US20040171147A1 - Cytotherapeutics, cytotherapeutic units and methods for treatments using them - Google Patents

Cytotherapeutics, cytotherapeutic units and methods for treatments using them Download PDF

Info

Publication number
US20040171147A1
US20040171147A1 US10/721,144 US72114403A US2004171147A1 US 20040171147 A1 US20040171147 A1 US 20040171147A1 US 72114403 A US72114403 A US 72114403A US 2004171147 A1 US2004171147 A1 US 2004171147A1
Authority
US
United States
Prior art keywords
unit
cytotherapeutic
cells
cell
potent
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.)
Abandoned
Application number
US10/721,144
Inventor
Robert Hariri
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.)
Celularity 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
Priority to US10/721,144 priority Critical patent/US20040171147A1/en
Assigned to ANTHROGENESIS CORPORATION reassignment ANTHROGENESIS CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HARIRI, ROBERT J.
Publication of US20040171147A1 publication Critical patent/US20040171147A1/en
Priority to US11/592,544 priority patent/US8617535B2/en
Priority to US15/362,300 priority patent/US20170290862A1/en
Assigned to CLARITY ACQUISITION II LLC reassignment CLARITY ACQUISITION II LLC MERGER (SEE DOCUMENT FOR DETAILS). Assignors: ANTHROGENESIS CORPORATION
Assigned to Celularity, Inc. reassignment Celularity, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CLARITY ACQUISITION II LLC
Priority to US16/532,080 priority patent/US20200009199A1/en
Abandoned legal-status Critical Current

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
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/14Drugs for genital or sexual disorders; Contraceptives for lactation disorders, e.g. galactorrhoea
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • 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
    • A61P19/08Drugs 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
    • 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
    • A61P25/14Drugs 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
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Definitions

  • Cytotherapeutic therapy involves the introduction of immature cells, especially stem cells, into a patient in order to secure palliation, amelioration or cure of a disease state.
  • the present invention is also directed to improved cytotherapeutic agents, to methods of producing them, to unit dosage forms of such agents and to novel paradigms for administering cytotherapeutic units to patients in need of therapy.
  • stem cells have been known heretofore to administer certain types of stem cells to humans and to animals in order to achieve a therapeutic end. Much of this has been done with stem cells from adults, such as those found in adult bone marrow, especially for the repopulation of depopulated interosseous spaces, which attend aggressive chemotherapy or radiation therapy, e.g., for treatment of certain cancers. Indeed, such cytotherapy has become relatively widespread and has achieved a level of success despite limitations including the lack of standardization as to cell numbers and types.
  • stem cell preparations from most sources, including from neonatal cord blood include a diverse population of cells with differing potentials for effective therapy and often do not contain a sufficient number of cells for an optimized therapeutic dose, particularly for an averaged size adult undergoing a transplant for leukemia, for example. It is believed that different scientific and medical groups likely achieve differing preparations with differing characteristics, even when supposedly following the same or similar protocols. Presently, most independent preparations, even those done by the same individual, can have different compositions with the specifics of the compositions undetermined. In short, there is a complete lack of unit to unit reproducibility and little standardization in the cellular units used in transplants.
  • cytotherapeutic unit refers to a cell preparation comprising a plurality of potent cells in which at, least one cell type has been tailored for a particular patient or particular disease state. Tailoring may include having a minimum number of said cell type or, alternatively, removal of a portion or all of said cell type.
  • Patent with respect to a cell or cell type, means that the cell or cell type is capable of differentiation into at least one type of cell.
  • “Pluripotent,” with respect to a cell or cell type, means that the cell or cell type is capable of differentiation into at least two different types of cells.
  • Antigenic determinant refers to the set of antigenic regions on the surface of a cell.
  • “Factor” refers to a cell type by reference to its antigenic determinant.
  • Exemplary factors include CD34, CD8, CD10 and the like.
  • a cell or cell preparation may also be considered to be positive or negative in regard to a particular factor by reference to whether or not a particular cell or cell type exhibits the characteristics of that particular factor.
  • the present invention provides for cytotherapeutic units comprising a plurality of potent cells, the contents of which are known with respect to the identities and numbers of at least some of the potent cells. To ensure that the identities and numbers of at least some of the potent cells are accurate at least one assay is performed. In some preferred embodiments, the provider of the unit certifies the accuracy of the assay. In other embodiments, the potent cells for which the identities and numbers are known are pluripotent cells. The identities of the potent cells preferably reflect the presence or absence of at least one antigenic determinant on the cells. In some embodiments, the cytotherapeutic unit comprises at least some potent cells exhibiting CD34, CD8, CD10, OCT4, CD38, CXCR4, or CD117, for example.
  • the cells may also exhibit CD33.
  • the cytotherapeutic unit comprises cells that lack specific antigenic determinants.
  • at least one identified potent cell that is derived from a source is specifically excluded or removed from the cellular preparation.
  • some or all cells may be characterized by the presence of one or more of the following cell surface markers: CD10+, CD29+, CD34 ⁇ , CD38 ⁇ , CD44+, CD45 ⁇ , CD54+, CD90+, SH2+, SH3+, SH4+, SSEA3 ⁇ , SSEA4 ⁇ , OCT ⁇ 4+, and ABC ⁇ p+.
  • the potent cells may be obtained from fetal cord blood or other fetal tissue.
  • potent cells are obtained from placenta, especially postpartum placenta, which has been metabolically supported and nurtured. Potent cells are preferably obtained from postpartum placenta perfusate.
  • the present invention also provides for cytotherapeutic units wherein the potent cells are derived from a plurality of sources. In some embodiments, the potent cells are derived from at least two individuals, at least five individuals, or at least ten individuals.
  • the unit comprises at least one cell that is autologous. In some other embodiments, the unit comprises at least one cell that is exogenous. In some embodiments the unit comprises a chimera of autologous and allogeneic cells. In another embodiment at least some of the cells are genetically modified.
  • the plurality of potent cells is selected to render the unit suitable for therapy for an indicated disease state or condition and/or the severity of the condition.
  • the cytotherapeutic units comprise a minimum number of preselected types of potent cells and may be based, for example, on the weight of the particular patient or that patient's medical status.
  • the cytotherapeutic unit is assayed to ensure the accuracy of its contents of preselected types of potent cells.
  • the contents of the preselected potent cells in the cytotherapeutic unit are certified.
  • the cytotherapeutic unit can be one of a group of substantially identical units wherein the additional units are stored for future transplants so that, if needed, the patient can receive a unit identical to one previously transplanted.
  • the additional like-units may be altered to optimize future transplants for that same patient.
  • At least one type of cell is excluded from the cytotherapeutic unit comprising preselected potent cells.
  • the cytotherapeutic unit is preferably certified as to its contents of the preselected potent cells and the absence of the types of cells to be excluded.
  • the identity and the numbers of a plurality of potent cells being selected to render the cytotherapeutic unit suitable for therapy for an indicated disease state or condition is certified.
  • the certification is preferably of a plurality of potent cell types, wherein the plurality and the numbers of each of said plurality being selected as well as excluded renders the cytotherapeutic unit suitable for therapy for an indicated disease state or condition.
  • the present invention provides for kits for the treatment of a person suspected of having a disease state or condition.
  • the kit preferably comprises a cytotherapeutic unit comprising a plurality of potent cells.
  • the kit comprises a cytotherapeutic unit wherein at least one type of cell that has been excluded from the cytotherapeutic unit.
  • the kit comprises potent cells wherein at least some of the potent cells have been identified and counted.
  • the kit comprises a unit that has been assayed to ensure the accuracy of the identities and numbers of the potent cells. In some more preferred embodiments of the kit, the accuracy of the assay has been certified.
  • kits for the treatment of a person suspected of having a disease state or condition comprising a cytotherapeutic unit having minimum numbers of identified potent cells and a certification of the potent cell composition.
  • the kits may also contain equipment or devices for administering the unit to the patient, materials for monitoring the administration and other attendant things.
  • the present invention provides for cytotherapeutic units comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, wherein at least one type of cell has been removed from the unit. In some embodiments, a plurality of cell types has been removed from the unit.
  • the present invention provides for a cytotherapeutic unit comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, wherein said cells comprise a plurality of different types.
  • a cytotherapeutic unit comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, wherein said cells comprise a plurality of different types.
  • the different types of cells are separated into components.
  • the components are recombined into the unit. It is preferred in some aspects of the invention that components are used to supplement a cytotherapeutic unit with a specific potent cell type.
  • the separated components can be frozen separately or otherwise stored prior to recombination.
  • the cytotherapeutic unit itself has been placed in a frozen state.
  • the separated cell types have been identified and/or counted.
  • the present invention provides methods of treating a disease in a mammal comprising administering to the mammal a therapeutically effective amount of a composition comprising a cytotherapeutic unit.
  • the unit used to treat the disease state or condition comprises a plurality of potent cells wherein the content of the unit is known with respect to the identities and numbers. At least some of the cells in the unit are assayed to ensure the accuracy of the identities and the numbers of the potent cells.
  • the cytotherapeutic unit is administered multiple times. In other cases, administering multiple doses of the cytotherapeutic units that are derived from different individuals or sources may be performed.
  • the methods may also comprise administering multiples doses of the cytotherapeutic unit that is derived from one individual.
  • the present invention provides for cytotherapeutic units comprising a plurality of potent cells with the content of the cytotherapeutic unit being known with respect to the identities and numbers of at least some of the potent cells.
  • the identities of the potent cells in the cytotherapeutic unit are an aspect of the invention that is important for the reliability and the quality of the unit being used.
  • the potent cells can be identified by any number of methods and based on any set of criteria that a person of ordinary skill may find useful. One such method is to identify the potent cells based on the presence of antigenic determinants on the surface of the cell.
  • Antigenic determinants can be any molecule that is recognizable by an antibody. Some examples of antigenic determinants include polypeptides, lipids, glycoproteins, sugars, and the like.
  • the cells may be characterized by the presence of one or more of the following cell surface markers: CD10+, CD29+, CD34 ⁇ , CD38 ⁇ , CD44+, CD45 ⁇ , CD54+, CD90+, SH2+, SH3+, SH4+, SSEA3 ⁇ , SSEA4 ⁇ , OCT ⁇ 4+, and ABC ⁇ p+.
  • potent cells may be identified by the presence of antigenic determinants or by certain expressed factors, it can be equally important to identify a cell based on what antigenic determinants the cell lacks. For example, it is known that the presence of certain determinants may lower the chances of a successful treatment and therefore, a person using the cytotherapeutic unit would want to know that the unit being used lacks certain antigenic determinants. Furthermore, the presence or absence of antigenic factors can aid in determining the maturity level of a particular cell or cell-type. A less mature cell has a wider range of differentiation and is therefore, potentially more useful. Depending on the use of the cytotherapeutic unit, different levels of differentiation of the cells may be required. The identification of some of the cells enables a person to obtain a unit, that when used, results in a better clinical outcome.
  • Methods to determine the presence or absence of antigenic factors on or in a cell are well known in the art. These methods include fluorescence activated cell sorting (FACS), Enzyme-Linked Immuno Sorbent Assay (ELISA), western blot, polymerase chain reaction (PCR), reverse-transcribed PCR (RT-PCR), and the like. The precise method or methods used to identify the potent cells is not essential.
  • FACS fluorescence activated cell sorting
  • ELISA Enzyme-Linked Immuno Sorbent Assay
  • PCR polymerase chain reaction
  • RT-PCR reverse-transcribed PCR
  • the methods to determine the genetic profile of a cell are well known to those of ordinary skill in the art. Any method used is sufficient, but some examples of methods or techniques that can be used to determine the genetic makeup of a cell include, without limitation, PCR, RT-PCR, northern blot, southern blot, single nucleotide polymorphism (SNP) analysis, gene-chip expression analysis, serial analysis of gene expression (SAGE), nucleotide sequencing, FACS, in situ hybridization, and the like.
  • a cell can be identified by any of the above-mentioned criteria: antigenic determinants, genetic makeup, a combination thereof, or a cell can be identified based upon another set of criteria. In some embodiments, at least 0.1%, 1%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or about 100% of the cells are identified.
  • Methods of identification and determining the number of cells include but are not limited to using standard cell detection techniques such as flow cytometry, cell sorting, immunocytochemistry (e.g., staining with tissue specific or cell-marker specific antibodies), FACS, magnetic activated cell sorting (MACS), by examination of the morphology of cells using light or confocal microscopy, or by measuring changes in gene expression using techniques well known in the art, such as PCR and gene expression profiling.
  • standard cell detection techniques such as flow cytometry, cell sorting, immunocytochemistry (e.g., staining with tissue specific or cell-marker specific antibodies), FACS, magnetic activated cell sorting (MACS), by examination of the morphology of cells using light or confocal microscopy, or by measuring changes in gene expression using techniques well known in the art, such as PCR and gene expression profiling.
  • relevant determinations can be made by techniques including, but not limited to, optical and electrooptical properties, morphological imaging methods, optophoresis (www.genoptix.com) microwave spectroscopy (Signature Bioscience www.signaturebio.com) and optical tweezers. Other methods may also be employed.
  • the present invention provides for cytotherapeutic units that have at least one cell type that is excluded.
  • the cell-type that is excluded will not always be the same.
  • all CD34 positive cells will be excluded.
  • all CD8 positive cells will be excluded.
  • multiple cell types are excluded.
  • the term “exclusion” or “elimination” as used in this context preferably means at least about 75% reduction in the number of a certain cell type in a cell preparation.
  • At least about 90% reduction is achieved, with at least about 95% reduction being even more preferred.
  • Essentially complete elimination is, of course, most desirable, although the same may be achievable in some cases.
  • the foregoing percentage reductions relate to numbers of cells relative to an original population of such cells using any appropriate assay.
  • Cell types can be excluded or reduced either by selecting cell-containing units which, naturally do not contain them (or many of them) or by employing a process that specifically removes selected cell-types. It is preferred to exclude cell types having antigenic determinants which are inconsistent with the therapeutic modality planned for the cytotherapeutic unit. For example, but not by way of limitation, T-lymphocytes and mature dendritic cells may be excluded to lower the expectation of graft versus host disease. In the treatment of adrenal leukodysplasia it may be desirable to delete some or all CD8 positive cells.
  • a cell-type can be excluded by a process that is used either before or after the cells are extracted from a source. Processes or methods that are used to exclude a specific cell-type are well known to the art-skilled. Examples of processes or methods include: FACS, centrifugation, immunochromatography, and the like.
  • the cells may be sorted using a fluorescence activated cell sorter (FACS).
  • Fluorescence activated cell sorting is a well-known method for separating particles, including cells, based on the fluorescent properties of the particles (Kamrach, 1987, Methods Enzymol, 151:150-165). Laser excitation of fluorescent moieties in the individual particles results in a small electrical charge allowing electromagnetic separation of positive and negative particles from a mixture.
  • cell surface marker-specific antibodies or ligands are labeled with distinct fluorescent labels. Cells are processed through the cell sorter, allowing separation of cells based on their ability to bind to the antibodies used.
  • FACS sorter particles may be directly deposited into individual wells of 96-well or 384-well plates to facilitate separation and cloning.
  • Reagents for cell surface markers or cluster designated reagents are available from a variety of sources including Becton Dickinson and Cell Pro Inc., for example.
  • Available reagents include but are not limited to reagents for identifying: CD1a; CD2; CD3; CD4; CD4 (Multi-Clone); CD4 v4; CD5; CD7; CD8 (Leu-2a); CD8 (Leu-2b); CD10 (Anti-CALLA); CD11a (Anti-LFA-1 ⁇ ); CD11b; CD11c; CD13; CD14; CD15; CD16 (Leu-11a, 11b, 11c); CD18 (Anti-LIFA-1 ⁇ ); CD19 (Leu-12); CD19(SJ25C1); CD20; CD21(Anti-CR 2 ); CD22; CD23; CD25(Anti-IL-2R); CD26; CD27; CD28; CD31(Anti-PECAM-1); CD33; CD34(Anti-HPCA-1&2); CD38; CD42a(Anti-gpIX); CD44; CD45(Anti-Hle-1); CD45RA; CD45RO; CD49d(Anti-VLA- ⁇ 4); CD
  • Non-cluster designated reagents include: Anti-BrdU; Anti-Cytokeratin (CAM 5.2); Anti-HER-2/neu; Anti-HLA-DP; Anti-HLA-DQ; Anti-HLA-DR; Anti-Hu KIR (NKB1); Anti-IgA 2 ; Anti-IgD; Anti-IgG; Anti-IgM (Ig Heavy Chain); Anti-Kappa (Ig Light Chain); Anti-Kappa F(ab′) 2 ; Anti-Lambda (Ig Light Chain); Anti-Lambda F(ab′) 2 ; Anti-P-glycoprotein (P-gp); Anti-TCR ⁇ / ⁇ -1 (WT31); Anti-TCR- ⁇ / ⁇ -1; PAC-1; Lineage Cocktail 1 (lin1) FITC.
  • the skilled artisan will use those reagents required for his/her particular needs in order to optimize the desired cytotherapeutic unit or tailor it for a particular patient or use.
  • magnetic beads can be used to separate cells.
  • the cells may be sorted using a magnetic activated cell sorting (MACS) technique, a method for separating particles based on their ability to bind magnetic beads (0.5-100 ⁇ m diameter).
  • a variety of useful modifications can be performed on the magnetic microspheres, including the covalent addition of an antibody which specifically recognizes a cell-solid phase surface molecule or hapten.
  • a magnetic field is then applied, to physically manipulate the selected beads.
  • the beads are then mixed with the cells to allow binding.
  • Cells are then passed through a magnetic field to separate out cells having cell surface markers. These cells can then isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers.
  • the cells are again passed through a magnetic field, isolating cells that bound both the antibodies.
  • Such cells can then be diluted into separate dishes, such as microtiter dishes for clonal isolation, if desired.
  • Knowing the composition of the cytotherapeutic unit will help fulfill the long-felt need of a reliable and certified cytotherapeutic unit.
  • the number of the cells will be determined at the same time the identities are determined, but the numbers can also be determined before or after the identities of some of the potent cells are determined.
  • the numbers can also be determined before or after the identities of some of the potent cells are determined.
  • Cytotherapeutic units can now be prepared that have a minimum numbers of preselected cells. It is also now possible to ensure that other cell types are excluded from the-units.
  • the cytotherapeutic unit will comprise at least about 100 selected potent cells. Such units having at least about 1,000 such cells are preferred, with at least about 10,000 being more preferred. Greater numbers of selected cells are still more preferred, especially when it is intended that the unit be administered to the same or different individuals a plurality of times. Thus, selected cell populations greater than about 100,000 or even about 500,000 can be useful. It is preferred that some or all of the cells in the unit be identified through assay and that the same be reflected in a certification of such presence. This certification ensures uniform and effective therapeutic application.
  • the cytotherapeutic units will have a minimum number of different, specific cell types. Advantages to having a minimum number of specific cell types are that it may improve the efficacy of the cytotherapeutic unit.
  • the cytotherapeutic unit could be assayed to comprise at least about 1,000 OCT4 positive cells, either with or without known quantities of other desirable cell types.
  • the unit may be caused to comprise specific percentages of CD34 positive cells, measured by reference to all nucleated cells in the preparation.
  • such preparations may contain at least 0.01%, 0.1%, 1%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or other percentages of CD34 positive cells may be made. Similar, known percentages of cells having other antigenic determinants or specific factors may, likewise, be created.
  • cytotherapeutic units comprising cells that have been derived from at least one source, wherein the source's cells have been separated into components.
  • components is synonymous to cell-types, identified cells, and the like.
  • Methods to separate cellular preparations into components include, without limitation, FACS, centrifugation, chromatography, HPLC, FPLC, and the like.
  • cytotherapeutic units can comprise components that are recombined.
  • at least one component is used in a cytotherapeutic unit.
  • at least two, at least three, at least 4, at least 5, at least 10, at least 100 components are recombined to make the cytotherapeutic unit.
  • the components of each source be known in terms of identity and relative numbers, with some cell types preferably being excluded from some or all of the components. It may be seen that the different components may be maintained separately,; e.g. frozen, and that the same may form a “formulary” or “library” of cells of known identity and abundance for formulation into combined cytotherapeutic units.
  • cytotherapeutic unit Separating the respective cellular preparations into components allows a cytotherapeutic unit to be created that has a specific composition both in terms of cells present and in types of cells excluded. Additionally, this allows an existing cytotherapeutic unit to be supplemented with a specific cell-type or component as may be indicated for a specific therapeutic modality.
  • cytotherapeutic units of the invention may be seen to comprise cells derived from one source or from many sources. Contrary to prevailing practice, it is believed that there are great benefits to providing cells from a plurality of sources and that therapeutic benefit and efficacy will derive therefrom.
  • the cells are derived from multiple sources and may derive from multiple organs in such sources.
  • the term “source” refers to any organism, tissue, or organ from which cells are derived or extracted.
  • the sources are fetal cord blood, fetal tissue, placenta, postpartum placenta, postpartum placenta perfusate, or a mixture thereof.
  • the cells are stimulated to proliferate, for example, by administration of erythropoietin, cytokines, lymphokines, interferons, colony stimulating factors (CSF's), interferons, chemokines, interleukins, recombinant human hematopoietic growth factors including ligands, stem cell factors, thrombopoeitin (TPO), interleukins, and granulocyte colony-stimulating factor (G-CSF) or other growth factors.
  • CSF's colony stimulating factors
  • chemokines chemokines
  • interleukins recombinant human hematopoietic growth factors including ligands, stem cell factors, thrombopoeitin (TPO), interleukins, and granulocyte colony-stimulating factor (G-CSF) or other growth factors.
  • G-CSF granulocyte colony-stimulating factor
  • cells are genetically engineered, for example, using a viral vector such as an adenoviral or retroviral vector, or by using mechanical means such as liposomal or chemical mediated uptake of the DNA.
  • a viral vector such as an adenoviral or retroviral vector
  • mechanical means such as liposomal or chemical mediated uptake of the DNA.
  • a vector containing a transgene can be introduced into a cell of interest by methods well known in the art, e.g., transfection, transformation, transduction, electroporation, infection, microinjection, cell fusion, DEAE extran, calcium phosphate precipitation, liposomes, LIPOFECTINTM, lysosome fusion, synthetic cationic lipids, use of a gene gun or a DNA vector transporter, such that the transgene is transmitted to daughter cells, e.g., the daughter embryonic-like stem cells or progenitor cells produced by the division of an embryonic-like stem cell.
  • daughter cells e.g., the daughter embryonic-like stem cells or progenitor cells produced by the division of an embryonic-like stem cell.
  • the cytotherapeutic units will preferably comprise minimum numbers of preselected types of potent cells and be certified as such.
  • preselected refers to the process of selecting the types of potent cells that are to be in the cytotherapeutic unit before it is administered. Preselecting the types of potent cells that will have a minimum number of those cells in the cytotherapeutic unit allows the cytotherapeutic unit to be tailored to a composition desired to achieve a specific therapeutic result in an individual or class of individuals. Likewise, certification as to the absence of other preselected types of cells is preferred for similar reasons.
  • the plurality of potent cells and of cell types present in the cytotherapeutic units of the invention are selected to render the units suitable for therapy for an indicated disease state or condition.
  • the phrase “selected to render” refers to the process of deciding that a cytotherapeutic unit comprising a plurality of potent cells is suitable for therapy. This decision can be based on the numbers of potent cells present in the cytotherapeutic unit. As discussed hereinbefore, the number of cells appears to be critical for the success rate of treating an individual or patient with cytotherapy. Therefore, not all cytotherapeutic units may be suitable for therapy for an indicated disease state or condition. Additionally, the types of potent cells will also aid in the decision process on whether or not a cytotherapeutic unit is suitable for therapy.
  • Certain types of potent cells can be detrimental or beneficial to the treatment of a specific disease state or condition.
  • the types of cells present in the unit can be another factor that is used to select a unit suitable for therapy.
  • the criteria that are used to select a unit that is suitable for therapy is not specific to those mentioned above. Any set of criteria can be used to decide whether or not a plurality of potent cells present in a cytotherapeutic unit are selected to render the unit suitable for therapy of an indicated disease state or condition.
  • the present invention provides for cytotherapeutic units wherein at least some of the potent cells present in the unit are identified and counted.
  • the units' contents must be preferably assayed to ensure the accuracy of the identities and numbers.
  • the assays can be done by the same group, individual, or machine that had determined the identities and the numbers of at least some of the potent cells in the cytotherapeutic units.
  • the assays can be performed by a different individual, group, or machine that had determined the identities and numbers of some of the potent cells. In some embodiments, only one assay needs to be performed to ensure the accuracy of the identities and the numbers.
  • At least 2, at least 5, or at least 10 assays are performed to ensure the accuracy of the identities and the numbers of the potent cells.
  • the types of assays to be done can be the same assay that was used to determine the numbers and the identities previously.
  • different assays are used to ensure the accuracy of the numbers and identities of some of the potent cells.
  • Some assays that can be used to ensure the accuracy include, without limitation, ELISA, FACS, western blot, and the like.
  • the provider of the unit certifies the accuracy of the assay.
  • the term “provider” refers to an individual, business, or facility that is providing the cytotherapeutic unit to the individual that is using the unit.
  • the certification comprises a written statement indicating that the assay was performed correctly and that the results are correct.
  • the certification comprises results from an assay done on a positive control to show that the assay was functioning properly.
  • the certification comprises both the results of the positive control and a written statement that the assay was functioning properly.
  • the certification comprises a list of the types of potent cells that have been excluded from the cytotherapeutic unit.
  • the certification comprises a list of at least some of the types of potent cells that are contained in the cytotherapeutic unit. In some embodiments, the certification comprises the numbers of all the cells. In some embodiments, the certification further comprises the quantity of at least some of the specific cell types. In some other embodiments, the certification comprises a list of the types of at least some of the potent cells that have been added to the unit to supplement the potent cells so that the unit comprises minimum numbers of potent cells.
  • kits for the treatment of a person suspected of having a disease state or condition comprising a cytotherapeutic unit comprising a plurality of potent cells with the content of the unit being known with respect to the identities and numbers of at least some of the potent cells. Additionally, the cytotherapeutic unit is assayed to ensure the accuracy of the identities and numbers of the potent cells. The kits further comprise a certification of the accuracy of the assay. In some embodiments, the kits comprise a cytotherapeutic unit having minimum numbers of identified potent cells and a certification of the potent cell composition of the unit. In some other embodiments, the kits comprise cytotherapeutic units that have at least one cell-type that has been excluded.
  • the present invention also provides for methods of treating a disease state or condition in a mammal.
  • the methods comprise administering to the mammal a therapeutically effective amount of a composition comprising a cytotherapeutic unit comprising potent cells, wherein some of the potent cells are known with respect to their identities and numbers.
  • the unit is also assayed to ensure the accuracy of the identities and the numbers.
  • the cytotherapeutic unit comprises minimum numbers of preselected types of potent cells.
  • a therapeutically effective amount for a mammal can vary, but for example could be approximately 0.01 cytotherapeutic units/kg to 100 units/kg.
  • the cytotherapeutic unit can be administered rapidly or slowly to the mammal. In some embodiments, the cytotherapeutic unit is administered at a rate of approximately 0.01 ⁇ l/minute, and in other embodiments, the unit is administered at a rate of approximately 100,000 ml/minute.
  • the unit can be administered, for example, intravenously, subcutaneously, intramuscularly, orally, or rectally. In some embodiments, the unit is administered multiple times to the mammal at different times. In some other embodiments, cytotherapeutic units derived from different sources or different individuals are administered to the mammal.
  • cytotherapeutic units are limitless, but some examples of disease states or conditions that cytotherapeutic units can be used to treat include cancer, acute leukemia, chronic leukemia as well as other cancers presently treated with bone marrow or cord blood cell transplants, myelodysplastic syndrome, stem cell disorder, myeloproliferative disorder, lymphoproliferative disorder, phagocyte disorder, liposomal storage disorder, histiocytic disorder, inherited erythrocyte abnormality, congenital (inherited) immune system disorder, inherited platelet abnormality, plasma cell disorder, Lesch-Nyhan Syndrome, Cartilage-Hair Hypoplasia, Glanzmann Thrombastenia, osteoporosis, breast cancer, Ewing Sarcoma, neuroblastoma, renal cell carcinoma, lung cancer, Alzheimer's disease, liver disease, hepatitis, Parkinson's disease, vision loss, memory loss, and the like.
  • the cytotherapeutic units may be optimized for enzyme replacement therapy to treat specific diseases or conditions, including, but not limited to, lysosomal storage diseases, such as Tay-Sachs, Niemann-Pick, Fabry's, Gaucher's, Hunter's, and Hurler's syndromes, as well as other gangliosidoses, mucopolysaccharidoses, and glycogenoses.
  • lysosomal storage diseases such as Tay-Sachs, Niemann-Pick, Fabry's, Gaucher's, Hunter's, and Hurler's syndromes, as well as other gangliosidoses, mucopolysaccharidoses, and glycogenoses.
  • the cytotherapeutic units in this case may be certified that the cells have been assayed to contain the desired number of cells capable of producing the necessary enzyme.
  • Said unit may contain either allogeneic cells containing the functional endogenous gene of the desired enzyme, autologous cells containing exogenous copies of
  • the cells may be used as autologous or heterologous transgene carriers in gene therapy to correct inborn errors of metabolism such as adrenoleukodystrophy, cystic fibrosis, glycogen storage disease, hypothyroidism, sickle cell anemia, Pearson syndrome, Pompe's disease, phenylketonuria (PKU), Tay-Sachs disease, porphyrias, maple syrup urine disease, homocystinuria, mucopolypsaccharidoses, chronic granulomatous disease, and tyrosinemia or to treat cancer, tumors or other pathological conditions.
  • AML acute myelogenous leukemia
  • the patient undergoes traditional chemotherapy followed by conventional preparation for transplant as determined by the patient's health care provider but includes destroying the diseased bone marrow.
  • the patient's weight is determined.
  • Appropriate HLA typing has been done by conventional methods.
  • the transplanter requests and is provided with a cytotherapeutic unit comprising a plurality of potent nucleated cells; the content of said unit being known with respect to the identities and numbers at least some of said plurality; the unit being assayed to ensure the accuracy of said identities and numbers, which is certified.
  • the unit is certified to contain about 1.4 ⁇ 10 7 nucleated cells per kilogram of the patient's body weight. Additional certified information includes HLA information. Because the patient suffers from AML, the cytotherapeutic unit contains no less than one (1) percent of CD34+ of the total nucleated cells and no less than 2.5 percent CD8+ cells to minimize graft versus tumor effect.
  • the transplanter requests twice the total number of cells needed for transplant (1.4 ⁇ 10 7 nucleated cells multiplied by the patient's weight in kilograms ⁇ 2).
  • the transplanter requests the 1 ⁇ amount just prior to the transplant in order to have the number of cells suitable for this transplant.
  • the second half of the cells is to be shipped in the event that a second transplant becomes necessary.
  • the second cytotherapeutic unit is the same as that to be used in the initial transplant.
  • the transplanter may request, based on alterations in the patient's weight, severity of disease or even changes in recommended treatment, that the second cytotherapeutic unit be altered in the appropriate manner (increased number of CD34 positive cells, etc.) and certified.
  • the transplant is performed in the same manner conventionally used by the transplanter.
  • a child having sickle cell anemia is in need of a cell transplant. It is determined that 1.7 ⁇ 10 7 nucleated cells per kilogram of body weight of the child is needed. Appropriate HLA typing is done by conventional methods. It is determined that the cytotherapeutic unit must have no less than 1% CD34+cells of the total nucleated cells. Said CD34+ cells are further described in a ratio of 2:1 as CD34+/CD33+: CD34+/CD33 ⁇ A cytotherapeutic unit having these parameters is provided. This unit comprises cells derived from cord blood as well as pluripotential placental cells such as those described in WO 02/064755, which are derived in the manner described in WO02/064755.
  • the ratio of CD34+/CD33+ cells is 2:1 to CD34+/CD33 ⁇ , a fact which is ascertained by assay and certified as being accurate.
  • the certified cells are determined using FACS; based on the fluorescent properties of the particles, cell surface marker-specific antibodies or ligands are labeled with distinct fluorescent labels.
  • Cells are processed through the cell sorter, allowing separation of cells based on their ability to bind to the antibodies used.
  • Cell surface marker-specific antibodies may be purchased from any company selling such reagents, including Becton Dickinson, for example.
  • the transplant is performed in the same manner conventionally used by the transplanter.
  • a child suffers from adrenal leukodysplasia. It is determined that a cellular transplant is appropriate. It is determined that 2 ⁇ 10 7 nucleated cells (derived from cord blood by a conventional technique) per kilogram of body weight of the child is needed. Appropriate HLA typing is done by conventional methods. A cytotherapeutic unit having these parameters is provided. In particular, the unit is certified to contain no less than 0.25% of CD34+/CD38 ⁇ cells and with no less than 0.5% depletion of CD8+ cells of the total nucleated cells. The transplant is performed in the same manner conventionally used by the transplanter.

Abstract

The present invention provides cytotherapeutic units comprising predetermined numbers of selected types of potent cells. Assurance of the nature and identities of such cells is achieved through assay and certification of said numbers and identities. Therapeutic modalities are provided. Libraries of cell preparations with assayed and preferably certified populations are preferred and the preparation of cell preparations tailored to specific patients or disease states are provided.

Description

    FIELD OF THE INVENTION
  • This Application claims priority to U.S. Provisional Application 60/429,702 filed on Nov. 26, 2002. The present invention is directed to improvements in therapeutics utilizing cytotherapeutic formulations. Cytotherapeutic therapy involves the introduction of immature cells, especially stem cells, into a patient in order to secure palliation, amelioration or cure of a disease state. The present invention is also directed to improved cytotherapeutic agents, to methods of producing them, to unit dosage forms of such agents and to novel paradigms for administering cytotherapeutic units to patients in need of therapy. [0001]
  • BACKGROUND OF THE INVENTION
  • It has been known heretofore to administer certain types of stem cells to humans and to animals in order to achieve a therapeutic end. Much of this has been done with stem cells from adults, such as those found in adult bone marrow, especially for the repopulation of depopulated interosseous spaces, which attend aggressive chemotherapy or radiation therapy, e.g., for treatment of certain cancers. Indeed, such cytotherapy has become relatively widespread and has achieved a level of success despite limitations including the lack of standardization as to cell numbers and types. [0002]
  • Many of these therapeutic regimes employ relatively mature cellular preparations, e.g. bone marrow. While these have a level of therapeutic potential, such cells possess quite a large number of surface antigens and require immunosupression attendant to administration. Additionally, most cells extracted from adult bone marrow are limited in the types of cells into which they can differentiate. There have been a number of reports that have indicated that most stem cells isolated from adult bone marrow are only able to differentiate into blood cells. While this is useful for the treatment of blood related diseases, e.g., leukemia, these cells are not very useful for treating other types of diseases that are localized to a specific type of tissue or organ. An additional problem with bone marrow preparations is that the process of extracting the marrow is often very painful, and although potential donors can be identified many do not consent to the procedure because of the potential for pain and discomfort. [0003]
  • Recently, cytotherapy employing less mature stem cells, such as, for example, those found in neonatal cord blood, has found some success. However, stem cell preparations from most sources, including from neonatal cord blood, include a diverse population of cells with differing potentials for effective therapy and often do not contain a sufficient number of cells for an optimized therapeutic dose, particularly for an averaged size adult undergoing a transplant for leukemia, for example. It is believed that different scientific and medical groups likely achieve differing preparations with differing characteristics, even when supposedly following the same or similar protocols. Presently, most independent preparations, even those done by the same individual, can have different compositions with the specifics of the compositions undetermined. In short, there is a complete lack of unit to unit reproducibility and little standardization in the cellular units used in transplants. [0004]
  • The foregoing practices can give rise to inconsistent therapeutic outcomes from different research and medical centers and make accurate, statistical analyses for cytotherapeutic procedures difficult or impossible to attain. There is, thus, a long-felt need for improved cytotherapeutic materials and procedures, ones amenable to reproducible outcomes and to scientific analysis. It is also desired to improve specificity of cytotherapeutic treatments and to affect improved efficiencies and outcomes. Importantly, there is also a need for unit to unit reproducibility which may further the ability to collect sufficient data to advance the medical area devoted to cellular therapies. The present invention provides solutions for these and other long-felt needs. [0005]
  • SUMMARY OF THE INVENTION
  • As used herein, “cytotherapeutic unit” refers to a cell preparation comprising a plurality of potent cells in which at, least one cell type has been tailored for a particular patient or particular disease state. Tailoring may include having a minimum number of said cell type or, alternatively, removal of a portion or all of said cell type. [0006]
  • “Potent,” with respect to a cell or cell type, means that the cell or cell type is capable of differentiation into at least one type of cell. [0007]
  • “Pluripotent,” with respect to a cell or cell type, means that the cell or cell type is capable of differentiation into at least two different types of cells. [0008]
  • “Antigenic determinant” refers to the set of antigenic regions on the surface of a cell. [0009]
  • “Factor” refers to a cell type by reference to its antigenic determinant. Exemplary factors include CD34, CD8, CD10 and the like. A cell or cell preparation may also be considered to be positive or negative in regard to a particular factor by reference to whether or not a particular cell or cell type exhibits the characteristics of that particular factor. [0010]
  • The present invention provides for cytotherapeutic units comprising a plurality of potent cells, the contents of which are known with respect to the identities and numbers of at least some of the potent cells. To ensure that the identities and numbers of at least some of the potent cells are accurate at least one assay is performed. In some preferred embodiments, the provider of the unit certifies the accuracy of the assay. In other embodiments, the potent cells for which the identities and numbers are known are pluripotent cells. The identities of the potent cells preferably reflect the presence or absence of at least one antigenic determinant on the cells. In some embodiments, the cytotherapeutic unit comprises at least some potent cells exhibiting CD34, CD8, CD10, OCT4, CD38, CXCR4, or CD117, for example. In some embodiments some portion of the cells may also exhibit CD33. In some preferred embodiments, the cytotherapeutic unit comprises cells that lack specific antigenic determinants. In other embodiments, at least one identified potent cell that is derived from a source is specifically excluded or removed from the cellular preparation. [0011]
  • In one embodiment of the invention, some or all cells may be characterized by the presence of one or more of the following cell surface markers: CD10+, CD29+, CD34−, CD38−, CD44+, CD45−, CD54+, CD90+, SH2+, SH3+, SH4+, SSEA3−, SSEA4−, OCT−4+, and ABC−p+. [0012]
  • The potent cells may be obtained from fetal cord blood or other fetal tissue. In some embodiments, potent cells are obtained from placenta, especially postpartum placenta, which has been metabolically supported and nurtured. Potent cells are preferably obtained from postpartum placenta perfusate. The present invention also provides for cytotherapeutic units wherein the potent cells are derived from a plurality of sources. In some embodiments, the potent cells are derived from at least two individuals, at least five individuals, or at least ten individuals. In some embodiments, the unit comprises at least one cell that is autologous. In some other embodiments, the unit comprises at least one cell that is exogenous. In some embodiments the unit comprises a chimera of autologous and allogeneic cells. In another embodiment at least some of the cells are genetically modified. [0013]
  • In other embodiments, the plurality of potent cells is selected to render the unit suitable for therapy for an indicated disease state or condition and/or the severity of the condition. In some preferred embodiments, the cytotherapeutic units comprise a minimum number of preselected types of potent cells and may be based, for example, on the weight of the particular patient or that patient's medical status. In some preferred embodiments, the cytotherapeutic unit is assayed to ensure the accuracy of its contents of preselected types of potent cells. In some preferred embodiments, the contents of the preselected potent cells in the cytotherapeutic unit are certified. In other embodiments, the cytotherapeutic unit can be one of a group of substantially identical units wherein the additional units are stored for future transplants so that, if needed, the patient can receive a unit identical to one previously transplanted. Alternatively, the additional like-units may be altered to optimize future transplants for that same patient. [0014]
  • In other embodiments, at least one type of cell is excluded from the cytotherapeutic unit comprising preselected potent cells. The cytotherapeutic unit is preferably certified as to its contents of the preselected potent cells and the absence of the types of cells to be excluded. In other embodiments, the identity and the numbers of a plurality of potent cells being selected to render the cytotherapeutic unit suitable for therapy for an indicated disease state or condition is certified. In some embodiments, the certification is preferably of a plurality of potent cell types, wherein the plurality and the numbers of each of said plurality being selected as well as excluded renders the cytotherapeutic unit suitable for therapy for an indicated disease state or condition. [0015]
  • In some embodiments, the present invention provides for kits for the treatment of a person suspected of having a disease state or condition. The kit preferably comprises a cytotherapeutic unit comprising a plurality of potent cells. In some embodiments, the kit comprises a cytotherapeutic unit wherein at least one type of cell that has been excluded from the cytotherapeutic unit. In some preferred embodiments, the kit comprises potent cells wherein at least some of the potent cells have been identified and counted. In some embodiments, the kit comprises a unit that has been assayed to ensure the accuracy of the identities and numbers of the potent cells. In some more preferred embodiments of the kit, the accuracy of the assay has been certified. [0016]
  • The present invention provides kits for the treatment of a person suspected of having a disease state or condition comprising a cytotherapeutic unit having minimum numbers of identified potent cells and a certification of the potent cell composition. The kits may also contain equipment or devices for administering the unit to the patient, materials for monitoring the administration and other attendant things. [0017]
  • In some embodiments, the present invention provides for cytotherapeutic units comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, wherein at least one type of cell has been removed from the unit. In some embodiments, a plurality of cell types has been removed from the unit. [0018]
  • The present invention provides for a cytotherapeutic unit comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, wherein said cells comprise a plurality of different types. In some embodiments at least some of the different types of cells are separated into components. In other embodiments, the components are recombined into the unit. It is preferred in some aspects of the invention that components are used to supplement a cytotherapeutic unit with a specific potent cell type. The separated components can be frozen separately or otherwise stored prior to recombination. In some other embodiments, the cytotherapeutic unit itself has been placed in a frozen state. In some further embodiments, the separated cell types have been identified and/or counted. [0019]
  • The present invention provides methods of treating a disease in a mammal comprising administering to the mammal a therapeutically effective amount of a composition comprising a cytotherapeutic unit. The unit used to treat the disease state or condition comprises a plurality of potent cells wherein the content of the unit is known with respect to the identities and numbers. At least some of the cells in the unit are assayed to ensure the accuracy of the identities and the numbers of the potent cells. In some preferred embodiments, the cytotherapeutic unit is administered multiple times. In other cases, administering multiple doses of the cytotherapeutic units that are derived from different individuals or sources may be performed. The methods may also comprise administering multiples doses of the cytotherapeutic unit that is derived from one individual. [0020]
  • The present invention provides for cytotherapeutic units comprising a plurality of potent cells with the content of the cytotherapeutic unit being known with respect to the identities and numbers of at least some of the potent cells. [0021]
  • The identities of the potent cells in the cytotherapeutic unit are an aspect of the invention that is important for the reliability and the quality of the unit being used. The potent cells can be identified by any number of methods and based on any set of criteria that a person of ordinary skill may find useful. One such method is to identify the potent cells based on the presence of antigenic determinants on the surface of the cell. Antigenic determinants can be any molecule that is recognizable by an antibody. Some examples of antigenic determinants include polypeptides, lipids, glycoproteins, sugars, and the like. Additionally, the cells may be characterized by the presence of one or more of the following cell surface markers: CD10+, CD29+, CD34−, CD38−, CD44+, CD45−, CD54+, CD90+, SH2+, SH3+, SH4+, SSEA3−, SSEA4−, OCT−4+, and ABC−p+. [0022]
  • Although some potent cells may be identified by the presence of antigenic determinants or by certain expressed factors, it can be equally important to identify a cell based on what antigenic determinants the cell lacks. For example, it is known that the presence of certain determinants may lower the chances of a successful treatment and therefore, a person using the cytotherapeutic unit would want to know that the unit being used lacks certain antigenic determinants. Furthermore, the presence or absence of antigenic factors can aid in determining the maturity level of a particular cell or cell-type. A less mature cell has a wider range of differentiation and is therefore, potentially more useful. Depending on the use of the cytotherapeutic unit, different levels of differentiation of the cells may be required. The identification of some of the cells enables a person to obtain a unit, that when used, results in a better clinical outcome. [0023]
  • Methods to determine the presence or absence of antigenic factors on or in a cell are well known in the art. These methods include fluorescence activated cell sorting (FACS), Enzyme-Linked Immuno Sorbent Assay (ELISA), western blot, polymerase chain reaction (PCR), reverse-transcribed PCR (RT-PCR), and the like. The precise method or methods used to identify the potent cells is not essential. [0024]
  • Other criteria to identify a cell can be based on the genetic makeup of the cell. Genes play an essential role in everything that occurs in a cell. Because of this fact, a person of ordinary skill in the art may identify a potent cell based on its genes. More specifically, a person of ordinary skill in the art may identify a cell based on the genes that are wild-type, mutant, being expressed, not being expressed, contain polymorphisms, or a combination thereof. As used herein, the term “expressed” means whether or not the gene is being transcribed into RNA or whether a protein is ultimately produced by that gene. [0025]
  • The methods to determine the genetic profile of a cell are well known to those of ordinary skill in the art. Any method used is sufficient, but some examples of methods or techniques that can be used to determine the genetic makeup of a cell include, without limitation, PCR, RT-PCR, northern blot, southern blot, single nucleotide polymorphism (SNP) analysis, gene-chip expression analysis, serial analysis of gene expression (SAGE), nucleotide sequencing, FACS, in situ hybridization, and the like. [0026]
  • In some embodiments of the present invention, a cell can be identified by any of the above-mentioned criteria: antigenic determinants, genetic makeup, a combination thereof, or a cell can be identified based upon another set of criteria. In some embodiments, at least 0.1%, 1%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or about 100% of the cells are identified. [0027]
  • Methods of identification and determining the number of cells are well known in the art, they include but are not limited to using standard cell detection techniques such as flow cytometry, cell sorting, immunocytochemistry (e.g., staining with tissue specific or cell-marker specific antibodies), FACS, magnetic activated cell sorting (MACS), by examination of the morphology of cells using light or confocal microscopy, or by measuring changes in gene expression using techniques well known in the art, such as PCR and gene expression profiling. Additionally, relevant determinations can be made by techniques including, but not limited to, optical and electrooptical properties, morphological imaging methods, optophoresis (www.genoptix.com) microwave spectroscopy (Signature Bioscience www.signaturebio.com) and optical tweezers. Other methods may also be employed. [0028]
  • It is known that specific cell-types or cells having particular antigenic determinants can have a deleterious effect on the success rate of cytotherapy. Therefore, the present invention provides for cytotherapeutic units that have at least one cell type that is excluded. The cell-type that is excluded will not always be the same. In some embodiments, all CD34 positive cells will be excluded. In some other embodiments all CD8 positive cells will be excluded. In some other embodiments multiple cell types are excluded. In some applications, it may be acceptable and convenient to reduce, rather than eliminate, selected cell types to improve therapeutic success. Thus, the term “exclusion” or “elimination” as used in this context preferably means at least about 75% reduction in the number of a certain cell type in a cell preparation. Preferably, at least about 90% reduction is achieved, with at least about 95% reduction being even more preferred. Essentially complete elimination is, of course, most desirable, although the same may be achievable in some cases. The foregoing percentage reductions relate to numbers of cells relative to an original population of such cells using any appropriate assay. [0029]
  • Cell types can be excluded or reduced either by selecting cell-containing units which, naturally do not contain them (or many of them) or by employing a process that specifically removes selected cell-types. It is preferred to exclude cell types having antigenic determinants which are inconsistent with the therapeutic modality planned for the cytotherapeutic unit. For example, but not by way of limitation, T-lymphocytes and mature dendritic cells may be excluded to lower the expectation of graft versus host disease. In the treatment of adrenal leukodysplasia it may be desirable to delete some or all CD8 positive cells. [0030]
  • To be excluded “naturally” means that the preparation of cells that is derived from a source does not contain a specific cell type without further manipulation or contains a very small population of such types. Alternatively, a cell-type can be excluded by a process that is used either before or after the cells are extracted from a source. Processes or methods that are used to exclude a specific cell-type are well known to the art-skilled. Examples of processes or methods include: FACS, centrifugation, immunochromatography, and the like. [0031]
  • In one embodiment, the cells may be sorted using a fluorescence activated cell sorter (FACS). Fluorescence activated cell sorting (FACS) is a well-known method for separating particles, including cells, based on the fluorescent properties of the particles (Kamrach, 1987, Methods Enzymol, 151:150-165). Laser excitation of fluorescent moieties in the individual particles results in a small electrical charge allowing electromagnetic separation of positive and negative particles from a mixture. In one embodiment, cell surface marker-specific antibodies or ligands are labeled with distinct fluorescent labels. Cells are processed through the cell sorter, allowing separation of cells based on their ability to bind to the antibodies used. FACS sorter particles may be directly deposited into individual wells of 96-well or 384-well plates to facilitate separation and cloning. Reagents for cell surface markers or cluster designated reagents are available from a variety of sources including Becton Dickinson and Cell Pro Inc., for example. [0032]
  • Available reagents include but are not limited to reagents for identifying: CD1a; CD2; CD3; CD4; CD4 (Multi-Clone); CD4 v4; CD5; CD7; CD8 (Leu-2a); CD8 (Leu-2b); CD10 (Anti-CALLA); CD11a (Anti-LFA-1α); CD11b; CD11c; CD13; CD14; CD15; CD16 (Leu-11a, 11b, 11c); CD18 (Anti-LIFA-1β); CD19 (Leu-12); CD19(SJ25C1); CD20; CD21(Anti-CR[0033] 2); CD22; CD23; CD25(Anti-IL-2R); CD26; CD27; CD28; CD31(Anti-PECAM-1); CD33; CD34(Anti-HPCA-1&2); CD38; CD42a(Anti-gpIX); CD44; CD45(Anti-Hle-1); CD45RA; CD45RO; CD49d(Anti-VLA-α4); CD54; CD56(MY31); CD56(NCAM16.2); CD57; CD58(Anti-LFA-3); CD61; CD62P; CD62L(Leu-8); CD69; CD71; CD80(Anti-BB1/B7); CD95; CD117; CD122(Anti-IL-2Rp75); CD123(Anti-IL-3Rα); CD134(Ox40); CD154(CD40L); CD158a; CD161; Lineage Cocktail 1 (lin1) FITC and others now known or hereafter discovered.
  • Non-cluster designated reagents include: Anti-BrdU; Anti-Cytokeratin (CAM 5.2); Anti-HER-2/neu; Anti-HLA-DP; Anti-HLA-DQ; Anti-HLA-DR; Anti-Hu KIR (NKB1); Anti-IgA[0034] 2; Anti-IgD; Anti-IgG; Anti-IgM (Ig Heavy Chain); Anti-Kappa (Ig Light Chain); Anti-Kappa F(ab′)2; Anti-Lambda (Ig Light Chain); Anti-Lambda F(ab′)2; Anti-P-glycoprotein (P-gp); Anti-TCR α/β-1 (WT31); Anti-TCR-γ/δ-1; PAC-1; Lineage Cocktail 1 (lin1) FITC. The skilled artisan will use those reagents required for his/her particular needs in order to optimize the desired cytotherapeutic unit or tailor it for a particular patient or use.
  • In another embodiment, magnetic beads can be used to separate cells. The cells may be sorted using a magnetic activated cell sorting (MACS) technique, a method for separating particles based on their ability to bind magnetic beads (0.5-100 μm diameter). A variety of useful modifications can be performed on the magnetic microspheres, including the covalent addition of an antibody which specifically recognizes a cell-solid phase surface molecule or hapten. A magnetic field is then applied, to physically manipulate the selected beads. The beads are then mixed with the cells to allow binding. Cells are then passed through a magnetic field to separate out cells having cell surface markers. These cells can then isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers. The cells are again passed through a magnetic field, isolating cells that bound both the antibodies. Such cells can then be diluted into separate dishes, such as microtiter dishes for clonal isolation, if desired. [0035]
  • Knowing the composition of the cytotherapeutic unit will help fulfill the long-felt need of a reliable and certified cytotherapeutic unit. In addition to the composition of the unit, it can be useful to know the numbers of at least some of the cells in the cytotherapeutic unit. In some embodiments, just the numbers of cells will be known without knowing the specific identity of any of the cells. In some other embodiments, the numbers of cells will be known, but also the numbers of the identified cells will be known. To determine the number of cells in total is well known to those of ordinary skill in the art. Examples of equipment that can be used to count cells are a machine that performs FACS or flow cytometry, or a much simpler piece of equipment, a hemacytometer. Often the number of the cells will be determined at the same time the identities are determined, but the numbers can also be determined before or after the identities of some of the potent cells are determined. By knowing the number of the cells present in a cytotherapeutic unit this will give a person using the unit the knowledge of what is being administered, something that is sorely lacking in present cytotherapies. [0036]
  • The knowledge of the numbers of total cells and also the numbers of specific cell types in a cytotherapeutic unit can be used to supplement the unit with additional cells or cell types so that a minimum number of cells or a minimum number of a specific cell type can be present in the unit. It is thought that the diverse responses seen in cytotherapy is in part due to the varying number of cells recovered from a source using the cellular preparation techniques in use today. [0037]
  • By identifying and counting the cells this will allow a more thorough analysis of what is required for a successful treatment as well as the ability to perform a thorough and complete analysis on the importance of a specific cell type in a cellular preparation. [0038]
  • Cytotherapeutic units can now be prepared that have a minimum numbers of preselected cells. It is also now possible to ensure that other cell types are excluded from the-units. In some embodiments the cytotherapeutic unit will comprise at least about 100 selected potent cells. Such units having at least about 1,000 such cells are preferred, with at least about 10,000 being more preferred. Greater numbers of selected cells are still more preferred, especially when it is intended that the unit be administered to the same or different individuals a plurality of times. Thus, selected cell populations greater than about 100,000 or even about 500,000 can be useful. It is preferred that some or all of the cells in the unit be identified through assay and that the same be reflected in a certification of such presence. This certification ensures uniform and effective therapeutic application. [0039]
  • In some embodiments of the present invention, the cytotherapeutic units will have a minimum number of different, specific cell types. Advantages to having a minimum number of specific cell types are that it may improve the efficacy of the cytotherapeutic unit. For example, the cytotherapeutic unit could be assayed to comprise at least about 1,000 OCT4 positive cells, either with or without known quantities of other desirable cell types. In other embodiments, the unit may be caused to comprise specific percentages of CD34 positive cells, measured by reference to all nucleated cells in the preparation. Thus, such preparations may contain at least 0.01%, 0.1%, 1%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or other percentages of CD34 positive cells may be made. Similar, known percentages of cells having other antigenic determinants or specific factors may, likewise, be created. [0040]
  • Other embodiments of the present invention provide for cytotherapeutic units comprising cells that have been derived from at least one source, wherein the source's cells have been separated into components. As used herein, the term “components” is synonymous to cell-types, identified cells, and the like. Methods to separate cellular preparations into components that are well known to those of skill in the art include, without limitation, FACS, centrifugation, chromatography, HPLC, FPLC, and the like. [0041]
  • Thus, cytotherapeutic units can comprise components that are recombined. In some embodiments, at least one component is used in a cytotherapeutic unit. In some other embodiments, at least two, at least three, at least 4, at least 5, at least 10, at least 100 components are recombined to make the cytotherapeutic unit. It is preferred that the components of each source be known in terms of identity and relative numbers, with some cell types preferably being excluded from some or all of the components. It may be seen that the different components may be maintained separately,; e.g. frozen, and that the same may form a “formulary” or “library” of cells of known identity and abundance for formulation into combined cytotherapeutic units. Separating the respective cellular preparations into components allows a cytotherapeutic unit to be created that has a specific composition both in terms of cells present and in types of cells excluded. Additionally, this allows an existing cytotherapeutic unit to be supplemented with a specific cell-type or component as may be indicated for a specific therapeutic modality. [0042]
  • Thus, cytotherapeutic units of the invention may be seen to comprise cells derived from one source or from many sources. Contrary to prevailing practice, it is believed that there are great benefits to providing cells from a plurality of sources and that therapeutic benefit and efficacy will derive therefrom. In some embodiments, the cells are derived from multiple sources and may derive from multiple organs in such sources. As used herein, the term “source” refers to any organism, tissue, or organ from which cells are derived or extracted. In some embodiments, the sources are fetal cord blood, fetal tissue, placenta, postpartum placenta, postpartum placenta perfusate, or a mixture thereof. It is well known to those of ordinary skill how to extract cells from different tissues or organs. Methods to extract cells from fetal cord blood can be found in, for example in U.S. Pat. No. 5,372,581, entitled “Method and apparatus for placental blood collection,” issued Dec. 13, 1994; Hessel et al., U.S. Pat. No. 5,415,665, entitled “Umbilical cord clamping, cutting, and blood collecting device and method”, issued May 16, 1995. The needle or cannula is usually placed in the umbilical vein and the placenta is gently massaged to aid in draining cord blood from the placenta. Methods to extract cells from placenta, post-partum placenta, or post-partum placenta perfusate can be found in, for example, International Patent Publications WO 02/46373 and WO 02/064755, each of which are herein incorporated by reference in their entireties. [0043]
  • In another embodiment, the cells are stimulated to proliferate, for example, by administration of erythropoietin, cytokines, lymphokines, interferons, colony stimulating factors (CSF's), interferons, chemokines, interleukins, recombinant human hematopoietic growth factors including ligands, stem cell factors, thrombopoeitin (TPO), interleukins, and granulocyte colony-stimulating factor (G-CSF) or other growth factors. [0044]
  • In another embodiment, cells are genetically engineered, for example, using a viral vector such as an adenoviral or retroviral vector, or by using mechanical means such as liposomal or chemical mediated uptake of the DNA. [0045]
  • A vector containing a transgene can be introduced into a cell of interest by methods well known in the art, e.g., transfection, transformation, transduction, electroporation, infection, microinjection, cell fusion, DEAE extran, calcium phosphate precipitation, liposomes, LIPOFECTIN™, lysosome fusion, synthetic cationic lipids, use of a gene gun or a DNA vector transporter, such that the transgene is transmitted to daughter cells, e.g., the daughter embryonic-like stem cells or progenitor cells produced by the division of an embryonic-like stem cell. For various techniques for transformation or transfection of mammalian cells, see Keown et al., 1990, Methods Enzymol. 185:527-37; Sambrook et al., 2001, Molecular Cloning, A Laboratory Manual, Third Edition, Cold Spring Harbor Laboratory Press, N.Y. [0046]
  • The cytotherapeutic units will preferably comprise minimum numbers of preselected types of potent cells and be certified as such. As used herein, “preselected” refers to the process of selecting the types of potent cells that are to be in the cytotherapeutic unit before it is administered. Preselecting the types of potent cells that will have a minimum number of those cells in the cytotherapeutic unit allows the cytotherapeutic unit to be tailored to a composition desired to achieve a specific therapeutic result in an individual or class of individuals. Likewise, certification as to the absence of other preselected types of cells is preferred for similar reasons. [0047]
  • The plurality of potent cells and of cell types present in the cytotherapeutic units of the invention are selected to render the units suitable for therapy for an indicated disease state or condition. As used herein, the phrase “selected to render” refers to the process of deciding that a cytotherapeutic unit comprising a plurality of potent cells is suitable for therapy. This decision can be based on the numbers of potent cells present in the cytotherapeutic unit. As discussed hereinbefore, the number of cells appears to be critical for the success rate of treating an individual or patient with cytotherapy. Therefore, not all cytotherapeutic units may be suitable for therapy for an indicated disease state or condition. Additionally, the types of potent cells will also aid in the decision process on whether or not a cytotherapeutic unit is suitable for therapy. Certain types of potent cells can be detrimental or beneficial to the treatment of a specific disease state or condition. Thus, the types of cells present in the unit can be another factor that is used to select a unit suitable for therapy. The criteria that are used to select a unit that is suitable for therapy is not specific to those mentioned above. Any set of criteria can be used to decide whether or not a plurality of potent cells present in a cytotherapeutic unit are selected to render the unit suitable for therapy of an indicated disease state or condition. [0048]
  • The present invention provides for cytotherapeutic units wherein at least some of the potent cells present in the unit are identified and counted. However, for the units to be relied upon in scientific research and to be used as a cytotherapeutic the units' contents must be preferably assayed to ensure the accuracy of the identities and numbers. The assays can be done by the same group, individual, or machine that had determined the identities and the numbers of at least some of the potent cells in the cytotherapeutic units. However, the assays can be performed by a different individual, group, or machine that had determined the identities and numbers of some of the potent cells. In some embodiments, only one assay needs to be performed to ensure the accuracy of the identities and the numbers. In some other embodiments, at least 2, at least 5, or at least 10 assays are performed to ensure the accuracy of the identities and the numbers of the potent cells. The types of assays to be done can be the same assay that was used to determine the numbers and the identities previously. In some other embodiments, different assays are used to ensure the accuracy of the numbers and identities of some of the potent cells. Some assays that can be used to ensure the accuracy include, without limitation, ELISA, FACS, western blot, and the like. [0049]
  • In some other embodiments, the provider of the unit certifies the accuracy of the assay. As used herein, the term “provider” refers to an individual, business, or facility that is providing the cytotherapeutic unit to the individual that is using the unit. In some embodiments, the certification comprises a written statement indicating that the assay was performed correctly and that the results are correct. In some other embodiments, the certification comprises results from an assay done on a positive control to show that the assay was functioning properly. In some other embodiments, the certification comprises both the results of the positive control and a written statement that the assay was functioning properly. In some further embodiments, the certification comprises a list of the types of potent cells that have been excluded from the cytotherapeutic unit. In some further embodiments, the certification comprises a list of at least some of the types of potent cells that are contained in the cytotherapeutic unit. In some embodiments, the certification comprises the numbers of all the cells. In some embodiments, the certification further comprises the quantity of at least some of the specific cell types. In some other embodiments, the certification comprises a list of the types of at least some of the potent cells that have been added to the unit to supplement the potent cells so that the unit comprises minimum numbers of potent cells. [0050]
  • The present invention also provides for kits for the treatment of a person suspected of having a disease state or condition comprising a cytotherapeutic unit comprising a plurality of potent cells with the content of the unit being known with respect to the identities and numbers of at least some of the potent cells. Additionally, the cytotherapeutic unit is assayed to ensure the accuracy of the identities and numbers of the potent cells. The kits further comprise a certification of the accuracy of the assay. In some embodiments, the kits comprise a cytotherapeutic unit having minimum numbers of identified potent cells and a certification of the potent cell composition of the unit. In some other embodiments, the kits comprise cytotherapeutic units that have at least one cell-type that has been excluded. [0051]
  • The present invention also provides for methods of treating a disease state or condition in a mammal. The methods comprise administering to the mammal a therapeutically effective amount of a composition comprising a cytotherapeutic unit comprising potent cells, wherein some of the potent cells are known with respect to their identities and numbers. The unit is also assayed to ensure the accuracy of the identities and the numbers. In some other embodiments, the cytotherapeutic unit comprises minimum numbers of preselected types of potent cells. [0052]
  • A therapeutically effective amount for a mammal can vary, but for example could be approximately 0.01 cytotherapeutic units/kg to 100 units/kg. The cytotherapeutic unit can be administered rapidly or slowly to the mammal. In some embodiments, the cytotherapeutic unit is administered at a rate of approximately 0.01 μl/minute, and in other embodiments, the unit is administered at a rate of approximately 100,000 ml/minute. The unit can be administered, for example, intravenously, subcutaneously, intramuscularly, orally, or rectally. In some embodiments, the unit is administered multiple times to the mammal at different times. In some other embodiments, cytotherapeutic units derived from different sources or different individuals are administered to the mammal. [0053]
  • The potential uses for cytotherapeutic units are limitless, but some examples of disease states or conditions that cytotherapeutic units can be used to treat include cancer, acute leukemia, chronic leukemia as well as other cancers presently treated with bone marrow or cord blood cell transplants, myelodysplastic syndrome, stem cell disorder, myeloproliferative disorder, lymphoproliferative disorder, phagocyte disorder, liposomal storage disorder, histiocytic disorder, inherited erythrocyte abnormality, congenital (inherited) immune system disorder, inherited platelet abnormality, plasma cell disorder, Lesch-Nyhan Syndrome, Cartilage-Hair Hypoplasia, Glanzmann Thrombastenia, osteoporosis, breast cancer, Ewing Sarcoma, neuroblastoma, renal cell carcinoma, lung cancer, Alzheimer's disease, liver disease, hepatitis, Parkinson's disease, vision loss, memory loss, and the like. [0054]
  • The cytotherapeutic units may be optimized for enzyme replacement therapy to treat specific diseases or conditions, including, but not limited to, lysosomal storage diseases, such as Tay-Sachs, Niemann-Pick, Fabry's, Gaucher's, Hunter's, and Hurler's syndromes, as well as other gangliosidoses, mucopolysaccharidoses, and glycogenoses. The cytotherapeutic units in this case may be certified that the cells have been assayed to contain the desired number of cells capable of producing the necessary enzyme. Said unit may contain either allogeneic cells containing the functional endogenous gene of the desired enzyme, autologous cells containing exogenous copies of the desired gene or a combination of both. [0055]
  • In other embodiments, the cells may be used as autologous or heterologous transgene carriers in gene therapy to correct inborn errors of metabolism such as adrenoleukodystrophy, cystic fibrosis, glycogen storage disease, hypothyroidism, sickle cell anemia, Pearson syndrome, Pompe's disease, phenylketonuria (PKU), Tay-Sachs disease, porphyrias, maple syrup urine disease, homocystinuria, mucopolypsaccharidoses, chronic granulomatous disease, and tyrosinemia or to treat cancer, tumors or other pathological conditions. [0056]
  • The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that present invention is not entitled to antedate such publication by virtue of prior invention. [0057]
  • The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims. [0058]
  • All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.[0059]
  • EXAMPLE 1
  • An adult having acute myelogenous leukemia (AML) is in need of hematopoetic reconstitution by way of a cell transplant. The patient undergoes traditional chemotherapy followed by conventional preparation for transplant as determined by the patient's health care provider but includes destroying the diseased bone marrow. The patient's weight is determined. Appropriate HLA typing has been done by conventional methods. Based on these parameters, which include the disease to be treated, the patient's body weight and HLA matching, the transplanter requests and is provided with a cytotherapeutic unit comprising a plurality of potent nucleated cells; the content of said unit being known with respect to the identities and numbers at least some of said plurality; the unit being assayed to ensure the accuracy of said identities and numbers, which is certified. In particular, the unit is certified to contain about 1.4×10[0060] 7 nucleated cells per kilogram of the patient's body weight. Additional certified information includes HLA information. Because the patient suffers from AML, the cytotherapeutic unit contains no less than one (1) percent of CD34+ of the total nucleated cells and no less than 2.5 percent CD8+ cells to minimize graft versus tumor effect. In this case the transplanter requests twice the total number of cells needed for transplant (1.4×107 nucleated cells multiplied by the patient's weight in kilograms ×2). The transplanter requests the 1× amount just prior to the transplant in order to have the number of cells suitable for this transplant. The second half of the cells is to be shipped in the event that a second transplant becomes necessary. Accordingly, the second cytotherapeutic unit is the same as that to be used in the initial transplant. Alternatively, the transplanter may request, based on alterations in the patient's weight, severity of disease or even changes in recommended treatment, that the second cytotherapeutic unit be altered in the appropriate manner (increased number of CD34 positive cells, etc.) and certified. The transplant is performed in the same manner conventionally used by the transplanter.
  • EXAMPLE 2
  • A child having sickle cell anemia is in need of a cell transplant. It is determined that 1.7×10[0061] 7 nucleated cells per kilogram of body weight of the child is needed. Appropriate HLA typing is done by conventional methods. It is determined that the cytotherapeutic unit must have no less than 1% CD34+cells of the total nucleated cells. Said CD34+ cells are further described in a ratio of 2:1 as CD34+/CD33+: CD34+/CD33 A cytotherapeutic unit having these parameters is provided. This unit comprises cells derived from cord blood as well as pluripotential placental cells such as those described in WO 02/064755, which are derived in the manner described in WO02/064755. The ratio of CD34+/CD33+ cells is 2:1 to CD34+/CD33−, a fact which is ascertained by assay and certified as being accurate. The certified cells are determined using FACS; based on the fluorescent properties of the particles, cell surface marker-specific antibodies or ligands are labeled with distinct fluorescent labels. Cells are processed through the cell sorter, allowing separation of cells based on their ability to bind to the antibodies used. Cell surface marker-specific antibodies may be purchased from any company selling such reagents, including Becton Dickinson, for example. The transplant is performed in the same manner conventionally used by the transplanter.
  • EXAMPLE 3
  • A child suffers from adrenal leukodysplasia. It is determined that a cellular transplant is appropriate. It is determined that 2×10[0062] 7 nucleated cells (derived from cord blood by a conventional technique) per kilogram of body weight of the child is needed. Appropriate HLA typing is done by conventional methods. A cytotherapeutic unit having these parameters is provided. In particular, the unit is certified to contain no less than 0.25% of CD34+/CD38− cells and with no less than 0.5% depletion of CD8+ cells of the total nucleated cells. The transplant is performed in the same manner conventionally used by the transplanter.

Claims (53)

1. A cytotherapeutic unit comprising a plurality of potent cells; the content of said unit being known with respect to the identities and numbers at least some of said plurality; the unit being assayed to ensure the accuracy of said identities and numbers.
2. The cytotherapeutic unit of claim 1 wherein the accuracy of the assay is certified by the provider of the unit.
3. The cytotherapeutic unit of claim 1 wherein the potent cells for which the identities and numbers are known are pluripotent cells.
4. The cytotherapeutic unit of claim 1 wherein said identities reflect the presence or absence of at least one antigenic determinant on identified cells.
5. The cytotherapeutic unit of claim 1 wherein said potent cells are obtained from fetal cord blood or other fetal tissue.
6. The cytotherapeutic unit of claim 1 wherein said potent cells are obtained from fetal cord blood.
7. The cytotherapeutic unit of claim 1 wherein said potent cells are obtained from placenta.
8. The cytotherapeutic unit of claim 1 wherein said potent cells are obtained from postpartum placenta.
9. The cytotherapeutic unit of claim 1 wherein said potent cells are obtained from postpartum placenta perfusate.
10. The cytotherapeutic unit of claim 1 wherein potent cells for which the identities and numbers are known comprise at least some of cells exhibiting CD34, CD8, CD10, OCT4.
11. The cytotherapeutic unit of claim 1 wherein potent cells are derived from a plurality of sources.
12. The cytotherapeutic unit of claim 1 wherein potent cells are derived from at least two individuals.
13. The cytotherapeutic unit of claim 1 wherein potent cells are derived from at least 5 individuals
14. The cytotherapeutic unit of claim 1 wherein potent cells are genetically modified.
15. The cytotherapeutic unit of claim 1 wherein at least one type of cell is excluded from the unit.
16. The cytotherapeutic unit of claim 1 wherein the plurality of potent cells is selected to render the cytotherapeutic unit suitable for therapy for an indicated disease state or condition.
17. The cytotherapeutic unit of claim 16 wherein at least one type of cell is excluded from the unit.
18. A cytotherapeutic unit comprising minimum numbers of preselected types of potent cells.
19. The cytotherapeutic unit of claim 18 which has been assayed to ensure accuracy of its contents of preselected types of potent cells.
20. The cytotherapeutic unit of claim 18 wherein the contents of preselected potent cells is certified.
21. The cytotherapeutic unit of claim 18 wherein at least one type of cell is excluded from the unit.
22. The cytotherapeutic unit of claim 21 wherein the contents of preselected potent cells and the absence the types of cells to be excluded is certified.
23. The cytotherapeutic unit of claim 18, wherein said certification is of a plurality of potent cell types, said plurality and the numbers of each of said plurality being selected to render the cytotherapeutic unit suitable for therapy for an indicated disease state or condition.
24. The cytotherapeutic unit of claim 23, wherein said certification is of a plurality of potent cell types, said plurality and the numbers of each of said plurality being selected as well as the types of cells excluded renders the cytotherapeutic unit suitable for therapy for an indicated disease state or condition.
25. The cytotherapeutic unit of claim 18 where at least some potent cells are genetically modified.
26. A kit for treatment of a person suspected of having a disease state or condition comprising a cytotherapeutic unit comprising a plurality of potent cells; the content of said unit being known with respect to the identities and numbers at least some of said plurality; the unit being assayed to ensure the accuracy of said identities and numbers; and a certification of the accuracy of the assay.
27. The kit of claim 26 wherein at least one type of cell has been excluded from the cytotherapeutic unit.
28. A kit for treatment of a person suspected of having a disease state or condition comprising a cytotherapeutic unit having minimum numbers of identified potent cells and a certification of the potent cell composition of the unit.
29. The kit of claim 28 wherein at least one type of cell has been excluded from the cytotherapeutic unit.
30. The kit of claim 28 wherein at least some cells are genetically modified.
31. A cytotherapeutic unit comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, wherein at least one type of cell has been removed from the unit.
32. The cytotherapeutic unit of claim 31 wherein a plurality of cell types have been removed from the unit.
33. The cytotherapeutic unit of claim 31 wherein at least some cells of the unit are genetically modified.
34. A cytotherapeutic unit comprising cells derived from umbilical cord blood, placenta, or a mixture thereof, said cells comprising a plurality of different types, at least some of the different types having been separated into components and recombined into said unit.
35. The cytotherapeutic unit of claim 34, wherein said separated cell types have been frozen separately.
36. The cytotherapeutic unit of claim 34, in a frozen state.
37. The cytotherapeutic unit of claim 34, wherein said separated cell types have been characterized.
38. The cytotherapeutic unit of claim 34, wherein said separated cell types have been genetically modified
39. A method of treating a disease in a mammal comprising: Administering to said mammal a therapeutically effective amount of a composition comprising: a cytotherapeutic unit comprising a plurality of potent cells; the content of said unit being known with respect to the identities and numbers of at least some of said plurality; the unit being assayed to ensure the accuracy of said identities and numbers.
40. The method of claim 39, wherein the cytotherapeutic unit comprises minimum numbers of preselected types of potent cells.
41. The method of claim 39, wherein the accuracy of said identities and numbers is certified.
42. The method of claim 39, wherein the cytotherapeutic unit is derived from post-partum placenta.
43. The method of claim 39, wherein the cytotherapeutic unit is derived from post-partum placenta perfusate.
44. The method of claim 39, wherein said unit comprises at least one cell that is autologous.
45. The method of claim 39, wherein said unit comprises at least one cell that is exogenous.
46. The method of claim 39, wherein said unit is administered multiple times.
47. The method of claim 39, wherein said method further comprises administering multiples of said units that are derived from different individuals.
48. The method of claim 39, wherein said method further comprises administering multiples of said units that are derived from different sources.
49. The method of claim 39, wherein said method further comprises administering multiple units that are genetically modified.
50. A library of cytotherapeutic units, each unit member of said library comprising a plurality of potent cells; the content of each of said units being known with respect to the identities and numbers at least some of the plurality of potent cells comprising said unit; each of said units being assayed to ensure the accuracy of said identities and numbers.
51. A method of treatment of a patient in need of cytotherapeutic treatment comprising selecting from a library of cytotherapeutic units at least two unit members of said library; combining aliquots from said unit members to form a treatment unit and administering said treatment unit to the patient.
52. The method of claim 51 wherein each of the unit members of the library has been assayed to determine the identity and numbers of potent cells present in said unit member.
53. The method of claim 51 wherein at least one of said unit members of the library has been reduced in at least one selected cell population.
US10/721,144 2002-11-26 2003-11-25 Cytotherapeutics, cytotherapeutic units and methods for treatments using them Abandoned US20040171147A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/721,144 US20040171147A1 (en) 2002-11-26 2003-11-25 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US11/592,544 US8617535B2 (en) 2002-11-26 2006-11-03 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US15/362,300 US20170290862A1 (en) 2002-11-26 2016-11-28 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US16/532,080 US20200009199A1 (en) 2002-11-26 2019-08-05 Cytotherapeutics, Cytotherapeutic Units and Methods for Treatments Using Them

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42970202P 2002-11-26 2002-11-26
US10/721,144 US20040171147A1 (en) 2002-11-26 2003-11-25 Cytotherapeutics, cytotherapeutic units and methods for treatments using them

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/592,544 Continuation US8617535B2 (en) 2002-11-26 2006-11-03 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US15/362,300 Division US20170290862A1 (en) 2002-11-26 2016-11-28 Cytotherapeutics, cytotherapeutic units and methods for treatments using them

Publications (1)

Publication Number Publication Date
US20040171147A1 true US20040171147A1 (en) 2004-09-02

Family

ID=32393577

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/721,144 Abandoned US20040171147A1 (en) 2002-11-26 2003-11-25 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US11/592,544 Active 2027-10-08 US8617535B2 (en) 2002-11-26 2006-11-03 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US15/362,300 Abandoned US20170290862A1 (en) 2002-11-26 2016-11-28 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US16/532,080 Abandoned US20200009199A1 (en) 2002-11-26 2019-08-05 Cytotherapeutics, Cytotherapeutic Units and Methods for Treatments Using Them

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/592,544 Active 2027-10-08 US8617535B2 (en) 2002-11-26 2006-11-03 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US15/362,300 Abandoned US20170290862A1 (en) 2002-11-26 2016-11-28 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US16/532,080 Abandoned US20200009199A1 (en) 2002-11-26 2019-08-05 Cytotherapeutics, Cytotherapeutic Units and Methods for Treatments Using Them

Country Status (11)

Country Link
US (4) US20040171147A1 (en)
EP (1) EP1571910A4 (en)
JP (2) JP2006509770A (en)
KR (3) KR101042448B1 (en)
CN (1) CN1717177A (en)
AU (2) AU2003298775B2 (en)
BR (1) BR0316695A (en)
CA (1) CA2505534A1 (en)
MX (1) MXPA05005673A (en)
WO (1) WO2004047770A2 (en)
ZA (1) ZA200504273B (en)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020160510A1 (en) * 2001-02-14 2002-10-31 Hariri Robert J. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US20030235909A1 (en) * 2002-04-12 2003-12-25 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20050019908A1 (en) * 2000-12-06 2005-01-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20050118715A1 (en) * 2002-04-12 2005-06-02 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20050148034A1 (en) * 2002-04-12 2005-07-07 Hariri Robert J. Methods for identification of modulators of angiogenesis, compounds discovered thereby, and methods of treatment using the compounds
US20050186218A1 (en) * 2001-04-13 2005-08-25 Wyeth Holdings Corporation Removal of bacterial endotoxin in a protein solution by immobilized metal affinity chromatography
US20050276792A1 (en) * 2004-03-26 2005-12-15 Kaminski Joseph K Systems and methods for providing a stem cell bank
US20060040392A1 (en) * 2004-04-23 2006-02-23 Collins Daniel P Multi-lineage progenitor cells
US20060147431A1 (en) * 2005-01-04 2006-07-06 Nobuko Uchida Methods for the treatment of lysosomal storage disorders
US20070134210A1 (en) * 2005-10-13 2007-06-14 Mohammad Heidaran Production of oligodendrocytes from placenta-derived stem cells
US20070190042A1 (en) * 2005-12-29 2007-08-16 Edinger James W Composition for collecting and preserving placental stem cells and methods of using the composition
US20070249047A1 (en) * 2006-04-17 2007-10-25 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Respiratory Epithelial Cells
US20090053805A1 (en) * 2000-12-06 2009-02-26 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US7670596B2 (en) 2004-04-23 2010-03-02 Bioe, Inc. Multi-lineage progenitor cells
US7682803B2 (en) 2005-10-13 2010-03-23 Anthrogenesis Corporation Immunomodulation using placental stem cells
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US20100104539A1 (en) * 2007-09-07 2010-04-29 John Daniel Placental tissue grafts and improved methods of preparing and using the same
US7790458B2 (en) 2004-05-14 2010-09-07 Becton, Dickinson And Company Material and methods for the growth of hematopoietic stem cells
US20110020293A1 (en) * 2009-07-21 2011-01-27 Abt Holding Company Use of Stem Cells to Reduce Leukocyte Extravasation
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US20110212069A1 (en) * 2010-02-25 2011-09-01 Abt Holding Company Modulation of Microglia Activation
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US8057789B2 (en) 2002-02-13 2011-11-15 Anthrogenesis Corporation Placental stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8562972B2 (en) 2006-10-23 2013-10-22 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US8586360B2 (en) 2009-07-02 2013-11-19 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
US8617535B2 (en) 2002-11-26 2013-12-31 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US10494607B2 (en) 2007-02-12 2019-12-03 Celularity, Inc. CD34+,CD45−placental stem cell-enriched cell populations

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
JP2009539378A (en) * 2006-06-09 2009-11-19 アントフロゲネシス コーポレーション Placental environment and its use for culturing stem cells
CA2688504A1 (en) 2007-06-18 2008-12-24 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
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
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
EP2623978A1 (en) * 2012-02-03 2013-08-07 Charité - Universitätsmedizin Berlin CD8+ T-cell subsets as markers for prediction of delayed fracture healing

Citations (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3862002A (en) * 1962-05-08 1975-01-21 Sanfar Lab Inc Production of physiologically active placental substances
US4008719A (en) * 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4798824A (en) * 1985-10-03 1989-01-17 Wisconsin Alumni Research Foundation Perfusate for the preservation of organs
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US4999291A (en) * 1985-08-23 1991-03-12 Amgen Inc. Production of human pluripotent granulocyte colony-stimulating factor
US5192312A (en) * 1991-03-05 1993-03-09 Colorado State University Research Foundation Treated tissue for implantation and methods of treatment and use
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
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
US5385901A (en) * 1991-02-14 1995-01-31 The Rockefeller University Method of treating abnormal concentrations of TNF α
US5391485A (en) * 1985-08-06 1995-02-21 Immunex Corporation DNAs encoding analog GM-CSF molecules displaying resistance to proteases which cleave at adjacent dibasic residues
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5487992A (en) * 1989-08-22 1996-01-30 University Of Utah Research Foundation Cells and non-human organisms containing predetermined genomic modifications and positive-negative selection methods and vectors for making same
US5591767A (en) * 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5591625A (en) * 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US5599705A (en) * 1993-11-16 1997-02-04 Cameron; Robert B. In vitro method for producing differentiated universally compatible mature human blood cells
US5605822A (en) * 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5605914A (en) * 1993-07-02 1997-02-25 Celgene Corporation Imides
US5709854A (en) * 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
US5716827A (en) * 1990-03-30 1998-02-10 Systemix, Inc. Human hematopoietic stem cell
US5716794A (en) * 1996-03-29 1998-02-10 Xybernaut Corporation Celiac antigen
US5716616A (en) * 1995-03-28 1998-02-10 Thomas Jefferson University Isolated stromal cells for treating diseases, disorders or conditions characterized by bone defects
US5733541A (en) * 1995-04-21 1998-03-31 The Regent Of The University Of Michigan Hematopoietic cells: compositions and methods
US5733542A (en) * 1990-11-16 1998-03-31 Haynesworth; Stephen E. Enhancing bone marrow engraftment using MSCS
US5733566A (en) * 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5855619A (en) * 1994-06-06 1999-01-05 Case Western Reserve University Biomatrix for soft tissue regeneration
US5858782A (en) * 1995-11-13 1999-01-12 Regents Of The University Of Michigan Functional human hematopoietic cells
US5861315A (en) * 1994-11-16 1999-01-19 Amgen Inc. Use of stem cell factor and soluble interleukin-6 receptor for the ex vivo expansion of hematopoietic multipotential cells
US5866414A (en) * 1995-02-10 1999-02-02 Badylak; Stephen F. Submucosa gel as a growth substrate for cells
US5874448A (en) * 1997-11-18 1999-02-23 Celgene Corporation Substituted 2-(2,6 dioxo-3-fluoropiperidin-3-yl)-isoindolines and method of reducing TNFα levels
US5874301A (en) * 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5877299A (en) * 1995-06-16 1999-03-02 Stemcell Technologies Inc. Methods for preparing enriched human hematopoietic cell preparations
US5877200A (en) * 1993-07-02 1999-03-02 Celgene Corporation Cyclic amides
US5879940A (en) * 1994-07-20 1999-03-09 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoieses
US5879318A (en) * 1997-08-18 1999-03-09 Npbi International B.V. Method of and closed system for collecting and processing umbilical cord blood
US6011000A (en) * 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US6010696A (en) * 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US6020469A (en) * 1993-12-22 2000-02-01 Amgen Inc. Stem cell factor formulations and methods
US6022848A (en) * 1993-03-31 2000-02-08 Pro-Neuron, Inc. Inhibitor of stem cell proliferation and uses thereof
US6022540A (en) * 1997-09-04 2000-02-08 Osiris Therapeutics, Inc. Ligands that modulate differentiation of mesenchymal stem cells
US6022743A (en) * 1986-04-18 2000-02-08 Advanced Tissue Sciences, Inc. Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
US6030836A (en) * 1998-06-08 2000-02-29 Osiris Therapeutics, Inc. Vitro maintenance of hematopoietic stem cells
US6174333B1 (en) * 1994-06-06 2001-01-16 Osiris Therapeutics, Inc. Biomatrix for soft tissue regeneration using mesenchymal stem cells
US6179819B1 (en) * 1996-08-30 2001-01-30 John N. Haswell Umbilical cord blood collection
US6184035B1 (en) * 1998-11-18 2001-02-06 California Institute Of Technology Methods for isolation and activation of, and control of differentiation from, skeletal muscle stem or progenitor cells
US6190368B1 (en) * 1996-05-14 2001-02-20 Children's Hospital Medical Center Of Northern California Apparatus and method for collecting blood from an umbilical cord
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6335349B1 (en) * 1996-07-24 2002-01-01 Celgene Corporation Substituted 2(2,6-dioxopiperidin-3-yl)isoindolines
US6337387B1 (en) * 1995-11-17 2002-01-08 Asahi Kasei Kabushiki Kaisha Differentiation-suppressive polypeptide
US6338942B2 (en) * 1995-05-19 2002-01-15 T. Breeders, Inc. Selective expansion of target cell populations
US20020028510A1 (en) * 2000-03-09 2002-03-07 Paul Sanberg Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US6355239B1 (en) * 1998-03-13 2002-03-12 Osiris Therapeutics, Inc. Uses for non-autologous mesenchymal stem cells
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US20030003573A1 (en) * 2000-04-27 2003-01-02 Lakshmi Rambhatle Hepatocytes for therapy and drug screening made from embryonic stem cells
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US20030032179A1 (en) * 2000-12-06 2003-02-13 Hariri Robert J. Post-partum mammalian placenta, its use and placental stem cells therefrom
US6528245B2 (en) * 1998-05-07 2003-03-04 University Of South Florida Bone marrow cells as a source of neurons for brain and spinal cord repair
US20030044976A1 (en) * 2001-08-27 2003-03-06 Advanced Cell Technology De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20030044977A1 (en) * 2001-08-10 2003-03-06 Norio Sakuragawa Human stem cells originated from human amniotic mesenchymal cell layer
US20030045552A1 (en) * 2000-12-27 2003-03-06 Robarge Michael J. Isoindole-imide compounds, compositions, and uses thereof
US6534084B1 (en) * 1999-06-30 2003-03-18 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6538023B1 (en) * 2000-09-15 2003-03-25 Tsuyoshi Ohnishi Therapeutic uses of green tea polyphenols for sickle cell disease
US20030059414A1 (en) * 2001-09-21 2003-03-27 Ho Tony W. Cell populations which co-express CD49c and CD90
US20030235563A1 (en) * 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20040018817A1 (en) * 2002-07-18 2004-01-29 Omron Corporation Communication system, communication apparatus, and communication control method
US20040018617A1 (en) * 2002-07-26 2004-01-29 Shiaw-Min Hwang Somatic pluripotent cells
US6685936B2 (en) * 1999-10-12 2004-02-03 Osiris Therapeutics, Inc. Suppressor cells induced by culture with mesenchymal stem cells for treatment of immune responses in transplantation
US20040028660A1 (en) * 2002-05-30 2004-02-12 Anthrogenesis Corporation Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US20040048796A1 (en) * 2002-03-26 2004-03-11 Hariri Robert J. Collagen biofabric and methods of preparation and use therefor
US6709864B1 (en) * 1996-07-30 2004-03-23 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
US20040058412A1 (en) * 2002-09-20 2004-03-25 Neuronyx, Inc. Cell populations which co-express CD49c and CD90
US20050009876A1 (en) * 2000-07-31 2005-01-13 Bhagwat Shripad S. Indazole compounds, compositions thereof and methods of treatment therewith
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US20050042595A1 (en) * 2003-08-14 2005-02-24 Martin Haas Banking of multipotent amniotic fetal stem cells
US20050058641A1 (en) * 2002-05-22 2005-03-17 Siemionow Maria Z. Tolerance induction and maintenance in hematopoietic stem cell allografts
US20050074435A1 (en) * 2001-12-21 2005-04-07 Robert Casper Cellular compositions and methods of making and using them
US20060004043A1 (en) * 2003-11-19 2006-01-05 Bhagwat Shripad S Indazole compounds and methods of use thereof
US20060008450A1 (en) * 1999-08-05 2006-01-12 Verfaillie Catherine M Use of multipotent adult stem cells in treatment of myocardial infarction and congestive heart failure
US6987184B2 (en) * 2001-02-15 2006-01-17 Signal Pharmaceuticals, Llc Isothiazoloanthrones, isoxazoloanthrones, isoindolanthrones and derivatives thereof as JNK inhibitors and compositions and methods related
US20060024280A1 (en) * 2002-01-22 2006-02-02 Advanced Cell Technology, Inc. Stem cell-derived endothelial cells modified to disrupt tumor angiogenesis
US20060030041A1 (en) * 1999-08-05 2006-02-09 Regents Of The University Of Minnesota Multipotent adult stem cells and methods for isolation
US20070031384A1 (en) * 2005-01-07 2007-02-08 Anthony Atala Regeneration of pancreatic islets by amniotic fluid stem cell therapy
US20070041954A1 (en) * 2005-07-14 2007-02-22 Ichim Thomas E Compositions of placentally-derived stem cells for the treatment of cancer
US7642091B2 (en) * 2005-02-24 2010-01-05 Jau-Nan Lee Human trophoblast stem cells and use thereof
US20110003387A1 (en) * 2009-07-02 2011-01-06 Abbot Stewart Method of producing erythrocytes without feeder cells
US20120020936A1 (en) * 2000-12-06 2012-01-26 Hariri Robert J Treatment of neurodegenerative disease using placental stem cells
US8105634B2 (en) * 2006-08-15 2012-01-31 Anthrogenesis Corporation Umbilical cord biomaterial for medical use
US20120034195A1 (en) * 2002-02-13 2012-02-09 Hariri Robert J Placental stem cells derive from post-partum manmalian placenta, and uses and methods of treatment using said cells

Family Cites Families (131)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4829000A (en) 1985-08-30 1989-05-09 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Reconstituted basement membrane complex with biological activity
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US5004681B1 (en) 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
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
US5399493A (en) 1989-06-15 1995-03-21 The Regents Of The University Of Michigan Methods and compositions for the optimization of human hematopoietic progenitor cell cultures
US5635386A (en) 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
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
US5763266A (en) 1989-06-15 1998-06-09 The Regents Of The University Of Michigan Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells
JPH04501510A (en) 1989-07-25 1992-03-19 セル ジェネシス,インコーポレイティド Homologous recombination for universal donor cells and chimeric mammalian hosts
CA2045175C (en) 1989-11-06 2003-03-18 Arthur I. Skoultchi Production of proteins using homologous recombination
US5272071A (en) 1989-12-22 1993-12-21 Applied Research Systems Ars Holding N.V. Method for the modification of the expression characteristics of an endogenous gene of a given cell line
US5635387A (en) 1990-04-23 1997-06-03 Cellpro, Inc. Methods and device for culturing human hematopoietic cells and their precursors
US6326198B1 (en) 1990-06-14 2001-12-04 Regents Of The University Of Michigan Methods and compositions for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5837539A (en) 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
US5226914A (en) 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5190556A (en) 1991-03-19 1993-03-02 O.B. Tech, Inc. Cord cutter sampler
US5744361A (en) 1991-04-09 1998-04-28 Indiana University Expansion of human hematopoietic progenitor cells in a liquid medium
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
WO1993013206A1 (en) 1991-12-23 1993-07-08 British Bio-Technology Limited Stem cell inhibiting proteins
EP0590156A4 (en) 1992-03-31 1994-07-13 Toray Industries Novel, physiologically active protein and hemopoietic stem cell growth promoter
US5460964A (en) 1992-04-03 1995-10-24 Regents Of The University Of Minnesota Method for culturing hematopoietic cells
WO1994000484A1 (en) 1992-06-22 1994-01-06 Young Henry E Scar inhibitory factor and use thereof
US5672346A (en) 1992-07-27 1997-09-30 Indiana University Foundation Human stem cell compositions and methods
US5849553A (en) 1992-07-27 1998-12-15 California Institute Of Technology Mammalian multipotent neural stem cells
US5693482A (en) 1992-07-27 1997-12-02 California Institute Of Technology Neural chest stem cell assay
CA2148715A1 (en) 1992-11-16 1994-05-26 John E. Wagner Pluripotential quiescent stem cell population
US5772992A (en) 1992-11-24 1998-06-30 G.D. Searle & Co. Compositions for co-administration of interleukin-3 mutants and other cytokines and hematopoietic factors
US5654186A (en) 1993-02-26 1997-08-05 The Picower Institute For Medical Research Blood-borne mesenchymal cells
GB9308271D0 (en) 1993-04-21 1993-06-02 Univ Edinburgh Method of isolating and/or enriching and/or selectively propagating pluripotential animal cells and animals for use in said method
US5372581A (en) 1993-07-21 1994-12-13 Minneapolis Children's Services Corporation Method and apparatus for placental blood collection
IL107483A0 (en) 1993-11-03 1994-02-27 Yeda Res & Dev Bone marrow transplantation
US6001654A (en) 1994-01-28 1999-12-14 California Institute Of Technology Methods for differentiating neural stem cells to neurons or smooth muscle cells using TGT-β super family growth factors
US5942496A (en) 1994-02-18 1999-08-24 The Regent Of The University Of Michigan Methods and compositions for multiple gene transfer into bone cells
DE4422667A1 (en) 1994-06-30 1996-01-04 Boehringer Ingelheim Int Process for the production and cultivation of hematopoietic progenitor cells
US5516532A (en) 1994-08-05 1996-05-14 Children's Medical Center Corporation Injectable non-immunogenic cartilage and bone preparation
US5827742A (en) 1994-09-01 1998-10-27 Beth Israel Deaconess Medical Center, Inc. Method of selecting pluripotent hematopioetic progenitor cells
US5665557A (en) 1994-11-14 1997-09-09 Systemix, Inc. Method of purifying a population of cells enriched for hematopoietic stem cells populations of cells obtained thereby and methods of use thereof
US5914268A (en) 1994-11-21 1999-06-22 National Jewish Center For Immunology & Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5789147A (en) 1994-12-05 1998-08-04 New York Blood Center, Inc. Method for concentrating white cells from whole blood by adding a red cell sedimentation reagent to whole anticoagulated blood
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5906934A (en) 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5677139A (en) 1995-04-21 1997-10-14 President And Fellows Of Harvard College In vitro differentiation of CD34+ progenitor cells into T lymphocytes
US5908782A (en) 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US5830708A (en) 1995-06-06 1998-11-03 Advanced Tissue Sciences, Inc. Methods for production of a naturally secreted extracellular matrix
AU6223296A (en) 1995-06-07 1996-12-30 Novartis Ag Methods for obtaining compositions enriched for hematopoieti c stem cells and antibodies for use therein
US6306575B1 (en) 1995-06-16 2001-10-23 Stemcell Technologies, Inc. Methods for preparing enriched human hematopoietic cell preparations
US5654381A (en) 1995-06-16 1997-08-05 Massachusetts Institute Of Technology Functionalized polyester graft copolymers
WO1997018298A1 (en) 1995-11-14 1997-05-22 Regents Of The University Of Minnesota Ex vivo culture of stem cells
CA2237890C (en) 1995-11-16 2011-03-29 Case Western Reserve University In vitro chondrogenic induction of human mesenchymal stem cells
US5851984A (en) 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US6227202B1 (en) 1996-09-03 2001-05-08 Maulana Azad Medical College Method of organogenesis and tissue regeneration/repair using surgical techniques
US5945337A (en) 1996-10-18 1999-08-31 Quality Biological, Inc. Method for culturing CD34+ cells in a serum-free medium
US5969105A (en) 1996-10-25 1999-10-19 Feng; Yiqing Stem cell factor receptor agonists
JPH10295369A (en) * 1997-02-26 1998-11-10 Japan Tobacco Inc Production of hematopoietic stem cell
US6152142A (en) 1997-02-28 2000-11-28 Tseng; Scheffer C. G. Grafts made from amniotic membrane; methods of separating, preserving, and using such grafts in surgeries
US6231880B1 (en) 1997-05-30 2001-05-15 Susan P. Perrine Compositions and administration of compositions for the treatment of blood disorders
CA2294944A1 (en) 1997-07-03 1999-01-14 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US6077708A (en) 1997-07-18 2000-06-20 Collins; Paul C. Method of determining progenitor cell content of a hematopoietic cell culture
US5968829A (en) 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
US6093531A (en) 1997-09-29 2000-07-25 Neurospheres Holdings Ltd. Generation of hematopoietic cells from multipotent neural stem cells
US6248587B1 (en) 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
WO1999030723A1 (en) * 1997-12-04 1999-06-24 University Of Medicine And Dentistry Of New Jersey Use of human umbilical cord blood for adoptive therapy
US6059968A (en) 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
NZ507730A (en) 1998-05-04 2004-02-27 Point Therapeutics Inc Hematopoietic stimulation
DE69937347T2 (en) 1998-06-08 2008-09-04 Osiris Therapeutics, Inc. REGULATION OF HEMATOPOIETIC STEM CELL DIFFERENTIATION THROUGH THE USE OF HUMAN MESENCHYMAL STEM CELLS
US6713245B2 (en) * 1998-07-06 2004-03-30 Diacrin, Inc. Methods for storing neural cells such that they are suitable for transplantation
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
JP3517359B2 (en) 1998-09-14 2004-04-12 テルモ株式会社 Cell separation / collection apparatus and cell separation / collection method
US6630349B1 (en) 1998-09-23 2003-10-07 Mount Sinai Hospital Trophoblast cell preparations
DE69937966D1 (en) 1998-11-12 2008-02-21 Cytomatrix Llc PRODUCTION OF LYMPHOIDER TISSUE-SPECIFIC CELLS OF HEMATOPOIETIC PRECURSOR CELLS IN A THREE-DIMENSIONAL SYSTEM
US6548299B1 (en) 1999-11-12 2003-04-15 Mark J. Pykett Lymphoid tissue-specific cell production from hematopoietic progenitor cells in three-dimensional devices
US6102871A (en) 1998-11-23 2000-08-15 Coe; Rosemarie O. Blood collection funnel
US6328765B1 (en) 1998-12-03 2001-12-11 Gore Enterprise Holdings, Inc. Methods and articles for regenerating living tissue
IN191359B (en) 1999-04-20 2003-11-29 Nat Inst Immunology
WO2000073421A2 (en) 1999-06-02 2000-12-07 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
WO2001075094A1 (en) 2000-04-04 2001-10-11 Thomas Jefferson University Application of myeloid-origin cells to the nervous system
ATE332708T1 (en) 2000-06-06 2006-08-15 Glaxo Group Ltd CANCER TREATMENT COMPOSITION CONTAINING AN ANTINEOPLASTIC AGENT AND PDE4 INHIBITOR
US7560280B2 (en) 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
AU2002239294B2 (en) 2000-11-22 2006-05-11 Asterias Biotherapeutics, Inc. Tolerizing allografts of pluripotent stem cells
US20080152629A1 (en) 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
EP3246396B1 (en) 2001-02-14 2020-01-29 Celularity, Inc. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US20020132343A1 (en) * 2001-03-19 2002-09-19 Clark Lum System and method for delivering umbilical cord-derived tissue-matched stem cells for transplantation
CA2466880A1 (en) 2001-11-09 2003-05-15 Artecel Sciences, Inc. Methods and compositions for the use of stromal cells to support embryonic and adult stem cells
WO2003042405A2 (en) 2001-11-15 2003-05-22 Children's Medical Center Corporation Methods of isolation, expansion and differentiation of fetal stem cells from chorionic villus, amniotic fluid, and placenta and therapeutic uses thereof
US20030099621A1 (en) 2001-11-29 2003-05-29 Robert Chow Stem cell screening and transplantation therapy for HIV infection
US7799324B2 (en) 2001-12-07 2010-09-21 Geron Corporation Using undifferentiated embryonic stem cells to control the immune system
US7736892B2 (en) 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
WO2003105908A2 (en) 2002-03-15 2003-12-24 Department Of Veterans Affairs, Rehabilitation R & D Service Methods and compositions using cellular asialodeterminants and glycoconjugates for targeting cells to tissues and organs
US20050118715A1 (en) 2002-04-12 2005-06-02 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
ZA200408369B (en) 2002-04-12 2006-11-29 Celgene Corp Methods for identification of modulators of angiogenesis, compounds discovered thereby, and methods of treatment using the compounds
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
MXPA05005673A (en) 2002-11-26 2005-11-23 Anthrogenesis Corp Cytotherapeutics, cytotherapeutic units and methods for treatments using them.
KR20050105467A (en) 2003-02-13 2005-11-04 안트로제네시스 코포레이션 Use of umbilical cord blood to treat individuals having a disease, disorder or condition
US20050089513A1 (en) 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
US20050186182A1 (en) 2003-11-10 2005-08-25 Theresa Deisher Methods of using G-CSF mobilized C-Kit+ cells in the production of embryoid body-like cell clusters for tissue repair and in the treatment of cardiac myopathy
AU2004296765B2 (en) 2003-12-02 2011-03-24 Celgene Corporation Methods and compositions for the treatment and management of hemoglobinopathy and anemia
TWI338714B (en) 2003-12-02 2011-03-11 Cathay General Hospital Method of isolation and enrichment of mesenchymal stem cells from amniotic fluid
US20050176139A1 (en) 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
US20050266391A1 (en) 2004-01-15 2005-12-01 Bennett Brydon L Methods for preserving tissue
KR20070002067A (en) 2004-03-26 2007-01-04 셀진 코포레이션 Systems and methods for providing a stem cell bank
US7244759B2 (en) 2004-07-28 2007-07-17 Celgene Corporation Isoindoline compounds and methods of making and using the same
US7909806B2 (en) 2004-09-23 2011-03-22 Anthrogenesis Corporation Cord blood and placenta collection kit
US7147626B2 (en) 2004-09-23 2006-12-12 Celgene Corporation Cord blood and placenta collection kit
KR20080031735A (en) 2005-06-10 2008-04-10 셀진 코포레이션 Human placental collagen compositions, processes for their preparation, methods of their use and kits comprising the compositions
JP2008544818A (en) 2005-06-30 2008-12-11 アントフロゲネシス コーポレーション Restoration of the tympanic membrane using placenta-derived collagen biofibers
EP1919365A2 (en) 2005-07-13 2008-05-14 Anthrogenesis Corporation Ocular plug formed from placenta derived collagen biofabric
US7928280B2 (en) 2005-07-13 2011-04-19 Anthrogenesis Corporation Treatment of leg ulcers using placenta derived collagen biofabric
WO2007047465A1 (en) 2005-10-13 2007-04-26 Anthrogenesis Corporation Production of oligodendrocytes from placenta-derived stem cells
SI1957633T1 (en) 2005-10-13 2014-04-30 Anthrogenesis Corporation Immunomodulation using placental stem cells
NZ568618A (en) 2005-12-29 2011-10-28 Anthrogenesis Corp Co-culture of placental stem cells and stem cells from a second source
CN101395266B (en) 2005-12-29 2018-06-15 人类起源公司 placental stem cell populations
KR20080097190A (en) 2005-12-29 2008-11-04 안트로제네시스 코포레이션 Improved composition for collecting and preserving placental stem cells and methods of using the composition
JP2009539378A (en) 2006-06-09 2009-11-19 アントフロゲネシス コーポレーション Placental environment and its use for culturing stem cells
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
WO2008042441A1 (en) 2006-10-03 2008-04-10 Anthrogenesis Corporation Use of umbilical cord biomaterial for ocular surgery
US8071135B2 (en) 2006-10-04 2011-12-06 Anthrogenesis Corporation Placental tissue compositions
KR20180086533A (en) 2006-10-06 2018-07-31 안트로제네시스 코포레이션 Native(telopeptide) Placental Collagen Compositions
EP2084268B1 (en) 2006-10-23 2018-09-26 Celularity, Inc. Methods and compositions for treatment of bone defects with placental cell populations
EP2687220A3 (en) 2007-02-12 2014-05-14 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
JP2010518812A (en) 2007-02-12 2010-06-03 アンスロジェネシス コーポレーション Hepatocytes and chondrocytes derived from adherent placental stem cells, and enriched cell populations of CD34 +, CD45− placental stem cells
KR20190050867A (en) 2007-09-26 2019-05-13 안트로제네시스 코포레이션 Angiogenic cells from human placental perfusate
CA3018281C (en) 2007-09-28 2022-02-22 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US20090136471A1 (en) 2007-11-07 2009-05-28 Anthrogenesis Corporation Treatment of premature birth complications
KR20200011604A (en) 2008-08-20 2020-02-03 안트로제네시스 코포레이션 Improved cell composition and methods of making the same
EP2329012B1 (en) 2008-08-20 2020-05-20 Celularity, Inc. Treatment of stroke using isolated placental cells
CA2965883C (en) 2008-08-22 2020-05-12 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
BRPI0922184A2 (en) 2008-11-19 2020-08-18 Anthrogenesis Corporation isolated cell, isolated cell population, degradable permanent or decellularized matrix or synthetic structure, and method for treating an individual
RU2732240C2 (en) 2008-11-21 2020-09-14 Антродженезис Корпорейшн Treating diseases, disorders or pathological conditions of the lungs using placental cells

Patent Citations (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3862002A (en) * 1962-05-08 1975-01-21 Sanfar Lab Inc Production of physiologically active placental substances
US4008719A (en) * 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US5391485A (en) * 1985-08-06 1995-02-21 Immunex Corporation DNAs encoding analog GM-CSF molecules displaying resistance to proteases which cleave at adjacent dibasic residues
US5393870A (en) * 1985-08-06 1995-02-28 Immunex Corporation Analogs of human granulocyte-macrophage colony stimulating factor
US4999291A (en) * 1985-08-23 1991-03-12 Amgen Inc. Production of human pluripotent granulocyte colony-stimulating factor
US4810643A (en) * 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US4798824A (en) * 1985-10-03 1989-01-17 Wisconsin Alumni Research Foundation Perfusate for the preservation of organs
US6022743A (en) * 1986-04-18 2000-02-08 Advanced Tissue Sciences, Inc. Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
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
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
US5605822A (en) * 1989-06-15 1997-02-25 The Regents Of The University Of Michigan Methods, compositions and devices for growing human hematopoietic cells
US5487992A (en) * 1989-08-22 1996-01-30 University Of Utah Research Foundation Cells and non-human organisms containing predetermined genomic modifications and positive-negative selection methods and vectors for making same
US5716827A (en) * 1990-03-30 1998-02-10 Systemix, Inc. Human hematopoietic stem cell
US5733566A (en) * 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US6010696A (en) * 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US5733542A (en) * 1990-11-16 1998-03-31 Haynesworth; Stephen E. Enhancing bone marrow engraftment using MSCS
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5385901A (en) * 1991-02-14 1995-01-31 The Rockefeller University Method of treating abnormal concentrations of TNF α
US5192312A (en) * 1991-03-05 1993-03-09 Colorado State University Research Foundation Treated tissue for implantation and methods of treatment and use
US5591767A (en) * 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6022848A (en) * 1993-03-31 2000-02-08 Pro-Neuron, Inc. Inhibitor of stem cell proliferation and uses thereof
US5709854A (en) * 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
US5605914A (en) * 1993-07-02 1997-02-25 Celgene Corporation Imides
US5877200A (en) * 1993-07-02 1999-03-02 Celgene Corporation Cyclic amides
US5599705A (en) * 1993-11-16 1997-02-04 Cameron; Robert B. In vitro method for producing differentiated universally compatible mature human blood cells
US5591625A (en) * 1993-11-24 1997-01-07 Case Western Reserve University Transduced mesenchymal stem cells
US6020469A (en) * 1993-12-22 2000-02-01 Amgen Inc. Stem cell factor formulations and methods
US5855619A (en) * 1994-06-06 1999-01-05 Case Western Reserve University Biomatrix for soft tissue regeneration
US6174333B1 (en) * 1994-06-06 2001-01-16 Osiris Therapeutics, Inc. Biomatrix for soft tissue regeneration using mesenchymal stem cells
US5879940A (en) * 1994-07-20 1999-03-09 Fred Hutchinson Cancer Research Center Human marrow stromal cell lines which sustain hematopoieses
US5861315A (en) * 1994-11-16 1999-01-19 Amgen Inc. Use of stem cell factor and soluble interleukin-6 receptor for the ex vivo expansion of hematopoietic multipotential cells
US5874301A (en) * 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5866414A (en) * 1995-02-10 1999-02-02 Badylak; Stephen F. Submucosa gel as a growth substrate for cells
US6011000A (en) * 1995-03-03 2000-01-04 Perrine; Susan P. Compositions for the treatment of blood disorders
US5716616A (en) * 1995-03-28 1998-02-10 Thomas Jefferson University Isolated stromal cells for treating diseases, disorders or conditions characterized by bone defects
US5733541A (en) * 1995-04-21 1998-03-31 The Regent Of The University Of Michigan Hematopoietic cells: compositions and methods
US6338942B2 (en) * 1995-05-19 2002-01-15 T. Breeders, Inc. Selective expansion of target cell populations
US5877299A (en) * 1995-06-16 1999-03-02 Stemcell Technologies Inc. Methods for preparing enriched human hematopoietic cell preparations
US5858782A (en) * 1995-11-13 1999-01-12 Regents Of The University Of Michigan Functional human hematopoietic cells
US6337387B1 (en) * 1995-11-17 2002-01-08 Asahi Kasei Kabushiki Kaisha Differentiation-suppressive polypeptide
US5716794A (en) * 1996-03-29 1998-02-10 Xybernaut Corporation Celiac antigen
US6190368B1 (en) * 1996-05-14 2001-02-20 Children's Hospital Medical Center Of Northern California Apparatus and method for collecting blood from an umbilical cord
US6335349B1 (en) * 1996-07-24 2002-01-01 Celgene Corporation Substituted 2(2,6-dioxopiperidin-3-yl)isoindolines
US6709864B1 (en) * 1996-07-30 2004-03-23 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US6179819B1 (en) * 1996-08-30 2001-01-30 John N. Haswell Umbilical cord blood collection
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US5879318A (en) * 1997-08-18 1999-03-09 Npbi International B.V. Method of and closed system for collecting and processing umbilical cord blood
US6022540A (en) * 1997-09-04 2000-02-08 Osiris Therapeutics, Inc. Ligands that modulate differentiation of mesenchymal stem cells
US5874448A (en) * 1997-11-18 1999-02-23 Celgene Corporation Substituted 2-(2,6 dioxo-3-fluoropiperidin-3-yl)-isoindolines and method of reducing TNFα levels
US6355239B1 (en) * 1998-03-13 2002-03-12 Osiris Therapeutics, Inc. Uses for non-autologous mesenchymal stem cells
US6528245B2 (en) * 1998-05-07 2003-03-04 University Of South Florida Bone marrow cells as a source of neurons for brain and spinal cord repair
US6030836A (en) * 1998-06-08 2000-02-29 Osiris Therapeutics, Inc. Vitro maintenance of hematopoietic stem cells
US6184035B1 (en) * 1998-11-18 2001-02-06 California Institute Of Technology Methods for isolation and activation of, and control of differentiation from, skeletal muscle stem or progenitor cells
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US6534084B1 (en) * 1999-06-30 2003-03-18 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US20060030041A1 (en) * 1999-08-05 2006-02-09 Regents Of The University Of Minnesota Multipotent adult stem cells and methods for isolation
US20060008450A1 (en) * 1999-08-05 2006-01-12 Verfaillie Catherine M Use of multipotent adult stem cells in treatment of myocardial infarction and congestive heart failure
US6685936B2 (en) * 1999-10-12 2004-02-03 Osiris Therapeutics, Inc. Suppressor cells induced by culture with mesenchymal stem cells for treatment of immune responses in transplantation
US20020028510A1 (en) * 2000-03-09 2002-03-07 Paul Sanberg Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US20030003573A1 (en) * 2000-04-27 2003-01-02 Lakshmi Rambhatle Hepatocytes for therapy and drug screening made from embryonic stem cells
US20050009876A1 (en) * 2000-07-31 2005-01-13 Bhagwat Shripad S. Indazole compounds, compositions thereof and methods of treatment therewith
US6538023B1 (en) * 2000-09-15 2003-03-25 Tsuyoshi Ohnishi Therapeutic uses of green tea polyphenols for sickle cell disease
US20040048372A1 (en) * 2000-12-06 2004-03-11 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20030032179A1 (en) * 2000-12-06 2003-02-13 Hariri Robert J. Post-partum mammalian placenta, its use and placental stem cells therefrom
US20120020936A1 (en) * 2000-12-06 2012-01-26 Hariri Robert J Treatment of neurodegenerative disease using placental stem cells
US20050019908A1 (en) * 2000-12-06 2005-01-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20030045552A1 (en) * 2000-12-27 2003-03-06 Robarge Michael J. Isoindole-imide compounds, compositions, and uses thereof
US6987184B2 (en) * 2001-02-15 2006-01-17 Signal Pharmaceuticals, Llc Isothiazoloanthrones, isoxazoloanthrones, isoindolanthrones and derivatives thereof as JNK inhibitors and compositions and methods related
US20030044977A1 (en) * 2001-08-10 2003-03-06 Norio Sakuragawa Human stem cells originated from human amniotic mesenchymal cell layer
US20030044976A1 (en) * 2001-08-27 2003-03-06 Advanced Cell Technology De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20030059414A1 (en) * 2001-09-21 2003-03-27 Ho Tony W. Cell populations which co-express CD49c and CD90
US20050074435A1 (en) * 2001-12-21 2005-04-07 Robert Casper Cellular compositions and methods of making and using them
US20060024280A1 (en) * 2002-01-22 2006-02-02 Advanced Cell Technology, Inc. Stem cell-derived endothelial cells modified to disrupt tumor angiogenesis
US20120034195A1 (en) * 2002-02-13 2012-02-09 Hariri Robert J Placental stem cells derive from post-partum manmalian placenta, and uses and methods of treatment using said cells
US20040048796A1 (en) * 2002-03-26 2004-03-11 Hariri Robert J. Collagen biofabric and methods of preparation and use therefor
US20030235563A1 (en) * 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20050058641A1 (en) * 2002-05-22 2005-03-17 Siemionow Maria Z. Tolerance induction and maintenance in hematopoietic stem cell allografts
US20040028660A1 (en) * 2002-05-30 2004-02-12 Anthrogenesis Corporation Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US20040018817A1 (en) * 2002-07-18 2004-01-29 Omron Corporation Communication system, communication apparatus, and communication control method
US20040018617A1 (en) * 2002-07-26 2004-01-29 Shiaw-Min Hwang Somatic pluripotent cells
US20040058412A1 (en) * 2002-09-20 2004-03-25 Neuronyx, Inc. Cell populations which co-express CD49c and CD90
US20050058629A1 (en) * 2003-06-27 2005-03-17 Harmon Alexander M. Soft tissue repair and regeneration using postpartum-derived cells
US20070036767A1 (en) * 2003-06-27 2007-02-15 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20050058631A1 (en) * 2003-06-27 2005-03-17 Kihm Anthony J. Postpartum cells derived from placental tissue, and methods of making and using the same
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US20050054098A1 (en) * 2003-06-27 2005-03-10 Sanjay Mistry Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20050058630A1 (en) * 2003-06-27 2005-03-17 Harris Ian Ross Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US20050032209A1 (en) * 2003-06-27 2005-02-10 Messina Darin J. Regeneration and repair of neural tissue using postpartum-derived cells
US20050037491A1 (en) * 2003-06-27 2005-02-17 Sanjay Mistry Repair and regeneration of ocular tissue using postpartum-derived cells
US20070009494A1 (en) * 2003-06-27 2007-01-11 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20070014771A1 (en) * 2003-06-27 2007-01-18 Ethicon, Incorporated Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20050054093A1 (en) * 2003-08-14 2005-03-10 Martin Haas Multipotent amniotic fetal stem cells
US20050042595A1 (en) * 2003-08-14 2005-02-24 Martin Haas Banking of multipotent amniotic fetal stem cells
US20060004043A1 (en) * 2003-11-19 2006-01-05 Bhagwat Shripad S Indazole compounds and methods of use thereof
US20070031384A1 (en) * 2005-01-07 2007-02-08 Anthony Atala Regeneration of pancreatic islets by amniotic fluid stem cell therapy
US7642091B2 (en) * 2005-02-24 2010-01-05 Jau-Nan Lee Human trophoblast stem cells and use thereof
US20070041954A1 (en) * 2005-07-14 2007-02-22 Ichim Thomas E Compositions of placentally-derived stem cells for the treatment of cancer
US8105634B2 (en) * 2006-08-15 2012-01-31 Anthrogenesis Corporation Umbilical cord biomaterial for medical use
US20110003387A1 (en) * 2009-07-02 2011-01-06 Abbot Stewart Method of producing erythrocytes without feeder cells

Cited By (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8580563B2 (en) * 2000-12-06 2013-11-12 Anthrogenesis Corporation Placental stem cells
US8545833B2 (en) 2000-12-06 2013-10-01 Anthrogenesis Corporation Treatment of radiation injury using placental stem cells
US20050019908A1 (en) * 2000-12-06 2005-01-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US8986984B2 (en) 2000-12-06 2015-03-24 Anthrogenesis Corporation Method of propagating cells
US9149569B2 (en) 2000-12-06 2015-10-06 Anthrogenesis Corporation Treatment of diseases or disorders using placental stem cells
US9387283B2 (en) 2000-12-06 2016-07-12 Anthrogenesis Corporation Method of collecting placental stem cells
US8293223B2 (en) 2000-12-06 2012-10-23 Anthrogenesis Corporation Treatment of organ injuries and burns using placental stem cells
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US7976836B2 (en) 2000-12-06 2011-07-12 Anthrogenesis Corporation Treatment of stroke using placental stem cells
US20100120015A1 (en) * 2000-12-06 2010-05-13 Hariri Robert J Method of collecting placental stem cells
US20090053805A1 (en) * 2000-12-06 2009-02-26 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20080131966A1 (en) * 2001-02-14 2008-06-05 Hariri Robert J Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US9139813B2 (en) 2001-02-14 2015-09-22 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US7311904B2 (en) 2001-02-14 2007-12-25 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making the same
US7914779B2 (en) 2001-02-14 2011-03-29 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making the same
US20020160510A1 (en) * 2001-02-14 2002-10-31 Hariri Robert J. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US8435788B2 (en) 2001-02-14 2013-05-07 Anthrogenesis Corporation Tissue matrices comprising placental stem cells
US20050186218A1 (en) * 2001-04-13 2005-08-25 Wyeth Holdings Corporation Removal of bacterial endotoxin in a protein solution by immobilized metal affinity chromatography
US8057789B2 (en) 2002-02-13 2011-11-15 Anthrogenesis Corporation Placental stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US8753883B2 (en) 2002-02-13 2014-06-17 Anthrogenesis Corporation Treatment of psoriasis using placental stem cells
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US8889411B2 (en) 2002-04-12 2014-11-18 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20050118715A1 (en) * 2002-04-12 2005-06-02 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20050148034A1 (en) * 2002-04-12 2005-07-07 Hariri Robert J. Methods for identification of modulators of angiogenesis, compounds discovered thereby, and methods of treatment using the compounds
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20030235909A1 (en) * 2002-04-12 2003-12-25 Hariri Robert J. Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US8617535B2 (en) 2002-11-26 2013-12-31 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US20150193581A1 (en) * 2004-03-26 2015-07-09 Celgene Corporation Systems and methods for providing a stem cell bank
US20050276792A1 (en) * 2004-03-26 2005-12-15 Kaminski Joseph K Systems and methods for providing a stem cell bank
US20120046968A1 (en) * 2004-03-26 2012-02-23 Kaminski Joseph K Systems and methods for providing a stem cell bank
US20060040392A1 (en) * 2004-04-23 2006-02-23 Collins Daniel P Multi-lineage progenitor cells
US8163275B2 (en) 2004-04-23 2012-04-24 Bioe Llc Multi-lineage progenitor cells
US7670596B2 (en) 2004-04-23 2010-03-02 Bioe, Inc. Multi-lineage progenitor cells
US7790458B2 (en) 2004-05-14 2010-09-07 Becton, Dickinson And Company Material and methods for the growth of hematopoietic stem cells
US20060147431A1 (en) * 2005-01-04 2006-07-06 Nobuko Uchida Methods for the treatment of lysosomal storage disorders
WO2006074387A1 (en) * 2005-01-04 2006-07-13 Stemcells California, Inc. Methods for the treatment of lysosomal storage disorders
US8071376B2 (en) 2005-10-13 2011-12-06 Anthrogenesis Corporation Production of oligodendrocytes from placenta-derived stem cells
US9539288B2 (en) 2005-10-13 2017-01-10 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8216566B2 (en) 2005-10-13 2012-07-10 Anthrogenesis Corporation Treatment of multiple sclerosis using placental stem cells
US20070134210A1 (en) * 2005-10-13 2007-06-14 Mohammad Heidaran Production of oligodendrocytes from placenta-derived stem cells
US7682803B2 (en) 2005-10-13 2010-03-23 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8895256B2 (en) 2005-10-13 2014-11-25 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8455250B2 (en) 2005-12-29 2013-06-04 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US9078898B2 (en) 2005-12-29 2015-07-14 Anthrogenesis Corporation Placental stem cell populations
US10383897B2 (en) 2005-12-29 2019-08-20 Celularity, Inc. Placental stem cell populations
US9598669B2 (en) 2005-12-29 2017-03-21 Anthrogenesis Corporation Composition for collecting placental stem cells and methods of using the composition
US9725694B2 (en) 2005-12-29 2017-08-08 Anthrogenesis Corporation Composition for collecting and preserving placental stem cells and methods of using the composition
US8691217B2 (en) 2005-12-29 2014-04-08 Anthrogenesis Corporation Placental stem cell populations
US8202703B2 (en) 2005-12-29 2012-06-19 Anthrogenesis Corporation Placental stem cell populations
US20070190042A1 (en) * 2005-12-29 2007-08-16 Edinger James W Composition for collecting and preserving placental stem cells and methods of using the composition
US8591883B2 (en) 2005-12-29 2013-11-26 Anthrogenesis Corporation Placental stem cell populations
US20070249047A1 (en) * 2006-04-17 2007-10-25 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Respiratory Epithelial Cells
US7727763B2 (en) 2006-04-17 2010-06-01 Bioe, Llc Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US9433647B2 (en) 2006-08-17 2016-09-06 Mimedx Group, Inc. Placental tissue grafts
US8597687B2 (en) 2006-08-17 2013-12-03 Mimedx Group, Inc. Methods for determining the orientation of a tissue graft
US11504449B2 (en) 2006-08-17 2022-11-22 Mimedx Group, Inc. Placental tissue grafts and methods of preparing and using the same
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
US8623421B2 (en) 2006-08-17 2014-01-07 Mimedx Group, Inc. Placental graft
US9272005B2 (en) 2006-08-17 2016-03-01 Mimedx Group, Inc. Placental tissue grafts
US8460716B2 (en) 2006-08-17 2013-06-11 Mimedx Group, Inc. Method for applying a label to a placental tissue graft
US9265800B2 (en) 2006-08-17 2016-02-23 Mimedx Group, Inc. Placental tissue grafts
US9956253B2 (en) 2006-08-17 2018-05-01 Mimedx Group, Inc. Placental tissue grafts
US8709494B2 (en) 2006-08-17 2014-04-29 Mimedx Group, Inc. Placental tissue grafts
US10406259B2 (en) 2006-08-17 2019-09-10 Mimedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
US9572839B2 (en) 2006-08-17 2017-02-21 Mimedx Group, Inc. Placental tissue grafts and methods of preparing and using the same
US9265801B2 (en) 2006-08-17 2016-02-23 Mimedx Group, Inc. Placental tissue grafts
US9463207B2 (en) 2006-08-17 2016-10-11 Mimedx Group, Inc. Placental tissue grafts
US8460715B2 (en) 2006-08-17 2013-06-11 Mimedx Group, Inc. Placental tissue grafts
US9339520B2 (en) 2006-10-23 2016-05-17 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US10105399B2 (en) 2006-10-23 2018-10-23 Celularity, Inc. Methods and compositions for treatment of bone defects with placental cell populations
US8562972B2 (en) 2006-10-23 2013-10-22 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8916146B2 (en) 2007-02-12 2014-12-23 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US10494607B2 (en) 2007-02-12 2019-12-03 Celularity, Inc. CD34+,CD45−placental stem cell-enriched cell populations
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
US8709493B2 (en) 2007-09-07 2014-04-29 Mimedx Group, Inc. Placental tissue grafts
US8372438B2 (en) 2007-09-07 2013-02-12 Mimedx Group, Inc. Method for inhibiting adhesion formation using an improved placental tissue graft
US9084767B2 (en) 2007-09-07 2015-07-21 Mimedx Group, Inc. Placental tissue grafts and methods of preparing and using the same
US20100104539A1 (en) * 2007-09-07 2010-04-29 John Daniel Placental tissue grafts and improved methods of preparing and using the same
US8372439B2 (en) 2007-09-07 2013-02-12 Mimedx Group, Inc. Method for treating a wound using improved placental tissue graft
US8357403B2 (en) 2007-09-07 2013-01-22 Mimedx Group, Inc. Placental tissue grafts
US8932643B2 (en) 2007-09-07 2015-01-13 Mimedx Group, Inc. Placental tissue grafts
US9789137B2 (en) 2007-09-07 2017-10-17 Mimedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
US8409626B2 (en) 2007-09-07 2013-04-02 Mimedx Group, Inc. Placental tissue grafts
US9533011B2 (en) 2007-09-07 2017-01-03 Mimedx Group, Inc. Placental tissue grafts and methods of preparing and using the same
US10874697B2 (en) 2007-09-07 2020-12-29 Mimedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
US8703207B2 (en) 2007-09-07 2014-04-22 Mimedx Group, Inc. Placental tissue grafts
US8703206B2 (en) 2007-09-07 2014-04-22 Mimedx Group, Inc. Placental tissue grafts
US9272003B2 (en) 2007-09-07 2016-03-01 Mimedx Group, Inc. Placental tissue grafts
US8642092B2 (en) 2007-09-07 2014-02-04 Mimedx Group, Inc. Placental tissue grafts
US11752174B2 (en) 2007-09-07 2023-09-12 Mimedx Group, Inc. Placental tissue grafts and improved methods of preparing and using the same
US8323701B2 (en) 2007-09-07 2012-12-04 Mimedx Group, Inc. Placental tissue grafts
US9415074B2 (en) 2007-09-07 2016-08-16 Mimedx Group, Inc. Placental tissue grafts
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US9216200B2 (en) 2007-09-28 2015-12-22 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US9198938B2 (en) 2008-11-19 2015-12-01 Antrhogenesis Corporation Amnion derived adherent cells
US9255248B2 (en) 2009-07-02 2016-02-09 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
US8586360B2 (en) 2009-07-02 2013-11-19 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
US20110020293A1 (en) * 2009-07-21 2011-01-27 Abt Holding Company Use of Stem Cells to Reduce Leukocyte Extravasation
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US20110212069A1 (en) * 2010-02-25 2011-09-01 Abt Holding Company Modulation of Microglia Activation
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US9464274B2 (en) 2010-07-13 2016-10-11 Anthrogenesis Corporation Methods of generating natural killer cells
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US11090339B2 (en) 2011-06-01 2021-08-17 Celularity Inc. Treatment of pain using placental stem cells
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta

Also Published As

Publication number Publication date
US20170290862A1 (en) 2017-10-12
AU2003298775A1 (en) 2004-06-18
KR20050086780A (en) 2005-08-30
CA2505534A1 (en) 2004-06-10
KR20090092351A (en) 2009-08-31
AU2008229977A1 (en) 2008-11-06
MXPA05005673A (en) 2005-11-23
ZA200504273B (en) 2006-08-30
KR101042448B1 (en) 2011-06-16
EP1571910A4 (en) 2009-10-28
KR20100125479A (en) 2010-11-30
CN1717177A (en) 2006-01-04
US8617535B2 (en) 2013-12-31
AU2003298775B2 (en) 2008-07-17
JP2011225606A (en) 2011-11-10
WO2004047770A2 (en) 2004-06-10
US20070092497A1 (en) 2007-04-26
US20200009199A1 (en) 2020-01-09
EP1571910A2 (en) 2005-09-14
BR0316695A (en) 2005-10-18
WO2004047770A3 (en) 2004-08-19
JP2006509770A (en) 2006-03-23

Similar Documents

Publication Publication Date Title
US8617535B2 (en) Cytotherapeutics, cytotherapeutic units and methods for treatments using them
Bednarski et al. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant
Choudhury et al. Dendritic cells derived in vitro from acute myelogenous leukemia cells stimulate autologous, antileukemic T-cell responses
Dolcetti et al. Hierarchy of immunosuppressive strength among myeloid‐derived suppressor cell subsets is determined by GM‐CSF
Mapara et al. Monitoring of tumor cell purging after highly efficient immunomagnetic selection of CD34 cells from leukapheresis products in breast cancer patients: comparison of immunocytochemical tumor cell staining and reverse transcriptase–polymerase chain reaction
Vescio et al. The hematopoietic stem cell antigen, CD34, is not expressed on the malignant cells in multiple myeloma
DE10132502A1 (en) Attack on tumor cells with missing, low or abnormal MHC expression by combining non-MHC-restricted T cells / NK cells and MHC-restricted cells
Guo et al. Direct recognition and lysis of leukemia cells by WT1-specific CD4+ T lymphocytes in an HLA class II-restricted manner
Sekhsaria et al. Granulocyte colony-stimulating factor recruitment of CD34+ progenitors to peripheral blood: impaired mobilization in chronic granulomatous disease and adenosine deaminase--deficient severe combined immunodeficiency disease patients
TW201130978A (en) Method of isolation of stem cell populations from peripheral blood using sized-based separation (elutriation)
US20070286807A1 (en) Method Of Inducing Or Modulating Immune Response
Ryu et al. Granulocyte macrophage-colony stimulating factor produces a splenic subset of monocyte-derived dendritic cells that efficiently polarize T helper type 2 cells in response to blood-borne antigen
Kufner et al. Leukemia‐derived dendritic cells can be generated from blood or bone marrow cells from patients with acute myeloid leukaemia: A methodological approach under serum‐free culture conditions
Carriglio et al. Good Laboratory Practice preclinical safety studies for GSK2696273 (MLV vector-based ex vivo gene therapy for adenosine deaminase deficiency severe combined immunodeficiency) in NSG mice
WO2023081320A1 (en) Therapeutic compositions and methods for allogeneic hematopoietic stem cell transplantation
US20230052157A1 (en) Method for obtaining nucleic acid for sequencing
NZ553745A (en) Cytotherapeutic units comprising CD34-, OCT-4+ postpartum placental cells and methods of using such in the manufacture of medicaments
US8802434B2 (en) Biological cell culture, cell culture media and therapeutic use of biological cells
To et al. Single-cell transcriptomics reveal distinct subsets of activated dendritic cells in the tumor microenvironment
Crough et al. Donor-derived b2a2-specific T cells for immunotherapy of patients with chronic myeloid leukemia
Danby et al. Clinical use of umbilical cord blood cells
Hassab et al. Study of CD25 expression on leukemic cells: a prognostic factor in acute myeloid leukemia
WO2023086595A1 (en) Biomarkers of megakaryocyte-derived extracellular vesicles
Yamagami Exploiting molecules involved in fetal-maternal tolerance to overcome immunologic barriers
De Silvestro et al. Legislative and ethical aspects of administering granulocyte colony-stimulating factor to normal donors

Legal Events

Date Code Title Description
AS Assignment

Owner name: ANTHROGENESIS CORPORATION, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HARIRI, ROBERT J.;REEL/FRAME:015340/0205

Effective date: 20040505

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: CLARITY ACQUISITION II LLC, NEW JERSEY

Free format text: MERGER;ASSIGNOR:ANTHROGENESIS CORPORATION;REEL/FRAME:044413/0680

Effective date: 20170815

AS Assignment

Owner name: CELULARITY, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CLARITY ACQUISITION II LLC;REEL/FRAME:044780/0261

Effective date: 20171103