2006-04-05
EP1649013A2 (en) 2006-04-26
ES2564044T3 (en) 2016-03-17
PL2338981T3 (en) 2016-01-29
US20180338999A1 (en) 2018-11-29
CA2530732A1 (en) 2005-01-06
US10195233B2 (en) 2019-02-05
WO2005001076A3 (en) 2005-03-31
US11191789B2 (en) 2021-12-07
US10039793B2 (en) 2018-08-07
AU2004252571C1 (en) 2012-03-01
US8658152B2 (en) 2014-02-25
US9504719B2 (en) 2016-11-29
EP2338980A3 (en) 2011-08-10
WO2005001077A3 (en) 2005-03-03
US20050019865A1 (en) 2005-01-27
EP2336298A2 (en) 2011-06-22
US7510873B2 (en) 2009-03-31
JP4950659B2 (en) 2012-06-13
US20050037491A1 (en) 2005-02-17
EP2322599A2 (en) 2011-05-18
US20100210013A1 (en) 2010-08-19
CA2530422A1 (en) 2005-01-06
CA2530416A1 (en) 2005-01-06
US7413734B2 (en) 2008-08-19
US20050058629A1 (en) 2005-03-17
US20060188983A1 (en) 2006-08-24
EP1641918A2 (en) 2006-04-05
EP1641914A2 (en) 2006-04-05
EP2338980A2 (en) 2011-06-29
WO2005001077A2 (en) 2005-01-06
PL1649013T3 (en) 2016-07-29
WO2005003334A2 (en) 2005-01-13
WO2005001080A3 (en) 2005-03-24
AU2004252566A1 (en) 2005-01-06
AU2004254616C1 (en) 2012-03-01
EP1641916B1 (en) 2016-02-17
EP2338982B1 (en) 2015-08-26
PL1641916T3 (en) 2016-08-31
AU2004254616A1 (en) 2005-01-13
EP2341131A2 (en) 2011-07-06
US10383898B2 (en) 2019-08-20
EP2399990B1 (en) 2015-07-22
CA2530422C (en) 2016-01-05
JP4950661B2 (en) 2012-06-13
EP2336298B1 (en) 2016-02-17
US20060154366A1 (en) 2006-07-13
US20170239299A1 (en) 2017-08-24
US11000554B2 (en) 2021-05-11
CA2530533C (en) 2015-02-10
US20060153817A1 (en) 2006-07-13
ES2582342T3 (en) 2016-09-12
WO2005001079A3 (en) 2005-04-28
PL1641913T3 (en) 2016-06-30
CA2530421C (en) 2015-04-21
JP4948164B2 (en) 2012-06-06
AU2004252568A1 (en) 2005-01-06
WO2005001078A2 (en) 2005-01-06
EP2341131A3 (en) 2011-08-03
US20170326183A1 (en) 2017-11-16
CA2530255A1 (en) 2005-01-06
US8703121B2 (en) 2014-04-22
AU2004252567A1 (en) 2005-01-06
PL2338982T3 (en) 2016-01-29
AU2004281371A1 (en) 2005-04-28
AU2004252568B2 (en) 2011-06-30

Similar Documents

Publication Publication Date Title
CA2530412C (en) Cartilage and bone repair and regeneration using postpartum-derived cells
US20060166361A1 (en) Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
WO2006071773A2 (en) Treatment of osteochondral diseases using postpartum-derived cells and products thereof

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20220627