EP2490702A1 - Treatment using reprogrammed mature adult cells - Google Patents

Treatment using reprogrammed mature adult cells

Info

Publication number
EP2490702A1
EP2490702A1 EP09736651A EP09736651A EP2490702A1 EP 2490702 A1 EP2490702 A1 EP 2490702A1 EP 09736651 A EP09736651 A EP 09736651A EP 09736651 A EP09736651 A EP 09736651A EP 2490702 A1 EP2490702 A1 EP 2490702A1
Authority
EP
European Patent Office
Prior art keywords
cells
committed
patient
cell
reprogrammed
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.)
Withdrawn
Application number
EP09736651A
Other languages
German (de)
English (en)
French (fr)
Inventor
Ilham Mohamed Saleh Saeed Abuljadayel
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.)
Tristem Trading Cyprus Ltd
Original Assignee
Tristem Trading Cyprus Ltd
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 Tristem Trading Cyprus Ltd filed Critical Tristem Trading Cyprus Ltd
Publication of EP2490702A1 publication Critical patent/EP2490702A1/en
Withdrawn legal-status Critical Current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
    • A61K35/14Blood; Artificial blood
    • 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
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised 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
    • A61K35/22Urine; Urinary tract, e.g. kidney or bladder; Intraglomerular mesangial cells; Renal mesenchymal cells; Adrenal gland
    • 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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived 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
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • 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
    • A61K35/34Muscles; Smooth muscle cells; Heart; Cardiac stem cells; Myoblasts; Myocytes; Cardiomyocytes
    • 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
    • A61K35/37Digestive system
    • A61K35/39Pancreas; Islets of Langerhans
    • 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
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • 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
    • A61P25/24Antidepressants
    • 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
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure

Definitions

  • a method of treating various diseases, disorders, or conditions in patient using reprogrammed cells such as retrodifferentiated, transdifferentiated, or redifferentiated cells.
  • the method comprises obtaining committed cells from the patient, retrodifferentiating the committed cells to obtain retrodifferentiated target cells, and administering the
  • the method comprises obtaining committed cells from the patient, transdifferentiating the committed cells to obtain transdifferentiated target cells, and administering the transdifferentiated target cells to the patient.
  • the retrodifferentiated or transdifferentiated target cells repair or replenish tissue or cells in the patient.
  • Stem cells are characterized by their ability to renew themselves through mitotic cell division and to differentiate into a diverse range of specialized cell types.
  • the two broad types of mammalian stem cells are embryonic stem cells, which are isolated from the inner cell mass of blastocysts, and adult stem cells, which are found in adult tissues.
  • stem cells can differentiate into all of the specialized embryonic tissues.
  • stem cells and progenitor cells replenish specialized cells and maintain the normal turnover of regenerative organs such as blood, skin or intestinal tissues.
  • Stem cells are abundant in a developing embryo, although, the quantity of stem cells decreases as development progresses.
  • an adult organism contains a limited number of stem cells which are confined to certain body compartments.
  • stem cells have the potential to alter treatments for multiple diseases or disorders. While some adult stem cell therapies, such as bone marrow transplants, already exist, medical researchers anticipate using stem cells to treat a wider variety of diseases including cancer, Parkinson's disease, spinal cord injuries, amyotrophic lateral sclerosis, multiple sclerosis, and muscle damage, among others. Such therapies may take advantage of the stem cells' capability of differentiating into cell types that are necessary to treat the disease.
  • haematopoietic stem cells are traditionally extracted by isolation from bone marrow, growth factor mobilized peripheral blood, or cord blood (placenta).
  • Haematopoietic stem cells may also be prepared from embryonic stem (ES) cells, which are extracted from embryos obtained using in vitro fertilization techniques.
  • ES embryonic stem
  • stem cells that may be obtained from these sources are limited. Moreover, the stem cells may experience difficulty in differentiating into the cells necessary to treat the ailment.
  • the present invention relates to the use of reprogrammed cells to repair tissue or replenish tissue or cells in a patient.
  • the present invention relates to the use of retrodifferentiated cells, obtained from retrodifferentiation of differentiated or committed cells, to repair tissue or replenish tissue or cells in a patient.
  • the present invention also relates to the use of transdifferentiated cells, obtained from transdifferentiation of differentiated or committed cells, to repair tissue or replenish tissue or cells in a patient.
  • the application is based, in part, on Applicant's discovery that committed or somatic cells obtained from a patient may be reprogrammed to result in cells of different lineages, and these reprogrammed cells can be administered to the patient to repair or replenish tissues or cells. Examples of the process of reprogramming include retrodifferentiation and transdifferentiation.
  • committed cells may undergo reprogramming to result in cells of a different lineage.
  • committed cells may undergo retrodifferentiation to result in retrodiffereiitiated cells, e.g., cells that are less differentiated, such as pluripotent stem cells, and that these retrodifferentiated cells can be administered to the patient to repair or replenish tissues or cells.
  • retrodiffereiitiated cells e.g., cells that are less differentiated, such as pluripotent stem cells
  • these retrodifferentiated cells can be administered to the patient to repair or replenish tissues or cells.
  • committed cells obtained fi-om a patient may undergo transdifferentiation to result in transdifferentiated cells, e.g., cells of a different lineage than the committed cells, and that these transdifferentiated cells may be administered to the patient to repair or replenish tissues or cells.
  • the invention encompasses a method of repairing or replenishing tissue or cells of a cell lineage in a patient by administering reprogrammed cells to the patient.
  • the invention encompasses a method of repairing or replenishing tissue or cells of a cell lineage in a patient, comprising (i) obtaining committed cells, (ii) retrodifferentiating the committed cells to obtain retrodifferentiated target cells, and (iii) administering the retrodifferentiated target cells to the patient, wherein the retrodifferentiated target cells redifferentiate into cells of the cell lineage.
  • These redifferentiated cells may be of the same cell lineage or of a different cell lineage as the committed cells.
  • the invention also encompasses a method of repairing or replenishing tissue or cells of a cell lineage in a patient, comprising (i) obtaining committed cells, (ii)
  • transdifferentiating the committed cells to obtain transdifferentiated target cells, and (iii) administering the transdifferentiated target cells to the patient.
  • the patient may be suffering from a disease, disorder, or condition including, but not limited to, bone marrow failure, haematological conditions, aplastic anemia, beta-thalassemia, diabetes, motor neuron disease, Parkinson's disease, spinal cord injury, muscular dystrophy, kidney disease, multiple sclerosis, congestive heart failure, hepatitis C virus, human immunodeficiency virus, head trauma, spinal cord injuries, lung disease, depression, non-obstructive azoospermia, andropause, menopause and infertility, rejuvenation, sclerodenna ulcers, psoriasis, wrinkles, liver cirrhosis, autoimmune disease, alopecia, retinitis pigmentosa, crystalline dystrophy/blindness, diabetes, and infertility.
  • the reprogrammed cells may be bone marrow cells that treat aplastic anemia, leukemia, lymphoma, or human immunodefic
  • the reprogrammed target cells may include, but are not limited to, pluripotent stem cells, pluripotent genu cells, haematopoietic stem cells, neuronal stem cells, epithelial stem cells, mesenchymal stem cells, endodermal and neuroectodermal stem cells, germ cells, extraembryonic, embryonic stem cells, kidney cells, alveolar epithelium cells, endoderm cells, neurons, ectoderm cells, islet cells, acinar cells, oocytes, sperm, haematopoietic cells, hepatocytes, skin/keratinocytes, melanocytes, bone/osteocytes, hair/dermal papilla cells, cartilage/chondrocytes, fats cells/adipocytes, skeletal muscular cells, endothelium cells, cardiac muscle/cardiomyocytes, and tropo
  • the committed cells are obtained from blood or related tissues including bone marrow.
  • the committed cells may be obtained from whole blood, and/or may be obtained through apheresis.
  • the blood may be mobilized or unmobilized blood.
  • Such committed cells include, but are not limited to, T cells, B cells, eosinophils, basophils, neutrophils, megakaryocytes, monocytes, erythrocytes, granulocytes, mast cells, lymphocytes, leukocytes, platelets, and red blood cells.
  • the committed cells may be obtained from neuronal tissue from the central nervous system or peripheral nervous system, muscle tissue, or epidermis and/or dermis tissue from skin.
  • the committed cells are obtained from the blood or tissue of a patient.
  • the patient from which the committed cells are obtained, and to which the reprogrammed target cells such as retrodifferentiated target cells or the transdifferentiated target cells are administered is the same patient.
  • the committed cells are retrodifferentiated by contacting the committed cells with an agent.
  • the committed cells may be incubated with the agent.
  • the agent engages a receptor that mediates capture, recognition or presentation of an antigen at the surface of the committed cells.
  • the receptor may be an MHC class I antigen or an MHC class II antigen.
  • the class I antigen is an HLA-A receptor, an F1LA-B receptor, an HLA-C receptor, an HLA-E receptor, an HLA-F receptor or an HLA-G receptor and said class II antigen is an HLA-DM receptor, an HLA-DP receptor, an HLA-DQ receptor or an HLA-DR receptor.
  • the agent may be an antibody to the receptor, such as a monoclonal antibody to the receptor.
  • the antibody is monoclonal antibody CR3/43 or monoclonal antibody TAL 1B5.
  • the agent modulates MHC gene expression, such as MHC class I + and/or MHC class II + expression.
  • the retrodifferentiated cells may undergo redifferentiation in a separate step.
  • the retrodifferentiated cells may be redifferentiated by contacting the retrodifferentiated cells with growth factors including, but not limited to, basic fibroblast growth factor , epidermal growth factor, granulocyte macrophage colony- stimulating factor, stem cell factor, interleukins-l, -3, -6, and -7, basic fibroblast growth factor, epidermal growth factor, granulocyte-macrophage colony stimulating factor, granulocyte-colony stimulating factor, erythropoietin, stem cell factor, and bone
  • growth factors including, but not limited to, basic fibroblast growth factor , epidermal growth factor, granulocyte macrophage colony- stimulating factor, stem cell factor, interleukins-l, -3, -6, and -7, basic fibroblast growth factor, epidermal growth factor, granulocyte-macrophage colony stimulating factor, granulocyte-colony stimulating factor,
  • the resulting redifferentiated target cells may then be administered to a patient.
  • the committed cells can be transdifferentiated by culturing the committed cells in particular culture conditions.
  • the committed cells may be cultured in particular types of culture media in conjunction with the retrodifferentiation agents. Examples of these culture media may include Dulbecco's
  • tissue culture media may also comprise differentiation promoting agent such as vitamin and/or mineral supplements, hydrocortisone, dexamethasone, ⁇ -mercaptoethanol, etc.
  • additional culturing conditions include using chelating agents or antibiotics, culturing at certain temperatures or carbon dioxide or oxygen levels, and culturing in certain vessels.
  • the culture conditions can determine the type of transdifferentiated target cell that results.
  • One aspect of the invention is thereby a method of obtaining target cells.
  • the method may comprise obtaining committed cells and then reprogramming the committed cells. These processes are as described in this application.
  • the method may comprise retrodifferentiating committed cells.
  • the method may comprise transdifferentiating committed cells.
  • the method may comprise retrodifferentiating committed cells, and then redifferentiating the retrodifferentiated cells .
  • Another aspect of the invention is use of one or more reprogrammed target cells in preparation of a medicament for repairing or replenishing tissue or cells of a cell lineage in a patient, or for treating a disease or tissue injury.
  • another aspect of the invention is a method of treating a disease or tissue injury in a patient in need thereof.
  • the method comprises obtaining committed cells, reprogramming the committed cells to obtain reprogrammed target cells, and administering reprogrammed target cells to the patient.
  • the target cells may be reprogrammed via retrodifferentiation, transdifferentiation, and/or ⁇ differentiation.
  • the target cells are retrodifferentiated target cells, transdifferentiated target cells, and/or redifferentiated target cells. The proceses of obtaining the committed cells and reprogramming the committed cells are as described in this application.
  • the reprogrammed target cells such as retrodifferentiated target cells, transdifferentiated target cells, or redifferentiated target cells
  • the reprogrammed target cells may be administered via injection, implantation, or infusion. These cells may be administered by parenteral, intramuscular, intravenous, subcutaneous, intraocular, oral, transdermal injection, or injection into spinal fluid.
  • the retrodifferentiated target cells or transdifferentiated target cells are administered in a pharmaceutical composition.
  • the pharmaceutical composition may comprise the retrodifferentiated target cells or
  • transdifferentiated target cells and at least one pharmaceutically acceptable excipient.
  • the pharmaceutical composition for administering the reprogrammed target cells, such as retrodifferentiated target cells or transdifferentiated target cells.
  • the pharmaceutical composition may comprise one or more types of target cells, and at least one pharmaceutically acceptable carrier.
  • the pharmaceutical composition may include adjuvants and/or other excipients suitable for administration into a patient.
  • Another aspect of the invention is a method of preparing a pharmaceutical composition or medicament comprising (i) obtaining committed cells, (ii) reprogramming the committed cells to obtain reprogrammed target cells, and (iii) optionally, combining the reprogrammed target cells with one or more pharmaceutical excipients.
  • the reprogrammed target cells are combined with one or more pharmaceutical excipients. In other embodiments, the reprogrammed target cells are not combined with one or more pharmaceutical excipients.
  • the processes of obtaining committed cells and reprogramming the committed cells are as described in this application.
  • FIG. 1 shows immunophenotyping of aphaeresed mononuclear cells before (top panel) and after (lower panel) induction of reprogramming.
  • Cells were labeled with monoclonal antibodies conjugated to R-phycoerythrin (RPE) Cy-5 or phycoerythrins (PE) (vertical legends) for immunoglobulin Gl (IgGl) isotype control and CD34 or CD19, respectively.
  • RPE R-phycoerythrin
  • PE phycoerythrins
  • IgGl immunoglobulin Gl
  • Cells were also stained for CD45, CD38, CD61 , and IgGl isotype controls monoclonal antibodies conjugated to fluorescein isothiocyanate (FITC) (horizontal legends).
  • Lower panel show increase in haematopoietic stem cells as depicted by in crease in the relative number of CD34 and CD34CD38- cells accompanied by decrease in leukocyte
  • FIG. 2a shows sequential immunophenotyping of peripheral blood samples of a patient with severe aplastic anemia following infusion of autologous HRSC (post Day 1, Day 2, Day 3, Day 6, and Day 14).
  • Cells were labeled with monoclonal antibodies against CD34 and CD45 (2 nd horizontal panel), and CD34 and CD38 (3 rd horizontal panel).
  • the top horizontal panel shows forward and side scatter is flow cytometry, bone marrow smear, and tehphin section of autologous HRSC.
  • Dayl to Day 14 shows an increase in cells having large forward and side scatter that is indicative of granulocytes
  • Dayl to Day3 shows an increase in the relative number of circulating CD34 haematopoietic stem cells.
  • FIG. 1 shows sequential immunophenotyping of peripheral blood samples of a patient with severe aplastic anemia following infusion of autologous HRSC (post Day 1, Day 2, Day 3, Day 6, and Day 14).
  • Cells were labeled with monoclonal antibodies against CD34 and CD
  • FIG. 2b shows sequential immunophenotyping of peripheral blood samples of a patient with severe aplastic anemia following infusion of autologous human reprogrammed stem cell (HRSC) (post Day 1, Day 2, Day 3, Day 6, and Day 14).
  • HRSC autologous human reprogrammed stem cell
  • Cells were labeled with monoclonal antibodies against CD34 and CD61 (1 st horizontal panel), CD19 and CD3 (2 nd panel), and CD33&13 and CD7 (3 rd horizontal panel).
  • the FACScan plots show increase in the number of myeloid cells, as depicted by a gradual increase in cells expressing CD33&13 including progenitor cells, which is depicted by an increase in the relative number of cell co- expressing CD33&13 with CD7. Also, there was a gradual increase in the relative number of lymphocytes, as shown by an increase in the relative number of CD 19 and CD3 lymphocytes.
  • FIG. 3 shows bone marrow analysis of a severe aplastic anaemia patient before and after infusion of autologous HRSC.
  • Bone marrow smear before and after therapy shows increase in red blood cells; terphine section before and after therapy (c and d) shows increase in bone marrow cellularity; clonal analysis of bone marrow following infusion of HRSC showing increased growth of colony forming unit megakarocyte (e); colony forming monocyte (f); colony forming granulocyte and macrophage (g); and colony forming myelocyte and erytliroid (h); and burst forming erythroid (i).
  • colony forming unit megakarocyte e
  • colony forming monocyte f
  • colony forming granulocyte and macrophage g
  • colony forming myelocyte and erytliroid h
  • burst forming erythroid i
  • FIG. 4 shows karyotyping and g-banding of peripheral blood sample of a patient with severe aplastic anemia following 4 years of infusion with autologous HRSC showing no genetic abnormalities.
  • FIG. 5 shows increase in absolute mean fetal hemoglobin levels in patients with thalassemia, post-treatment with autologous reprogrammed cells.
  • FIG. 6 shows increase in mean corpuscular volume, which represents the average size of red blood cells, expressed in femtoliters, in patients with thalassemia, post-treatment with autologous reprogrammed cells.
  • FIG. 7 shows increase in mean cell hemoglobin, which is the weight of the hemoglobin per cell, expressed in picograms, in patients with thalassemia, post-treatment with autologous reprogrammed cells.
  • FIG. 8 shows decrease in serum ferritin levels, expressed in nanograms per milliliter, in patients with thalassemia, post-treatment with autologous reprogrammed cells.
  • FIG. 9 shows increase in C-peptide levels fasting and post mixed meal diet intake in patients with diabetes, post-treatment with autologous reprogrammed cells.
  • FIG. 10 shows decrease in glycosylated hemoglobin (HbAlC) levels in patients with diabetes, post-treatment with autologous reprogrammed cells.
  • HbAlC glycosylated hemoglobin
  • FIG. 11 shows decrease in creatine phosphokinase (CPK) and lactate dehydrogenase (LDH) levels in patients with muscular dystrophy, post- treatment with autologous reprogrammed cells.
  • FIG. 12 shows decrease in liver enzymes alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels in patients with muscular dystrophy, post-treatment with autologous reprogrammed cells.
  • CPK creatine phosphokinase
  • LDH lactate dehydrogenase
  • FIG. 13 shows decrease in microalbumin urea levels in 12 patients with kidney disease, post-treatment with autologous reprogrammed cells.
  • FIG. 14 shows decrease in glycosylated hemoglobin (HbAlC) levels in 12 patients with kidney disease due to diabetes, post-treatment with autologous reprogrammed cells.
  • HbAlC glycosylated hemoglobin
  • FIG. 15a shows magnetic resonance imaging (MRI) scans of the brain of a patient with multiple sclerosis before (left scan) and three months after (right scan) treatment with autologous reprogrammed cells depicting decrease in lesion enhancement post stem cell therapy.
  • FIG. 15b shows MRI scans of a different part of the brain of the patient before (left scan) and three months after (right scan) treatment with autologous reprogrammed cells.
  • the arrows point to a lesion in the brain, while in the scans depicting after treatment, the arrows point to the improvement in the lesion.
  • FIG. 16a shows magnetic resonance imaging (MRI) scans of the brain of a patient with multiple sclerosis before (top scans) and six months after (bottom scans) treatment with autologous reprogrammed cells.
  • FIG. 16b shows additional MRI scans of the brain of the patient before (top scans) and six months after (bottom scans) treatment with autologous reprogrammed cells.
  • the arrows point to a lesion in the brain, while in the scans depicting after treatment, the arrows point to the improvement in the lesion with reduction in brain atrophy as depicted by reduction in ventricle and sulci dilatation.
  • FIG. 17a shows sagittal magnetic resonance imaging (MRI) scans of a patient with multiple sclerosis before (left scan) and six months after (right scan) treatment with autologous reprogrammed cells.
  • FIG. 17b shows transverse MRI scans of the spinal cord of the patient before (left scan) and six months after (right scan) treatment with autologous reprogrammed cells.
  • the arrows point to a lesion on the spinal cord, while in the scans depicting after treatment, the arrows point to the improvement in the lesion.
  • FIG. 18a shows decrease liver enzymes alanine aminotransferase (ALT) levels
  • FIG. 18b shows decrease in aspartate aminotransferase (AST) levels in patients infected with hepatitis C, post-treatment with autologous reprogrammed cells
  • FIG. 19a shows magnetic resonance imaging (MRI) scans of the brain of a patient with head trauma due to a motor accident. Before treatment (top scans), the ventricles show dilatation and displacement with a wide speared haematoma. After treatment with autologous reprogrammed cells (bottom scans), the ventricles show decrease in brain atrophy parameters such as reduction in ventricle and sulci dilatation with amelioration of haematoma.
  • FIG. 19b shows additional MRI scans of the brain of the patient before (top scans) and after (bottom scans) treatment.
  • FIG. 20 shows chest x-rays of a patient with lung disease before (left x-ray) and after (right x-ray) treatment with autologous reprogrammed cells. After treatment, the patient shows improvement in lung volume and reduction in lesion size as depicted by decrease in hypo-dense areas.
  • FIG. 21 shows sex hormone levels for follicle-stimulating hormone (fsh), luteinizing hormone (lh), progesterone (pro), and testosterone (test) in patients with non-obstructive azoospermia and treated with autologous reprogrammed cells.
  • the levels show significant increase in free testosterone with an increase in testis size (data not shown) determined by ultrasound.
  • FIG. 22 shows retinal sensitivity and visual impairment in a patient suffering from impaired vision before (top panel) and after (bottom panel) treatment with autologous reprogrammed cells.
  • the orange area indicates decreased retinal sensitivity.
  • the white areas indicate normal vision and the pink, orange, and black areas indicate increased visual impairment.
  • the patient experienced improvement in his visual field, as orange areas in the retinal sensitivity results before treatment turned green and white after treatment, and black areas in the visual impairment results before treatment turned white after treatment.
  • mitted cells are cells that display a differentiated character. These cells are often considered mature and specialized. Examples include white blood cells, red blood cells, epithelial cells, neurons, and chondrocytes.
  • uncommitted cells are cells that do not display a mature differentiated character. These cells are often considered immature and are not specialized.
  • An example of an uncommitted cell is a stem cell, which is an immature cell that is capable of self-renewal (division without limit) and differentiation (specialization).
  • reprogramming refers to a process by which a committed cell of a first cell lineage is changed into a cell of a different cell type. This different cell type may be of a different cell lineage. Reprogramming may occur through such processes as retrodifferentiation, transdifferentiation, or redifferentiation.
  • a "reprogrammed cell” is a cell that underwent reprogramming of a committed cell.
  • a reprogrammed cell may include a retrodifferentiated cell,
  • transdifferentiated cell and/or a redifferentiated cell.
  • “retrodifferentiation” is the process by which a committed cell, i.e., mature, specialized cell, reverts back to a more primitive cell stage.
  • “Retrodifferentiated cell” is a cell that results from retrodifferentiation of a committed cell.
  • transdifferentiation is the process by which a committed cell of a first cell lineage is changed into another cell of a different cell type.
  • transdifferentiation may be a combination of retrodifferentiation and redifferentiation.
  • Transdifferentiated cell is a cell that results from transdifferentiation of a committed cell.
  • a committed cell such as a whole blood cell may be transdifferentiated into a neuron.
  • redifferentiation refers to the process by which an uncommitted cell or a retrodifferentiated cell differentiates into a more mature, specialized cell.
  • Redifferentiated cell refers to a cell that results from redifferentiation of an uncommitted cell or a retrodifferentiated cell. If a redifferentiated cell is obtained through redifferentiation of a retrodifferentiated cell, the redifferentiated cell may be of the same or different lineage as the committed cell that had undergone retrodifferentiation. For example, a committed cell such as a white blood cell may be retrodifferentiated to form a
  • retrodifferentiated cell such as a pluripotent stem cell
  • the retrodifferentiated cell may be redifferentiated to form a lymphocyte, which is of the same lineage as the white blood cell (committed cell), or redifferentiated to form a neuron, which is of a different lineage than the white blood cell (committed cell).
  • target cell is a cell that is obtained for administration into a patient to repair or replenish tissue or cells.
  • a target cell may be a reprogrammed target cell, such as a retrodifferentiated target cell or a transdifferentiated target cell, whereby the retrodifferentiated or transdifferentiated target cell is administered to the patient.
  • committed cells of the invention are cells that display a differentiated character.
  • the committed cell may comprise any components that are concerned with antigen presentation, capture or recognition.
  • the committed cell may be an MHC Class I + and/or an MHC Class II + cell.
  • the committed cell may also be any cell derived or derivable from an
  • the committed cell is also an
  • the committed cell can be a lymphoid stem cell or a myeloid stem cell, which is differentiated relative to a pluripotent stem cell.
  • Committed cells may be derived from biological material, such as blood or related tissues including bone marrow or cord blood, neuronal tissue from the central nervous system or peripheral nervous system, muscle tissue, or epidermis and/or beis tissue from skin (i.e. by way of oral scraping for instance).
  • biological material may be of post-natal origin.
  • the biological material may be obtained using methods known in the art that are suitable for the tissue type. Examples include, but are not limited to, excision, needle withdrawal, swabbing, and apheresis.
  • the committed cells are derived from whole blood or processed products thereof, such as plasma or the buffy coat, since their removal from subjects can be carried out with the minimum of medical supervision.
  • Blood samples are typically treated with anticoagulants such as heparin or citrate.
  • Cells in the biological sample may be treated to enrich certain cell types, remove certain cell types or dissociate cells from a tissue mass.
  • Useful methods for purifying and separating cells include centrifugation (such as density gradient centrifugation), flow cytometry and affinity chromatography (such as the use of magnetic beads comprising monoclonal antibodies to cell surface markers or panning) (see Vettese-Dadey, The Principle 1999, 13: 21).
  • Ficoll-Hypaque separation is useful for removing erythrocytes and granulocytes to leave mononuclear cells such as lymphocytes and monocytes.
  • Examples of committed cells that can be derived from blood include, but are not limited to, CFC-T cells, CFC-B cells, CFC-Eosin cells, CFC-Bas cells, CFC-Bas cells, CFC-GM cells, CFC-M, CFC-MEG cells, BFC-E cells, CFC-E cells, T cells, B cells, eosinophils, basophils, neutrophils, monocytes, megakaryocytes, and erythrocytes.
  • Blood derived committed cells may be identified by their expression of particular antigens.
  • B cells are CD19 + , CD21 + , CD22 + and DR + cells.
  • T cells are CD2 + , CD3 + , and either CD4 + or CD8 + cells.
  • Immature lymphocytes are CD4 + and CD8 + cells.
  • Activated T cells are DR + cells.
  • Natural killer cells (NKs) are CD56 + and CD16 + cells.
  • T lymphocytes are CD7 cells.
  • Leukocytes are CD45 cells.
  • Granulocytes are CD13 + and CD33 + cells.
  • Monocyte macrophage cells are CD14 + and DR + cells.
  • the committed cell may be a B lymphocyte (activated or non-activated), a T lymphocyte (activated or non-activated), a cell from the macrophage monocyte lineage, a nucleated cell capable of expressing class I or class II antigens, a cell that can be induced to express class I or class II antigens or an enucleated cell (i.e. a cell that does not contain a nucleus—such as a red blood cell).
  • the committed cell may be selected from any one of a group of cells comprising large granular lymphocytes, null lymphocytes and natural killer cells, each expressing the CD56 and/or CD 16 cell surface receptors.
  • the committed cells are essentially primary cultures, it may necessary to supplement populations of cells with suitable nutrients to maintain viability. Suitable culture conditions are known by the skilled person in the art. Nonetheless, treatment of cell populations is preferably initiated as soon as possible after removal of biological samples from patients, typically within 12 hours, preferably within 2 to 4 hours. Cell viability can be checked using well known techniques such as trypan blue exclusion or propidium iodide.
  • Retrodifferentiation is a type of a reprogramming process whereby structures and functions of cells are progressively changed to give rise to less specialized cells.
  • Retrodifferentiation can occur naturally, wherein cells may undergo limited reverse differentiation in vivo in response to tissue damage. Alternatively, retrodifferentiation may be induced using the methods described in U.S. application Serial No. 08/594,164, now U.S.
  • Retrodifferentiated cells of the invention may include, but are not limited to pluripotent stem cells, lymphoid stem cells, myeloid stem cells, neural stem cells, skeletal muscle satellite cells, epithelial stem cells, endodermal stem cells, mesenchymal stem cells, and embryonic stem cells.
  • the committed cells are derived from blood and are retrodifferentiated to form retrodifferentiated cells of the haematopoietic cell lineage.
  • retrodifferentiated cells examples include, but are not limited to, pluripotent stem cells, lymphoid stem cells, and myeloid stem cells.
  • Committed cells may be retrodifferentiated by contacting the cells with an agent that operably engages the cells. The cells are then incubated so as to allow those cells that have been operably engaged by the agent to progress tlirough the retrodifferentiation process and ultimately become undifferentiated.
  • the contacting step may comprise the agent engaging with surface antigens on the committed cell.
  • the agent may act in direct engagement or in indirect engagement with the committed cell.
  • An example of direct engagement is when the committed cell has at least one cell surface receptor on its cell surface, such as a ⁇ -chain having homologous regions (regions that are commonly found having the same or a similar sequence) such as those that may be found on B cells, and wherein the agent directly engages the cell surface receptor.
  • Another example is when the committed cell has a cell surface receptor on its cell surface such as an a-chain having homologous regions such as those that may be found on T cells, and wherein the agent directly engages the cell surface receptor.
  • An example of indirect engagement is when the committed cell has at least two cell surface receptors on its cell surface and engagement of the agent with one of the receptors affects the other receptor which then induces retrodifferentiation of the committed cell.
  • the agent for the retrodifferentiating the committed cell may be a chemical compound or composition.
  • the agent may be capable of engaging a cell surface receptor on the surface of the committed cell.
  • the agent operably engages a receptor present on the surface of the committed cell - which receptor may be expressed by the committed cell, such as a receptor that is capable of being expressed by the committed cell.
  • agents may include, but are not limited to, any one or more of cyclic adenosine monophosphate (cAMP), a CD4 molecule, a CD8 molecule, a part or all of a T- cell receptor, a ligand (fixed or free), a peptide, a T-cell receptor (TCR), an antibody, a cross-reactive antibody, a monoclonal antibody, or a polyclonal antibody.
  • Growth factors may also be used, such as haematopoietic growth factors, for example erythropoietin and granulocyte-monocyte colony stimulating factor (GM-CSF).
  • the agent may be any one or more of an antibody, a cross-reactive antibody, a monoclonal antibody, or a polyclonal antibody to any one or more of: the .beta, chain of a MHC class II antigen, the ⁇ -chain of a MHC HLA-DR antigen, the a-chain of a MHC class I or class II antigen, the a-chain of HLA-DR antigen, the a- and the ⁇ -chain of MHC class II antigen or of a MHC class I antigen.
  • An example of an antibody is CR3/43 (supplied by Dako).
  • antibody may include the various fragments (whether derived by proteolytic cleavage or recombinant technology) and derivatives that retain binding activity, such as Fab, F(ab') 2 and scFv antibodies, as well as mimetics or bioisosteres thereof. Also included as antibodies are genetically engineered variants where some of the amino acid sequences have been modified, for example by replacement of amino acid residues to enhance binding or, where the antibodies have been made in a different species to the organism whose cells it is desired to treat according to the methods of the invention, to decrease the possibility of adverse immune reactions (an example of this is "humanized' mouse monoclonal antibodies).
  • Agents used to effect the conversion of a committed cell to a retrodifferentiated cell preferably may act extracellularly of the committed cell.
  • the committed cell may comprise a receptor that is operably engageable by the agent and the agent operably engages the receptor.
  • the receptor may be a cell surface receptor.
  • cell surface receptors include, but are not limited to, MHC class I and class II receptors.
  • the receptor may comprise an a-component and/or a ⁇ -component, as is the case for MHC class I and class II receptors.
  • the receptor may comprises a ⁇ -chain having homologous regions, for example at least the homologous regions of the ⁇ -chain of HLA-DR.
  • the receptor may comprise an a-chain having homologous regions, for example at least the homologous regions of the a-chain of HLA- DR.
  • the receptor may be a Class I or a Class II antigen of the major histocompatibility complex (MHC).
  • MHC major histocompatibility complex
  • the cell surface receptor may include, but are not limited to, an HLA-DR receptor, a DM receptor, a DP receptor, a DQ receptor, an HLA-A receptor, an HLA-B receptor, an HLA-C receptor, an HLA-E receptor, an HLA-F receptor, or an HLA-G receptor.
  • the cell surface receptor may be an HLA-DR receptor.
  • the agent may be an antibody to the receptor, such as a monoclonal antibody to the receptor.
  • An example of an agent may be one that modulates MHC gene expression such as MHC Class I + and/or MHC Class IF " expression.
  • the agent may be used in conjunction with a biological response modifier.
  • biological response modifiers include, but are not limited to, an alkylating agent, an immunomodulator, a growth factor, a cytokine, a cell surface receptor, a hormone, a nucleic acid, a nucleotide sequence, an antigen or a peptide.
  • an alkylating agent may be or may comprise cyclophosphoamide.
  • Other biological response modifiers may include compounds capable of upregulating
  • MHC class I and/or class II antigen expression which, in some embodiments, may allow an agent that binds to an MHC receptor to work more effectively.
  • any cell type can be made to express MHC class I and/or class II antigens, this may provide a method for retrodifferentiating a wide variety of cell types whether they constitutively express class I and/or class II MHC antigens or not.
  • Committed cells are generally incubated with an agent for at least two hours, typically between 2 and 24 hours, preferably between 2 and 12 hours. Incubations are typically performed at from about room temperature or for example about 22° C, up to about 37° C, including 33° C.
  • the progress of the retrodifferentiating procedure can be checked periodically by removing a small aliquot of the sample and examining cells using microscopy and/or flow cytometry.
  • the device can comprise tracking means for on-line monitoring the progress of the retrodifferentiating procedure.
  • the committed cells may be cultured in autologous plasma or serum, or in fetal blood serum or horse serum.
  • the committed cells may be cultured with anticoagulants, chelating agents, or antibiotics.
  • the temperature range for incubating the cells may be extended to 18-40 °C, and may also include 4-10 % C0 2 and/or 10-35 % 0 2 .
  • incubation may occur in blood bags, tissue culture bags, or plastic vessels, which are coated or uncoated.
  • retrodifferentiated cells may be obtained by retrodifferentiating using particular culture conditions.
  • committed cells may be retrodifferentiated into pluripotent cells by culturing the committed cells in Dulbecco's Modified Eagle Medium (DMEM), non-essential amino acids (NEAA), L-glutamine (L-glu), and ⁇ - mercaptoethanol (2 ⁇ ), in conjunction with the retrodifferentiating agents.
  • DMEM Dulbecco's Modified Eagle Medium
  • NEAA non-essential amino acids
  • L-glu L-glutamine
  • ⁇ - mercaptoethanol (2 ⁇ ) ⁇ -mercaptoethanol
  • committed cells may be cultured - in conjunction with retrodifferentiating agent(s) - using DMEM (low glucose) and L-glu, or DMEM (low glucose), L-glu, 2 ⁇ , and NEAA.
  • DMEM low glucose
  • L-glu low glucose
  • DMEM low glucose
  • NEAA low glucose
  • the antibiotic gentamycin may also be used in the cell culture.
  • Transdifferentiated cells are obtained by culturing committed cells with a tissue culture media in conjunction with retrodifferentiating agents.
  • the committed cells thereby undergo transdifferentiation, wherein the committed cells are transformed to cells of another cell type; in some embodiments, the committed cells are transformed to cells of a different lineage.
  • the type of target cell obtained through transdifferentiation is dependent on the culturing conditions. These conditions vary according to the type of tissue culture media, the presence/absence of various differentiation promoting agents, the presence/absence of different serums, the incubation temperature, the presence/absence of oxygen or carbon dioxide, and the type of container or vessel used for incubation.
  • tissue culture media used for transdifferentiation include, but are not limited to, Iscove's Modified Dulbecco's Medium (IMDM), Dulbecco's Modified Eagle Medium (DMEM), Eagle's minimum essential (EME) medium, alpha- minimum essential medium ( ⁇ - ⁇ ), Roswell Park Memorial Institute (RPMI; site where medium was developed) 1640, Ham-F-12, El 99, MCDB, Leibovitz L-15, Williams Medium E, or any commercially formulated tissue culture medium.
  • IMDM Iscove's Modified Dulbecco's Medium
  • DMEM Dulbecco's Modified Eagle Medium
  • EME Eagle's minimum essential
  • RPMI Roswell Park Memorial Institute
  • Differentiation promoting agents include anticoagulants, chelating agents, and antibiotics.
  • agents may be one or more of the following: vitamins and minerals or derivatives thereof, such as A (retinol), B 3 , C (ascorbate), ascorbate 2- phosphate, D 2 , D 3 , K, retinoic acid, nicotinamide, zinc or zinc compound, and calcium or calcium compounds; natural or synthetic hormones such as hydrocortisone, and
  • dexamethasone amino acids or derivatives thereof, such as L-glutamine (L-glu), ethylene glycol tetracetic acid (EGTA), proline, and non-essential amino acids (NEAA); compounds or derivatives thereof, such as ⁇ -mercaptoethal, dibutyl cyclic adenosine monophosphate (db-cAMP), monothioglycerol (MTG), putrescine, dimethyl sulfoxide (DMSO),
  • amino acids or derivatives thereof such as L-glutamine (L-glu), ethylene glycol tetracetic acid (EGTA), proline, and non-essential amino acids (NEAA); compounds or derivatives thereof, such as ⁇ -mercaptoethal, dibutyl cyclic adenosine monophosphate (db-cAMP), monothioglycerol (MTG), putrescine, dimethyl sulfoxide (DMSO),
  • db-cAMP dibutyl cyclic adeno
  • hypoxanthine adenine, forskolin, cilostamide, and 3-isobutyl-l-methylxanthine
  • nucleosides and analogues thereof such as 5-azacytidine
  • acids or salts thereof such as ascorbic acid, pyruvate, okadic acid, linoleic acid, ethylenediaminetetraacetic acid (EDTA), anticoagulant citrate dextrose formula A (ACDA), disodium EDTA, sodium butyrate, and
  • glycerophosphate glycerophosphate
  • antibiotics or drugs such as G418, gentamycine, Pentoxifylline (l-(5- oxohexyl)-3, 7-dimethylxanthine), and indomethacin
  • proteins such as tissue plasminogen activator (TP A).
  • differentiation promoting agents may be used to obtain particular types of target cells.
  • vitamin B 3 may be used to yield acinar cells such as islet cells or hydrocortisone; dexamethasone may be used to yield cells of mesenchymal origin or epithelial origins (e.g. kidney epithelial cells, skin and associated structures such as dermal papilla cells); and ⁇ -mercaptoethal may be used to yield ectodermal cells such as neuronal cells, including accessory cells of the CNS.
  • the culture medium may contain autologous plasma; platelets; serum such as fetal blood sampling; or sera of mammalian origin such as horse serum. Furthermore, the conversion process can occur inside blood bags, scaffolds, tissue culture bags, or plastic tissue culture vessels.
  • the tissue culture vessels may be adherent or non-adherent tissue culture vessels, or may be coated or uncoated with agents such as gelatin, collagen, matrigels or extracellular matrices that either promote adherence or floatation depending on the required type of tissue or specialized cells to be prepared.
  • Additional culturing conditions include the temperature, which may be between about 10 and about 60 °C, or between about 18 and about 40 °C; levels of carbon dioxide (C0 2 ), which may be between about 0 and about 20 %, or about 4 and about 10 %; and oxygen (0 2 ), which may be between about 0 and about 50 %, or about 10 and about 35 %.
  • C0 2 carbon dioxide
  • oxygen (0 2 ) oxygen
  • Medium 199 can be used as a basal medium instead
  • hydrocortisone optionally with
  • hydrocortisone hydrocorbate
  • Haematopoietic e IMDM optionally with • Incubation can be at 33 °C cells hydrocortisone; or e Incubation can be at room
  • Culture conditions for • differentiated cells can be pluripotent stem cells listed exposed to chelating agents prior above, with MTG substituted for to conversion in order to amplify 2 ⁇ , optionally with vitamins for erythroid progenitors in culture
  • RPMI 1640 can be used as a basal medium for enrichment of lymphoid progenitors
  • Sodium butyrate and/or 5- azacytidine can be added to culture to promote primitive erythroid differentiation
  • liver essential medium L-glu
  • hydrocortisone optionally with hydrocortisone
  • dexamethasone, L-glutamine and • Can be with low serum sodium pyruvate; or concentration.
  • DMEM and Ham F12 or F10 as • Can be with culture vessels with basal medium, or DMEM and gelatin
  • NEAA NEAA
  • DMEM low glucose
  • the culture medium optionally containing the various components
  • differentiation promoting agents may be diluted by the addition of more medium but without the retrodifferentiating agent.
  • dilution seems to increase differentiation because cells become less dense and proliferation stimulating factors are less concentrated.
  • the addition of medium may further enhance transdifferentiation and will affect what type of cell within the cell lineage is obtained. For instance, if the target cell is a neuron, the addition of culture medium may result in a shift in development towards a more mature neuron rather than a neuron progenitor (both are of the same lineage).
  • forward differentiation skeletal muscle progenitors will only differentiate by consecutive dilution of culture medium which is achieved by gradually decreasing the serum concentration.
  • the retrodifferentiated cells may be used to obtain target cells by recommitting or redifferentiating the retrodifferentiated cells to a target cell type. This may be performed by contacting the retrodifferentiated cells with growth factors. For example, retinoic acid has been used to differentiate stem cells into neuronal cells. Methylcellulose followed by co- culture with a bone marrow stromal line and IL-7 has been used to differentiate stem cells into lymphocyte precursors (Nisitani et al., Int Immuno 1994, 6: 909-916). Le Page (New Scientist Dec. 16, 2000) teaches that stem cells can be differentiated into lung epithelial cells.
  • Neural precursor cells can be expanded with basic fibroblast growth factor and epidermal growth factor (Nakafuku and Nakamura, J Neurosci Res 1995, 41 : 153-168).
  • Haematopoietic stem cells can be expanded using a number of growth factors including GM-CSF, erythropoietin, stem cell factor and interleukins (IL-1 , IL-3, IL-6)— see Metcalf (Nature 1989, 339: 27-30) for a review of these various factors.
  • the redifferentiated cell may be of the same lineage as the committed cell from which the retrodifferentiated cell was derived.
  • the redifferentiated cell may be of a different lineage than the committed cell from which the retrodifferentiated cell was derived.
  • a B lymphocyte may be retrodifferentiated to a CD34 + CD38 " HLA- DR " stem cell. This stem cell may be subsequently redifferentiated or recommitted along a B cell lineage (the same lineage) or a lymphoid lineage (different lineage).
  • target cells of the invention are reprograrnmed cells that can be obtained by retrodifferentiation, transdifferentiation, or redifferentiation as described above.
  • target cells may include, but are not limited to, pluripotent stem cells, lymphoid stem cells, myeloid stem cells, neural stem cells, skeletal muscle satellite cells, epithelial stem cells, endodermal and neuroectodermal stem cells, germ cells,
  • extraembryonic and embryonic stem cells mesenchymal stem cells, , kidney cells, alveolar epithelium cells, endoderm cells, neurons, ectoderm cells, islet cells, acinar cells, oocytes, sperm, haematopoietic cells, hepatocytes, skin/keratinocytes, melanocytes, bone/osteocytes, hair/dermal papilla cells, cartilage/chondrocytes, fats cells/adipocytes, skeletal muscular cells, endothelium cells, cardiac muscle/cardiomyocytes, and tropoblasts.
  • committed cells and/or retrodifferentiated cells are cultured under particular conditions to induce retrodifferentiation and/or transdifferentiation and/or redifferentiation and obtain the target cells.
  • the duration for which the committed cells and/or retrodifferentiated cells are cultured is not controlled by a particular length of time, but rather by a determination that the target cells have been produced. The determination of the production of, or the changes in the number of,
  • retrodifferentiated, transdifferentiated, or redifferentiated target cells may be performed by monitoring changes in the relative number of committed cells that downregulates expression of lineage-associated markers or transcription factors, and/or changes in the relative number of cells having cell surface markers characteristic of the target cells. Alternatively, or in addition, decreases in the numbers of cells having cell surface markers typical of the committed cells and not the target cells may be monitored.
  • the target cell may be an embryonic stem cell, which are characterized by many stage-specific markers such as POU5F1 (OCT-4), TERT, KLF4, UTFl , SOX2, Nanog or stage-specific embryonic markers 3 and 4 (SSEA-3 and SSEA-4), high molecular weight glycoproteins TRA- 1 -60 and TRA- 1 - 81 and alkaline phosphatase (Andrews et al., Hybridoma 1984, 3: 347-361 ; Kannagi et al., EMBO J 1983, 2: 2355-2361 ; Fox et al., Dev Biol 1984, 103: 263-266; Ozawa et al., Cell Differ 1985, 16: 169-173).
  • stage-specific markers such as POU5F1 (OCT-4), TERT, KLF4, UTFl , SOX2, Nanog or stage-specific embryonic markers 3 and 4 (SSEA-3 and SSEA-4), high molecular weight glycoproteins TRA- 1
  • SSEA-1 SSEA-1
  • Other markers are known for other types of stem cells, such as Nestein for neuroepithelial stem cells (J Neurosci 985, 5: 3310).
  • Mesenchymal stem cells are positive for SH2, SH3, CD29, CD44, CD71, CD90, CD106, CD120a and CD124, for example, and negative for CD34, CD45 and CD14.
  • Pluripotent stem cells are CD34 + DR " TdT " cells (other useful markers being CD38 " and CD36 + ).
  • Lymphoid stem cells are DR + , CD34 + and TdT + cells (also CD38 + ).
  • Myeloid stem cells are CD34 + , DR + , CD13 + , CD33 + , CD7 + and TdT + cells.
  • Additional cell markers for target cells may be discovered through microarray analysis.
  • the analysis may involve isolating RNA from target cells that were
  • the microarray may comprise genes or oligonucleotides representing a whole genome, or may comprise genes or oligonucleotides directed to a particular organ system, tissue system, disease, pathology, etc.
  • Cell markers may be identified by which genes/oligonucleotides exhibit high signal intensity, and are thereby upregulated or downregulated in the target cells. This information can then be applied to determining target cells based on the presence of markers, or even groups or a partem of markers, that were identified by the microarray analysis.
  • Confirmation of target cells can also be performed using a number of in vitro assays such as CFC assays (see also, the examples).
  • Very primitive haematopoietic stem cells are often measured using the long-term culture initiating cell (LTC-IC) assay (Eaves et al, J Tiss Cult Meth 1991, 13: 55-62). LTC-ICs sustain haemopoiesis for 5 to 12 weeks.
  • LTC-IC long-term culture initiating cell
  • cell culturing may continue until the target cells emerge as characterized by immunohistochemistry, flow cytometry, microarrays, or reverse-transcription polymerase chain reaction (RT-PCR), which are techniques known in the art.
  • RT-PCR reverse-transcription polymerase chain reaction
  • This may also include functional assays, for example, engrafting an immunodeficient host or correcting or ameliorating an underlying clinical condition, as observed herein.
  • the target cells can be identified by microarray or RT-PCR that show acquisition of new lineage-specific transcription factors, proteins, and signals in the target cells.
  • retrodifferentiated stem cells converted to target cells of the ectodermal lineage may express genes such as Nestin, Criptol, isll, LHX1, and/or EN1, and if further differentiated into neurons, will be expressing neurofilaments (NF).
  • cells converted into target cells of the endodermal lineage may express genes such as sox7, soxl 7, Nodal, PDX1, and/or FOXA2; but target cells further differentiated towards pancreatic islet cells may express genes such as insulin (INS) and neurog3 (NGN3).
  • INS insulin
  • NTN3 neurog3
  • the conversion towards the desired target cells may be accompanied by down regulation of mature transcription factors associated with the original starting population that has undergone conversion.
  • the determination of the production of target cells may occur by recognizing particular structural and/or morphological characteristics of the target cells, e.g., cell shape, size, etc. These characteristics are known in the art for the target cells of the invention.
  • the resulting altered cell populations may be used in a number of ways.
  • target cells e.g., pluripotent stem cells
  • the numbers of target cells or other retrodifferentiated cells formed may appear to be low, studies have shown that only 50 pluripotent haematopoietic stem cells can reconstitute an entire haematopoietic system in a donor mouse. Thus therapeutic utility does not require the formation of a large number of cells.
  • Conversion of committed cells to retrodifferentiated, transdifferentiated, or redifferentiated target cells may also be carried out in vivo by administration of the agent, admixed with a pharmaceutically carrier or diluent, to a patient. However it is preferred in many cases that retrodifferentiating, transdifferentiating, or redifferentiating is performed in vitro lex vivo.
  • Treated populations of cells obtained in vitro may be used subsequently with minimal processing. For example they may be simply combined with a pharmaceutically acceptable carrier or diluent and administered to a patient in need of stem cells.
  • the retrodifferentiated, transdifferentiated, or redifferentiated target cells may be desirable to enrich the cell population for the retrodifferentiated, transdifferentiated, or redifferentiated target cells or purify the cells from the cell population.
  • This can conveniently be performed using a number of methods (see Vattese-Dadey ⁇ The Engineer 1999, 13).
  • cells may be purified on the basis of cell surface markers using chromatography and/or flow cytometry. Nonetheless, it will often be neither necessary nor desirable to extensively purify retrodifferentiated, transdifferentiated, or redifferentiated target cells from the cell population since other cells present in the population (for example stromal cells) may maintain stem cell viability and function.
  • the purification or isolation means may comprise a flow cytometer.
  • Flow cytometry operates on the basis of physical characteristics of particles in liquid suspension, which can be distinguished when interrogated with a beam of light. Such particles may of course be cells. Physical characteristics include cell size and structure or, as has become very popular in recent years, cell surface markers bound by monoclonal antibodies conjugated to fluorescent molecules.
  • Visser et al. (Blood Cells 1980, 6:391-407) teach that stem cells may be isolated on the basis of their size and degree of structuredness.
  • Grogan et al. (Blood Cells 1980, 6: 625-44) also teach that "viable stem cells may be sorted from simple haematopoietic tissues in high and verifiable purity".
  • cell populations may be enriched, purified using negative criteria.
  • cells that possess lineage specific markers such as CD4, CD8, CD42 and CD3 may be removed from the cell population by flow cytometry or affinity chromatography.
  • a very useful technique for purifying cells involves the use of antibodies or other affinity ligands linked to magnetic beads.
  • the beads are incubated with the cell population and cells that have a cell surface marker, such as CD34, to which the affinity ligand binds are captured.
  • the sample tube containing the cells is placed in a magnetic sample concentrator where the beads are attracted to the sides of the tube. After one or more wash stages, the cells of interest have been partially or substantially completely purified from other cells.
  • the liquid phase is kept and consequently, the cells bound to the beads are effectively removed from the cell population.
  • Cell populations comprising reprogrammed target cells such as retrodifferentiated, transdifferentiated, or redifferentiated target cells, and/or purified reprogrammed target cells, such as retrodifferentiated, transdifferentiated, or redifferentiated target cells produced by the methods of the invention, may be maintained in vitro using known techniques.
  • minimal growth media such as Hanks, RPMI 1640, Dulbecco's Minimal Essential Media (DMEM) or Iscove's Modified Dulbecco Medium
  • mammalian serum such as FBS
  • autologous plasma mammalian serum
  • Stem cells may be cultured on feeder layers such as layers of stromal cells (see Deryugina et al. Crit Rev Immunology 1993, 13: 1 15-150). Stromal cells are believed to secrete factors that maintain progenitor cells in an undifferentiated state.
  • a long term culture system for stem cells is described by Dexter et al. (J Cell Physiol 1977, 91 : 335) and Dexter et al. (Acta Haematol 1979, 62: 299).
  • Lebkowski et al. (Transplantation 1992, 53: 1011-9) teaches that human CD34 + haematopoietic cells can be purified using a technology based on the use of monoclonal antibodies that are covalently immobilized on polystyrene surfaces and that the CD34 + cells purified by this process can be maintained with greater than 85% viability.
  • Lebkowski et al. (J Hematother 1993, 2: 339-42) also teaches how to isolate and culture human CD34 + cells. See also Haylock et al. (Immunomethods 1994, 5: 217-25) for a review of various methods. Cell populations comprising stem cells and purified preparations comprising stem cells may be frozen/cryopreserved for future use. Suitable techniques for freezing cells and subsequently reviving them are known in the art.
  • the retrodifferentiating, transdifferentiating, or ⁇ differentiating occurs to cells from or in buffy coat blood samples.
  • buffy coat means the layer of white cells that forms between the layer of red cells and the plasma when unclotted blood is centrifuged or allowed to stand.
  • Reprogrammed target cells of the present invention may be combined with various components to produce compositions of the invention.
  • the compositions may be combined with one or more pharmaceutically acceptable carriers or diluents to produce a pharmaceutical composition (which may be for human or animal use).
  • Suitable carriers and diluents include, but are not limited to, isotonic saline solutions, for example phosphate- buffered saline.
  • the composition of the invention may be administered by direct injection.
  • the composition may be formulated for parenteral, intramuscular, intravenous,
  • Compositions comprising target cells may be delivered by injection or implantation.
  • Cells may be delivered in suspension or embedded in a support matrix such as natural and/or synthetic biodegradable matrices.
  • Natural matrices include, but are not limited to, collagen matrices.
  • Synthetic biodegradable matrices include, but are not limited to, polyanhydrides and polylactic acid. These matrices may provide support for fragile cells in vivo.
  • compositions may also comprise the retrodifferentiated or transdifferentiated or redifferentiated target cells of the present invention, and at least one pharmaceutically acceptable excipient, carrier, or vehicle.
  • Delivery may also be by controlled delivery, i.e., delivered over a period of time which may be from several minutes to several hours or days. Delivery may be systemic (for example by intravenous injection) or directed to a particular site of interest. Cells may be introduced in vivo using liposomal transfer.
  • Target cells may be administered in doses of from 1 x 10 5 to 1 x 10 7 cells per kg.
  • a 70 kg patient may be administered 14 x 10 6 CD34 + cells for reconstitution of tissues.
  • the dosages may be any combination of the target cells listed in this application.
  • the methods of the invention can be used to treat a variety of diseases, conditions, or disorders. Such conditions include, but are not limited to, bone marrow failure,
  • haematological conditions aplastic anemia, beta-thalassemia, diabetes, motor neuron disease, Parkinson's disease, spinal cord injury, muscular dystrophy, kidney disease, liver disease, multiple sclerosis, congestive heart failure, hepatitis C virus, human
  • azoospermia andropause, menopause and infertility, rejuvenation, scleroderma ulcers, psoriasis, wrinkles, liver cirrhosis, autoimmune disease, alopecia, retinitis pigmentosa, and crystalline dystrophy/blindness or any disorder associated with tissue degeneration.
  • Aplastic anemia is a rare but fatal bone marrow disorder, marked by pancytopaenia and hypocellular bone marrow (Young et al. Blood 2006, 108: 2509-2519).
  • the disorder may be caused by an immune-mediated pathophysiology with activated type I cytotoxic T cells expressing Thl cytokine, especially ⁇ -interferon targeted towards the haematopoietic stem cell compartment, leading to bone marrow failure and hence anhaematoposis
  • graft failure winch may ensue weeks or months after stem cell transplantation (Gottdiener et al., Arch Intern Med 1981, 141 : 758- 763; Sanders et al. Semin Hematol 1991, 28: 244-249). Moreover the risk of graft failure increases with the number of blood transfusion received prior stem cell transplantation.
  • Thalassaemia is an inherited autosomal recessive blood disease marked by a reduced synthesis rate of one of the globin chains that make up hemoglobin. Thus, there is an underproduction of normal globin proteins, often due to mutations in regulatory genes, which results in formation of abnormal hemoglobin molecules, causing anemia.
  • Different types of thalassemia include alpha thalassemia, beta thalassemia, and delta thalassemia, which affect production of the alpha globin, beta globin, and delta globin, respectively.
  • Treatments include chronic blood transfusion, iron chelation, splenectomy, and allogeneic haematopoietic transplantation. However, chronic blood transfusion is not available to most patients due to the lack of an HLA-matched bone marrow donor, while allogeneic hematopoietic transplantation is associated with many possible complications such as infections and graft-versus-host disease.
  • Diabetes is a syndrome resulting in abnormally high blood sugar levels
  • Diabetes refers to a group of diseases that lead to high blood glucose levels due to defects in either insulin secretion or insulin action in the body. Diabetes is typically separated into two types: type 1 diabetes, marked by a diminished production of insulin, or type 2 diabetes, marked by a resistance to the effects of insulin. Both types lead to hyperglycemia, which largely causes the symptoms generally associated with diabetes, e.g., excessive urine production, resulting compensatory thirst and increased fluid intake, blurred vision, unexplained weight loss, lethargy, and changes in energy metabolism.
  • Diabetes is considered to be a chronic disease, without a cure.
  • Treatment options are limited to insulin injections, exercise, proper diet, or, for patients who have type 2 diabetes, some medications, e.g., those that promote insulin secretion by the pancreas, decrease glucose produced by the liver, increase sensitivity of cells to insulin, etc.
  • Motor neuron diseases refer to a group of neurological disorders that affect motor neurons. Such diseases include amyotrophic lateral sclerosis (ALS), primary lateral sclerosis (PLS), and progressive muscular atrophy (PMA). ALS is marked by degeneration of both the upper and lower motor neurons, which ceases messages to the muscles and results in their weakening and eventual atrophy. PLS is a rare motor neuron disease affecting upper motor neurons only, which causes difficulties with balance, weakness and stiffness in legs, spasticity, and speech problems. PMA is a subtype of ALS that affects only the lower motor neurons, which can cause muscular atrophy, fasciculations, and weakness. There are no known cures for motor neuron diseases.
  • ALS amyotrophic lateral sclerosis
  • PLS primary lateral sclerosis
  • PMA progressive muscular atrophy
  • Riluzole which is believed to reduce damage to motor neurons, has been has approved as a medication for ALS, although it slows down the progression of ALS rather than improves its effects.
  • treatments only address symptoms, such as baclofen which can reduce spasticity or quinine which may decrease cramps.
  • Parkinson's disease is a neurodegenerative disorder marked by the loss of the nigrostriatal pathway, resulting from degeneration of dopaminergic neurons within the substantia nigra.
  • the cause of PD is not known, but is associated with the progressive death of dopaminergic (tyrosine hydroxylase (TH) positive) mesencephalic neurons, inducing motor impairment.
  • TH dopaminergic
  • PD is characterized by muscle rigidity, tremor, bradykinesia, and potentially akinesia.
  • Symptomatic treatment of the disease-associated motor impairments involves oral administration of dihydroxyphenylalanine (L-DOPA), which can lead to a substantial improvement of motor function, but its effects are reduced as the degeneration of dopaminergic neurons progresses.
  • Alternative strategies include neural grafting, which is based on the idea that dopamine supplied from cells implanted into the striatum can substitute for lost nigrostriatal cells, and gene therapy, which can be used to replace dopamine in the affected striatum by introducing the enzymes responsible for L-DOPA or dopamine synthesis such as by introducing potential neuroprotective molecules that may either prevent the TH-positive neurons from dying or stimulate regeneration and functional recovery in the damaged nigrostriatal system.
  • Spinal cord injury is characterized by damage to the spinal cord and, in particular, the nerve fibers, resulting in impairment of part or all muscles or nerves below the injury site. Such damage may occur through trauma to the spine that fractures, dislocates, crushes, or compresses one or more of the vertebrae, or through nontraumatic injuries caused by arthritis, cancer, inflammation, or disk degeneration. While treatments following spinal cord injury may involve medications such as methylprednisolone, which is a corticosteroid that reduce damage to nerve sells and decreases inflammation in the injured area, or medications that control pain and muscle spasticity, as well as immobilization of the spine or surgery to remove herniated disks or any objects that may be damaging the spine, there is no known means to reverse the damage to the spinal cord.
  • methylprednisolone which is a corticosteroid that reduce damage to nerve sells and decreases inflammation in the injured area, or medications that control pain and muscle spasticity, as well as immobilization of the spine or surgery to remove herniated
  • MD Muscular dystrophy
  • Kidney disease refers to conditions that damage the kidneys and decrease their ability to function, which includes removal of wastes and excess water fro the blood, regulation of electrolytes, blood pressure, acid-base balance, and reabsorption of glucose and amino acids.
  • the two main causes of kidney disease are diabetes and high blood pressure, although other causes include glomerulonephritis, lupus, and malformations and obstructions in the kidney.
  • There is no cure for kidney disease, and thereby therapy focuses on slowing the progression of the disease and treating the causes of the disease, such as through controlling blood glucose and high blood pressure and monitoring diet; treating complications of the disease, for example, by addressing fluid retention, anemia, bone disease; and replacing lost kidney function, such as through dialysis or transplantation.
  • MS multiple sclerosis is an autoimmune condition in which the immune system attacks the central nervous system, leading to demyelination.
  • MS affects the ability of nerve cells in the brain and spinal cord to communicate with each other, as the body's own immune system attacks and damages the myelin which enwraps the neuron axons. When myelin is lost, the axons can no longer effectively conduct signals. This can lead to various neurological symptoms which usually progresses into physical and cognitive disability.
  • treatments attempt to return function after an attack (sudden onset or worsening of MS symptoms), prevent new attacks, and prevent disability. For example, treatment with corticosteroids may help end the attack, while treatment with interferon during an initial attack has been shown to decrease the chance that clinical MS will develop.
  • HIV Human immunodeficiency virus
  • AIDS acquired immunodeficiency syndrome
  • HIV primarily infects vital cells in the human immune system such as helper T cells, macrophages, and dendritic cells. HIV infection leads to low levels of CD4 + T cells by direct viral killing of infected cells, by increased rates of apoptosis in infected cells, or by killing of infected CD4 + T cells by CD8 cytotoxic lymphocytes that recognize infected cells.
  • Treatment for HIV infection consists of highly active antiretroviral therapy, or HAART.
  • NARTIs nucleoside analogue reverse transcriptase inhibitors
  • RTI non- nucleoside reverse transcriptase inhibitor
  • Congestive heart failure refers to a condition in which the heart cannot pump enough blood to the body's other organs. This condition can result from coronary artery disease, scar tissue on the heart cause by myocardial infarction, high blood pressure, heart valve disease, heart defects, and heart valve infection.
  • Treatment programs typically consist of rest, proper diet, modified daily activities, and drugs such as angiotensin-converting enzyme (ACE) inhibitors, beta blockers, digitalis, diuretics, vasodilators. However, the treatment program will not reverse the damage or condition of the heart.
  • ACE angiotensin-converting enzyme
  • Hepatitis C is an infectious disease in the liver, caused by hepatitis C virus. Hepatitis C can progress to scarring (fibrosis) and advanced scarring (cirrhosis). Cirrhosis can lead to liver failure and other complications such as liver cancer.
  • Current treatments include use of a combination of pegylated interferon alpha and the antiviral drug ribavirin. Success rates can vary between 50-80% depending on the virus genotype.
  • Head trauma refers to an injury of the head that may or may not cause injury to the brain.
  • head trauma includes traffic accidents, home and occupational accidents, falls, and assaults.
  • Various types of problems may result from head trauma, including skull fracture, lacerations of the scalp, subdural hematoma (bleeding below the dura mater), epidural hematoma (bleeding between the dura mater and the skull), cerebral contusion (brain bruise), concussion (temporary loss of function due to trauma), coma, or even death.
  • Treatment for head trauma will vary with the type of injury. If the brain is damaged, there is no quick means to fix it, and often the damage may be irreversible by available treatment means.
  • Lung disease is a broad term for diseases of the respiratory system, which includes the lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, and nerves and muscles for breathing.
  • lung diseases include obstructive lung diseases, in which the bronchial tubes become narrowed; restrictive or fibrotic lung diseases, in which the lung loses compliance and causes incomplete lung expansion and increased lung stiffness; respiratory tract infections, which can be caused by the common cold or pneumonia; respiratory tumors, such as those caused by cancer; pleural cavity diseases; and pulmonary vascular diseases, which affect pulmonary circulation.
  • Treatment for lung disease varies according to the type of disease, but can include medication such as corticosteroids and antibiotics, oxygen, mechanical ventilation, radiotherapy, and surgery.
  • Depression is a mental disorder characterized by a low mood accompanied by low self-esteem and loss of interest or pleasure in normally enjoyable activities.
  • Biologically depression is accompanied by altered activity in multiple parts of the brain, including raphe nuclei, which are a group of small nuclei in the upper brain stem that is a source of serotonin; suprachiasmatic nucleus, which controls biological rhythms such as the sleep/wake cycle; hypothalamic-pituitary-adrenal axis, which is a chain of structures that are activated during the body's response to various stressors; ventral tegmental area, which is considered to be responsible for the "reward" circuitry of the brain; nucleus accumbens, which us thought to play a role in reward, laughter, pleasure, addiction, and fear; and the anterior cingulated cortex, which is activated by negative experiences.
  • Treatments for depression include antidepressants that increase the amount of extracellular serotonin in the brain, exercise, and psychotherapy.
  • Non-obstructive azoospermia is a medical condition of a male not having any measureable level of sperm in his semen due to a problem with spermatogenesis. This is often caused by hormonal imbalance and can be treated using medications which restore the imbalance.
  • Andropause is a menopause- like condition experienced in middle-aged men, which involves a reduction in the production of the hormones testosterone and
  • Treatments include hormone replacement therapy and exercise.
  • Scleroderma is a chronic autoimmune disease that affects comiective tissue.
  • Hardening of the skin is the most visible manifestation of the disease, although it can affect connective tissue throughout the body. There is no known direct cure for scleroderma.
  • Psoriasis is a chronic autoimmune disease that causes red, scaly patches to appear on the skin. The cause is psoriasis is linked to the excessive growth of skin cells.
  • T-cells which migrate to the dermis and trigger the release of cytokines that induce the rapid production of skin cells.
  • Treatments for psoriasis include drugs that target the T-cells.
  • Retinitis pigmentosa is type of progressive retinal dystrophy in which the photoreceptors or the retinal pigment epithelium is abnormal and leads to visual loss.
  • the conditions described herein may be treated with a particular type, or a combination of types, of target cells.
  • the condition described herein may be treated by infusion of the cell types outlined in Table 2. Table 2.
  • Treatment regimes of various conditions using reprogrammed, i.e., retrodifferentiated or transdifferentiated or redifferentiated, target cells Autoimmune Disease mesenchymal stem cells and/or pluripotent stem cells
  • a patient may be treated for a condition as described above through the following steps:
  • Fistula-cannula is inserted in a patient's arm
  • White blood cells are harvested through aphaeresis using an automated system, such as the COBE ® Spectra Device (Gambro PCT);
  • the autologous retrodifferentiated stem cells are washed and then infused intravenously into the patient;
  • Patient's progress is monitored, including taking blood tests and assessing the injured areas.
  • This clinical study assessed the safety of infusing a single dose of autologous 3 hr reprogrammed cells following exposure to haematopoietic inductive culture condition into four patients with aplastic anemia.
  • the patients were transfused with 2 units of irradiated packed red blood cells and 4 units of platelets to maintain their hemoglobin level above 8 g/dl and platelets counts above 50,000.
  • Patient were apheresed by processing 2-3 times their total blood volume using the Cobe Spectra apheresis machine and the white blood cells separation kit (both from Gambro BCT). Apheresis involved jugular and anticubetal venous catheterization with single lumen catheter for venous access.
  • Table 4 Clinical and treatment history of aplastic anemia patients up to Autologous human reprogrammed stem cell (HRSC) infusion.
  • HRSC Autologous human reprogrammed stem cell
  • Table 5 Patient, date of infusion, weight and height, mononuclear cells collect and CD34+ cell infused.
  • Panel 1 consisted of Isotype negative control IgGl-FITC, IgGl-PE-Cy5 and IgGl- PvPE conjugates
  • Panel 2 consisted of anti-human CD45-FITC and CD34-RPE-Cy5
  • Panel 3 consisted of anti-human CD38-FITC and CD34-RPE-Cy5
  • Panel 4 consisted of CD61-FITC and CD34-RPE-Cy5
  • Panel 5 consisted of CD33/13 RPE and CD7-FITC
  • Panel 6 consisted of CD45 and Glycophorin-A-RPE
  • Panel 7 consisted of CD3-FITC and CD19-RPE
  • bone marrow mononuclear cells (M C) of patient prior and post infusion of the reprogrammed cells were seeded into methocult GFH4434 supplemented with recombinant growth factors according to the manufacturer's instructions (Stem Cell Technologies). Differentiation into haematopoietic cell colonies was assessed and scored with time using phase contrast inverted microscopy.
  • CBC liver enzymes and haemoglobin variants were continuously monitored before and post procedure. Following release from hospital patients, CBC, liver enzymes, hemoglobin variants, and peripheral blood karyotyping and G banding were monitored by an independent laboratory for reconfirmation purposes. These tests were performed frequently following infusion of the autologous reprogrammed cells.
  • Peripheral blood samples and bone marrow cells were analyzed before and following infusion of the autologous reprogrammed cells. This test was repeated in six-month intervals for the first year and on a yearly basis following 2 years post-initiation of autologous reprogrammed stem cell therapy. In addition, reprogrammed cells were analyzed prior infusion to look into the stability of the cells, which was also performed following the 3hr conversion step, as well as post-establislunent of a maximum 1 month long term culture of the converted cells. Karyotyping and G banding were monitored by a third independent laboratory.
  • Bone marrow smears and trephine section was performed before and post infusion of the autologous reprogrammed cells. This test was performed 14-20 days post infusion of the autologous reprogrammed cells and thereafter on a yearly basis. All smear and trephine sections were scanned using a microscope hooked to a camcorder before and following infusion of the reprogrammed stem cells to assess and keep record of engraftment.
  • Liver enzymes started to normalize post infusion of RHSC and reached normal levels 4 years post infusion of RHSC.
  • Patient B and Patient C died 2 years and six months post infusion, respectively.
  • Fetal Hb switching was noted in patients 001 and 004 (see Tables 6 and 7). These two patients exhibited long term engraftment post single infusion of the autologous HRSC.
  • WBC white blood cell
  • HB hemoglobin
  • RBC red blood cell
  • RETIC reticulocyte
  • MCV mean corpuscular volume
  • MCH mean corpuscular hemoglobin
  • MCHC mean corpuscular hemoglobin concentration
  • ESO eosinophils
  • BASO basophils
  • LYMPH lymphocytes
  • MONO monocytes
  • HB A hemoglobin A
  • HBA2 hemoglobin A2; HBF
  • SGOT serum glutamic oxaloacetic transaminase
  • Table 7 Patient D's complete blood counts, hemoglobin variant and liver enzymes of a severe aplastic anemia patient before and after in fusion of autologous HRSC.
  • MCV mean corpuscular volume
  • MCH mean corpuscular hemoglobin
  • MCHC mean corpuscular hemoglobin concentration
  • ESO eosinophils
  • BASO basophils
  • LYMPH lymphocytes
  • MONO monocytes
  • HB A hemoglobin A
  • HBA2 hemoglobin A2
  • HBF fetal hemoglobin
  • SGOT serum glutamic oxaloacetic transaminase
  • SGPT serum glutamic pyruvic transaminase
  • FIG. 1 Flow cytometry of aphaeresed mononuclear cells before and 3 hrs post induction of haematopoietic reprogramming are shown in FIG. 1.
  • the number of CD34 positive cell generated post haematopoietic reprogramming is listed in Table 4.
  • a representative flow cytometry of apheresed mononuclear before and post haematopoietic reprogramming show significant increase in the number of CD34 positive cells with and without expression of CD45, CD38 and CD7 (FIG. 1).
  • the CD34 cells circulated peripheral blood for 3-6 days and thereafter differentiated at a sustainable level into myelocyte as depicted by significant increase in cells expressing CD33&13 with and without CD7 having high forward and side scatter (see FIG. 2). Tins pattern of reprogramming was observed in all patients.
  • Hb F haemoglobin level (Tables 6 and 7). This was not noted in the other 2 patients that died. Hb F switching in these two patients confirms the reprogramming capabilities of the infused H SC towards juvenile Hb phenotype and hence engraftment and reconstitution as observed with cord blood stem cells transplant (Elhasid et al., Leukemia 2000, 14: 931-934; Locatelli et al, Bone Marrow Transplant 1996, 18: 1095-101).
  • the autologous HRSC were capable of long term engraftment and survival rate in a subset of severe aplastic anaemia patients without use of any
  • reprogrammed cells have exhibited a normal karyotype and genetic stability after infusion. Finally, engraftment and long term repopulation was observed in 3 more patient suffering from aplastic anemia.
  • beta thalassemia Autologous reprogrammed haematopoietic cells (target cells) were tested in 21 patients with beta thalassemia. Nineteen patients had beta-thalassemia major and 2 had thalassemia intermedia. One of the beta thalassemia intermedia patient was thalassemia/Hb E variant (common in patient of far eastern and Indian origin) and the other had
  • the autologous reprogrammed cells were generated through reprogramming white blood cells until the target cells were obtained, as indicated by their distinguishing characteristics as described above.
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • the mean weight and height following infusion of the reprogrammed cells were significantly greater when compared to base line, and the organ size in thalassemic patients with enlarged spleen and/or liver was normalized.
  • the absolute mean fetal haemoglobin concentration significantly increased in patients with thalassemia major and intermedia following infusion of the reprogrammed cells when compared to baseline (FIG. 5).
  • the mean red blood cell indices as reflected by improvement in red blood cell size haemoglobin content (FIG. 6) and concentration (FIG. 7) was also significantly improved as compared to baseline.
  • iron overload is the major cause of mortality and morbidity in patients with thalassemia; sickle cell anaemia and any transfusional iron overload induced disorder.
  • Autologous reprogrammed mesenchymal stem cells, pluripotent stem cells, and islet cells (target cells) were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • ALS amyotrophic lateral sclerosis
  • Autologous reprogrammed pluripotent stem cells, alveolar epithelium cells, and neurons (target cells) were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • PFT Pulmonary Function test
  • Autologous reprogrammed pluripotent stem cells and neurons were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above. .
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • Autologous retrodifferentiated pluripotent stem cells and neurons were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the patients were administered the autologous reprogi'ammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • MD Muscular Dystrophy
  • target cells were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the first patient was inflicted with limb-girdle MD, which refers to a class of MD wherein the muscles that are most severely affected generally those of the hips and shoulders, and the second patient was inflicted with nemelin MD.
  • the autologous reprogrammed cells were generated through reprogramming in accordance to the invention.
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • the muscle atrophy associated with MD can be measured by monitoring the level of the muscle enzyme creatine phosphokinase (CPK). This enzyme decreased in response to infusion of the retrodifferentiated stem cells (FIG. 11). Lactate dehydrogenase, an enzyme which is elevated during tissue breakdown, also decreased (FIG. 11). Patients also experienced a decrease in liver enzymes alanine aminotransferase (ALT) and aspartate aminotransferase (AST), which are both associated with inflammation and injury to liver cells, as well as skeletal muscle.
  • CPK creatine phosphokinase
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • patients showed an improvement in patient mobility as determined by cam recording of patients before and following infusion of the reprogrammed cells, and in pulmonary function test following infusion of the reprogrammed cells when compared to baseline
  • kidney disease Patients with kidney disease were treated with autologous reprogrammed pluripotent stem cells, mesenchymal stem cells, and kidney cells (target cells) were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the patients were apheresed by processing 2-3 times their total blood volume.
  • the autologous reprogrammed cells were generated through reprogramming in accordance to the invention.
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • kidney function such as urine output, serum creatinine and BUN or UREA.
  • fluid marker levels indicative of healthier kidney function
  • a 75 -year old diabetic male patient showed improved kidney function 24 months after treatment with the autologous reprogrammed cells ⁇ see Table 8).
  • the patient showed increased levels in hemoglobin, which is the protein molecule in red blood cells that carries oxygen, and insulin-like growth factor- 1 (IGF-1), a growth factor.
  • IGF-1 insulin-like growth factor- 1
  • urea which is an organic compound
  • creatinine which is a breakdown product of creatine phosphate in muscle
  • uric acid which is an organic compound excreted in urine
  • phosphorus which is a mineral found in bone
  • HbAlC hemoglobin
  • a 51 -year old diabetic male exhibited similar improvement 12 months after treatment ⁇ see Table 8).
  • This patient showed increased levels of hemoglobin and decreased levels of creatinine, HbAlC, and blood urea nitrogen (BUN), which is a measurement of the amount of nitrogen in the blood in the form of urea.
  • BUN blood urea nitrogen
  • Table 8 Kidney function marker levels in two diabetic male patients.
  • HbAlC glycosylated hemoglobin
  • IGF-1 Insulin-like Growth Factor 1
  • BUN blood urea nitrogen
  • FIG. 13 Analysis of 12 patients having Type II diabetes and treated with autologous reprogrammed cells revealed decreased levels of microalbumin (FIG. 13), which is associated with leakage of albumin into the urine and is an indicator of kidney disease, well as vascular endothelial dysfunction and cardiovascular disease. The 12 patients also experienced decreased levels of HbAlC (FIG. 14).
  • Table 9 Kidney function marker levels in 45-year old female patient suffering from autoimmune glomerulonephritis.
  • BUN blood urea nitrogen
  • Kidney function marker levels in 45-year old female patient suffering from autoimmune The patient also decreased in the number of hemodialysis sessions that she attended per month, from 12 sessions to about 8 sessions.
  • Table 10 Kidney function marker levels in 59-year old female patient suffering from end stage renal disease before and after therapy with reprogrammed cells.
  • TLC total leukocyte count
  • WBC white blood cells
  • PLT platelet
  • HbAlC glycosylated hemoglobin
  • AST aspartate aminotransferase
  • ALT alanine
  • PT pro-thrombin time
  • IN international normalized ratio(for blood coagulation)
  • PTH parathyroid hormone
  • a 46-year old patient suffering from chronic renal failure due to diabetes also showed improvement in creatinine level, urea, and hemoglobin level upon treatment with reprogrammed cells ⁇ see Table 11).
  • the patient decreased in the number of hemodialysis sessions that he attended per month, from 12 sessions to about 5 sessions, and decreased in treatment with epoetin alfa (EPREX ® ) delivered subcutaneously, from 4000 units twice a week to 4000 units once every two weeks.
  • EPREX ® epoetin alfa
  • TLC total leukocyte count
  • WBC white blood cells
  • PP post prandial
  • PLT platelet
  • FBS fasting blood sugar
  • AST aspartate aminotransferase
  • ALT alanine
  • Patients with multiple sclerosis were treated with autologous reprogrammed pluripotent stem cells, mesenchymal stem cells, and neurons (target cells).
  • the target cells were through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the patients were apheresed by processing 2-3 times their total blood volume.
  • the autologous reprogrammed cells were generated through reprogramming in accordance to the invention.
  • the patients were administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • Patients treated with autologous reprogrammed cells showed reduction in lesions in the brain and in the spinal cord. A reduction in lesions can occur within three months of receiving the treatment (FIGS. 15a-b). A reduction in damage to brain tissue occurred within six months (FIGS. 16a-b).
  • the patient was apheresed by processing 2-3 times her total blood volume.
  • the target cells were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above.
  • the patient was administered the autologous reprogrammed cells via intravenous infusion into the jugular or veins in the arm or thigh.
  • the screening test for HIV-1 and HIV-2 antibodies showed a test value of 3.68. A value of 1.0 or greater is considered positive.
  • test value for the HIV-1 and HIV-2 antibody screening was 0.46, which indicated that the patient did not show a positive result for HIV-1 and HIV-2.
  • test value for the HIV-1 and HIV-2 screening was 0.48, further demonstrating that the patient was not showing a positive result for HIV.
  • the effects of treating with autologous reprogrammed cells are demonstrated in patients suffering from other conditions and diseases.
  • the target cells listed herein were obtained through reprogramming apheresed white blood cells until the target cells developed, as indicated by their distinguishing characteristics as described above
  • EF ejection fraction
  • Pro BNP brain natriuretic peptide precursor levels
  • the heart which was previously dilated, returned to normal size as evidenced in Table 12 by decrease in end-diastolic left ventricular internal dimension (LVID/D), end-diastolic left ventricular internal dimension (LVID/S), and end-diastolic intraventricular septal thickness (IVSD) as measured by echocardiogram.
  • LVID/D end-diastolic left ventricular internal dimension
  • LVID/S end-diastolic left ventricular internal dimension
  • IVSD end-diastolic intraventricular septal thickness
  • LVID/D end-diastolic left ventricular internal dimension
  • LVID/S end systolic left ventricular internal dimension
  • EF ejection fraction
  • IVSD end-diastolic intraventricular septal thickness
  • LVPWD end-diastolic left ventricular posterior wall thickness
  • Pro-BNP precursor of brain natriuretic peptide
  • HbAlC glycosylated hemoglobin
  • HCV hepatitis C virus
  • AST aspartate aminotransferase
  • WBC white blood cells
  • PP post prandial
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • INR international normalized ratio (for blood coagulation)
  • PT prothrombin time
  • a 44-year old male patient suffering from liver cirrhosis due to hepatitis C virus infection also showed improvement in liver enzymes alanine aminotransferase and aspartate aminotransferase, the international normalized ratio (for blood coagulation), bilirubin, and albumin, as well as random blood sugar ⁇ see Table 15).
  • this patient was on albumin treatment before the therapy with reprogrammed cells, but did not receive albumin after the therapy.
  • Table 15 Blood markers in a 44-year old male patient suffering from liver cirrhosis due to hepatitis C virus infection at baseline and after therapy with reprogrammed cells.
  • TLC total leukocyte count
  • PLT platelet
  • AST aspartate aminotransferase
  • GGT gamma-glutamyl transferase
  • RBS random blood
  • INR international normalized ratio (for blood coagulation) Head Trauma
  • a patient suffering head trauma from a motor accident was treated with an infusion of autologous reprogrammed pluripotent stem cells and neurons.
  • a patient suffering from restrictive lung disease associated with a motor neuron disease was treated with an infusion of autologous reprogrammed pluripotent stem cells, mesenchymal stem cells, alveolar epithelium cells, and endothelium cells.
  • forced vital capacity FVC
  • FEVi forced expiratory volume in 1 second
  • the ratio of FEVi to FVC which is approximately 75-80 % in healthy adults, decreased from 100 % to 82 %.
  • An x-ray scan of the lungs shows less opacity of the lung cavity after treatment (FIG. 20).
  • a 51 -year old patient in menopause was administered an infusion of autologous reprogrammed pluripotent stem cells, pluripotent germ cells, and oocytes. Following the treatment, the patient experienced an increase in various hormone and protein levels, including insulin-like growth factor (IGF-1), esterdiaol, and low-density lipoprotein (LDL) (see Table 16). Table 16. Effects of administering patient in menopause with autologous reprogrammed stem cells.
  • IGF-1 insulin-like growth factor
  • LDL low-density lipoprotein
  • a patient suffering from depression was treated with an infusion of autologous reprogrammed pluripotent stem cells and neurons. Following the treatment, the patient experienced an increase in various hormone and protein levels, including insulin-like growth factor- 1 (IGF-1), Cortisol, and testosterone (see Table 17). Table 17. Effects of administering patient in menopause with autologous reprogrammed cells.
  • IGF-1 insulin-like growth factor- 1
  • Cortisol cortisol
  • testosterone see Table 17
  • GH growth hormone
  • IGF-1 insulin- like growth factor 1
  • IGF-bp insulin-like growth factor binding protein
  • Adrenocort adrenocortical hormone
  • Sh-bg sex hormone-binding globulin
  • FSH follicle- stimulating hormone
  • LH luteinizing hormone
  • e2 estradiol
  • DHEA-S dehydroepiandrosterone sulfate ester
  • TSH thyroid stimulating hormone
  • FT3 free triiodothryronine
  • FT4 free thyroxine
  • a patient suffering from non-obstructive azoospermia was treated with an infusion of autologous reprogrammed pluripotent stem cells, pluripotent germ cells, and sperm.
  • a patient suffering from vision loss due to a benign tumor that had been removed was treated with an infusion of autologous reprogrammed pluripotent stem cells and neurons. Following treatment, the patient experienced increased retinal sensitivity and improvement in vision (FIG. 22).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Neurosurgery (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Cardiology (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Communicable Diseases (AREA)
EP09736651A 2009-10-19 2009-10-19 Treatment using reprogrammed mature adult cells Withdrawn EP2490702A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/GB2009/051396 WO2011048350A1 (en) 2009-10-19 2009-10-19 Treatment using reprogrammed mature adult cells

Publications (1)

Publication Number Publication Date
EP2490702A1 true EP2490702A1 (en) 2012-08-29

Family

ID=42199514

Family Applications (1)

Application Number Title Priority Date Filing Date
EP09736651A Withdrawn EP2490702A1 (en) 2009-10-19 2009-10-19 Treatment using reprogrammed mature adult cells

Country Status (16)

Country Link
EP (1) EP2490702A1 (es)
JP (1) JP6073135B2 (es)
KR (1) KR101766203B1 (es)
CN (1) CN102647989A (es)
AU (1) AU2009354350B2 (es)
CA (1) CA2781522A1 (es)
CH (1) CH704227B1 (es)
CU (1) CU24006B1 (es)
IL (1) IL218902A (es)
MA (1) MA33740B1 (es)
MX (1) MX2012004522A (es)
RU (1) RU2635317C2 (es)
TN (1) TN2012000157A1 (es)
UA (1) UA109265C2 (es)
WO (1) WO2011048350A1 (es)
ZA (1) ZA201202664B (es)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3194571A4 (en) * 2014-09-18 2018-04-18 North Carolina State University Mammalian lung spheroids and lung spheroid cells and uses thereof
JP6113133B2 (ja) 2014-11-06 2017-04-12 日本メナード化粧品株式会社 幹細胞の未分化状態維持剤及び増殖促進剤
EP4265633A3 (en) 2015-10-16 2024-01-24 The Trustees Of Columbia University In The City Of New York Compositions and methods for inhibition of lineage specific antigens
CN107114355A (zh) * 2016-08-01 2017-09-01 北京世纪劲得生物技术有限公司 一种脂肪细胞保护液及其制备方法
CN110662554A (zh) 2017-02-28 2020-01-07 Vor生物制药股份有限公司 用于抑制谱系特异性蛋白质的组合物和方法
CN109731012A (zh) * 2017-10-30 2019-05-10 邹兆中 提高生物免疫治疗有效物质活性的方法
CN109706119B (zh) * 2018-04-13 2020-11-27 诺未科技(北京)有限公司 扩增造血干细胞的培养体系、方法及其用途
SG11202101994XA (en) 2018-08-28 2021-03-30 Vor Biopharma Inc Genetically engineered hematopoietic stem cells and uses thereof
KR20200025210A (ko) * 2018-08-29 2020-03-10 주식회사 스템모어 모유두 세포의 배양용 조성물 및 이를 이용한 모유두 세포의 배양 방법
WO2021240204A1 (en) * 2020-05-23 2021-12-02 Pakravan Nafiseh Sperm head for resolution of inflammation, tissue repair & regeneration, and restoration of tissue function: new approach of cell therapy
CN114231481B (zh) * 2021-12-21 2023-07-18 中国人民解放军总医院 一种重编程真皮成纤维细胞为内皮祖细胞的化学诱导方法
WO2023217132A1 (zh) * 2022-05-10 2023-11-16 上海赛立维生物科技有限公司 胆囊前体样细胞的制备方法和应用
WO2024050517A1 (en) * 2022-09-02 2024-03-07 The University Of North Carolina At Chapel Hill Methods of making and using anticancer reprogrammed b cells

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5554512A (en) * 1993-05-24 1996-09-10 Immunex Corporation Ligands for flt3 receptors
US7112440B2 (en) * 1995-02-02 2006-09-26 Ghazi Jaswinder Dhoot Method of increasing the relative number of CD45 low cells in a cell population
GB9502022D0 (en) * 1995-02-02 1995-03-22 Abuljadayel Ilham M S A method for preparing lymphohaematopoietic progenitor cells
US9068164B1 (en) * 1995-02-02 2015-06-30 Tristem Trading (Cyprus) Limited Method of preparing an undifferentiated cell
KR100404276B1 (ko) * 1995-05-02 2004-03-20 코닌클리케 필립스 일렉트로닉스 엔.브이. 칼러음극선관장치
US20050037490A1 (en) * 1999-10-29 2005-02-17 Lawrence Rosenberg Medium for preparing dedifferentiated cells
TWI288779B (en) * 2002-03-28 2007-10-21 Blasticon Biotech Forschung Dedifferentiated, programmable stem cells of monocytic origin, and their production and use
US20080112937A1 (en) * 2004-07-22 2008-05-15 Lewis T Williams Method of Producing Autologous Embryonic Stem Cells
KR20110134939A (ko) * 2009-04-03 2011-12-15 더 맥클린 하스피털 코퍼레이션 유도된 다능성 줄기 세포

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None *
See also references of WO2011048350A1 *

Also Published As

Publication number Publication date
IL218902A (en) 2017-06-29
CU20120063A7 (es) 2012-10-15
CU24006B1 (es) 2014-06-27
IL218902A0 (en) 2012-06-28
CA2781522A1 (en) 2011-04-28
KR101766203B1 (ko) 2017-08-08
JP6073135B2 (ja) 2017-02-01
AU2009354350B2 (en) 2015-09-03
AU2009354350A1 (en) 2012-04-19
RU2635317C2 (ru) 2017-11-10
MA33740B1 (fr) 2012-11-01
CH704227B1 (fr) 2015-05-15
CN102647989A (zh) 2012-08-22
MX2012004522A (es) 2012-05-08
UA109265C2 (uk) 2015-08-10
KR20120099686A (ko) 2012-09-11
RU2012120713A (ru) 2013-11-27
JP2013508343A (ja) 2013-03-07
WO2011048350A1 (en) 2011-04-28
ZA201202664B (en) 2013-02-27
TN2012000157A1 (en) 2013-12-12

Similar Documents

Publication Publication Date Title
AU2009354350B2 (en) Treatment using reprogrammed mature adult cells
US20140234268A1 (en) Treatment using reprogrammed mature adult cells
JP5636415B2 (ja) 幹細胞及び前駆細胞の分化のモジュレーション、アッセイ、並びにそれらの使用
US8828375B2 (en) Expansion of haemopoietic precursors
CN108478599A (zh) 间充质基质细胞及其相关用途
JP5431146B2 (ja) 免疫学的寛容および調節性前駆細胞
JP2002502617A (ja) 幹細胞および前駆細胞の増殖と分化を制御する方法
US20170106021A1 (en) Compositions enriched for hox11+ stem cells and methods of preparing the same
CN107641617B (zh) 体外高效制备非人灵长类动物巨核细胞及血小板的体系及其应用
US20080206196A1 (en) Differentiation of cord blood into neural like cells, and method to treat neurological condition patients
GB2474492A (en) Treatment of medical conditions using reprogrammed cells
JP2017008049A (ja) 再プログラム化成熟成体細胞を用いる治療
TW201114895A (en) Treatment using reprogrammed mature adult cells
CN114901806A (zh) 细胞群以及其取得方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120503

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1175111

Country of ref document: HK

17Q First examination report despatched

Effective date: 20140804

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20181027