US20130195817A1 - Methods and compositions for modulating angiogenesis and vasculogenesis - Google Patents

Methods and compositions for modulating angiogenesis and vasculogenesis Download PDF

Info

Publication number
US20130195817A1
US20130195817A1 US13/750,772 US201313750772A US2013195817A1 US 20130195817 A1 US20130195817 A1 US 20130195817A1 US 201313750772 A US201313750772 A US 201313750772A US 2013195817 A1 US2013195817 A1 US 2013195817A1
Authority
US
United States
Prior art keywords
cells
growth factor
subject
angiogenic
pro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/750,772
Other languages
English (en)
Inventor
Monique Dao
Ciara Tate
Casey Case
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.)
Sanbio Inc
Original Assignee
Sanbio Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanbio Inc filed Critical Sanbio Inc
Priority to US13/750,772 priority Critical patent/US20130195817A1/en
Assigned to SANBIO, INC. reassignment SANBIO, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CASE, CASEY, DAO, Monique, TATE, CIARA
Publication of US20130195817A1 publication Critical patent/US20130195817A1/en
Priority to US15/063,290 priority patent/US20160263159A1/en
Priority to US15/608,656 priority patent/US10245286B2/en
Priority to US16/269,000 priority patent/US11304982B2/en
Priority to US17/654,778 priority patent/US20220193144A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K35/34Muscles; 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
    • A61K35/28Bone 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
    • A61K35/44Vessels; Vascular smooth muscle cells; Endothelial cells; Endothelial progenitor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • A61L27/3834Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem cells
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0697Artificial constructs associating cells of different lineages, e.g. tissue equivalents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/42Notch; Delta; Jagged; Serrate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • C12N2502/1347Smooth muscle cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/28Vascular endothelial cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the present disclosure is in the fields of angiogenesis and vasculogenesis; e.g., for the treatment of ischemic events such as stroke. It is also in the field of stem cells and cells derived from stem cells by genetic manipulation.
  • the first step is angiogenic sprouting of endothelial cells (ECs); this process entails the initial proliferation of endothelial cells and remodeling of the surrounding extracellular matrix.
  • VEGF-mediated proliferation of ECs and matrix metalloproteinases are among the major components of angiogenic sprouting.
  • the second step is vessel stabilization; a process that relies on recruitment of vascular smooth muscle cells to encase the young vessels.
  • Monocytes and pericytes are also involved in vessel stabilization, producing the appropriate arteriogenic factors and extracellular matrix proteins. In the absence of vessel stabilization by smooth muscle cells and pericytes, regression of nascent vasculature can occur.
  • Marrow stromal cells (MSCs, also known as mesenchymal stem cells) have been shown to promote revascularization after cerebral artery occlusion and traumatic brain in jury.
  • MSCs also known as mesenchymal stem cells
  • SB623 cells are a derivative of marrow stromal cells, obtained by transfecting marrow stromal cells with a vector containing sequences encoding a Notch intracellular domain (NICD). See, for example, U.S.
  • SB623 cells elicit functional improvement in experimental stroke models. See, for example, U.S. Pat. No. 8,092,792 and Yasuhara et al. (2009) Stem Cells and Development 18:1501-1514.
  • the secretome of SB623 cells is comparable to that of the parental MSCs; different levels of specific trophic factors have been observed to be secreted by MSCs, as compared to SB623 cells. See, for example, Tate et al. (2010) Cell Transplantation 19:973-984; U.S. Patent Application Publication No. 2010/0266554.
  • many of the factors whose expression levels differ between MSCs and SB623 cells have been reported to be involved in vascular regeneration.
  • the present inventors have discovered that descendents of mesenchymal stem cells that have been transfected with sequences encoding a Notch intracellular domain (i.e., SB623 cells) have the surprising property of being able to synthesize and secrete factors that promote angiogenesis. Because angiogenesis, i.e., the formation of new blood vessels, is a critical part of the endogenous repair process in brain injury and disease, this discovery provides new methods of treatment for vascular disorders such as stroke.
  • the present disclosure provides, inter alia:
  • a method for augmenting angiogenesis in a subject comprising administering to the subject a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • a method for repairing ischemic damage in a subject comprising administering to the subject a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • a method for enhancing survival of endothelial cells comprising contacting the endothelial cells with a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • a method for stimulating proliferation of endothelial cells comprising contacting the endothelial cells with a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • a method for enhancing the branching of blood vessels comprising contacting the vessels with a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • a method for augmenting angiogenesis in a subject comprising administering to the subject (1) a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection; and (2) a pro-angiogenic agent.
  • NBD Notch intracellular domain
  • VEGF vascular endothelial growth factor
  • a method for repairing ischemic damage in a subject comprising administering to the subject (1) a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection; and (2) a pro-angiogenic agent.
  • NBD Notch intracellular domain
  • polypeptide is a transcription factor that activates expression of a pro-angiogenic protein.
  • VEGF vascular endothelial growth factor
  • a method for treating stroke in a subject comprising administering to the subject (1) a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection; and (2) a pro-angiogenic agent.
  • NBD Notch intracellular domain
  • polypeptide is a transcription factor that activates expression of a pro-angiogenic protein.
  • VEGF vascular endothelial growth factor
  • polypeptide is a transcription factor that activates expression of a pro-angiogenic protein.
  • VEGF vascular endothelial growth factor
  • polypeptide is a transcription factor that activates expression of a pro-angiogenic protein.
  • VEGF vascular endothelial growth factor
  • a method for providing an angiogenic factor to a subject comprises administering to the subject a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • the trophic factor is selected from the group consisting of one or more of angiogenin, angiopoietin-2, epidermal growth factor, basic fibroblast growth factor, heparin-binding epithelial growth factor-like growth factor, hepatocyte growth factor, leptin, platelet-derived growth factor-BB, placental growth factor and vascular endothelial growth factor.
  • a method for providing vascular endothelial growth factor to a subject comprises administering to the subject a population of SB623 cells; wherein the SB623 cells are obtained by (a) providing a culture of mesenchymal stem cells, (b) contacting the cell culture of step (a) with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein, (c) selecting cells that comprise the polynucleotide of step (b), and (d) further culturing the selected cells of step (c) in the absence of selection.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD) wherein said polynucleotide does not encode a full-length Notch protein
  • FIG. 1 shows measurements of the fraction of cells permeable to propidium iodide in cultures of HUVECs that have been starved for serum and growth factors.
  • Left-most bar shows results obtained from control serum/growth factor-starved HUVECs; center bar shows results for serum/growth factor-starved HUVECs cultured for seven days in the presence of conditioned medium from MSCs, and the right-most bar shows results for serum/growth factor-starved HUVECs cultured for seven days in the presence of conditioned medium from SB623 cells.
  • Values shown are mean ⁇ SD for three separate donors of MSCs and SB623 cells; * indicates p ⁇ 0.05 compared to control group.
  • FIG. 2 shows measurement of the fraction of cells expressing Bcl-2 in cultures of HUVECs that have been starved for serum and growth factors.
  • Left-most bar shows results obtained from control serum/growth factor-starved HUVECs; center bar shows results for serum/growth factor-starved HUVECs cultured for seven days in the presence of conditioned medium from MSCs, and the right-most bar shows results for serum/growth factor-starved HUVECs cultured for seven days in the presence of conditioned medium from SB623 cells.
  • Results were obtained by measuring fluorescence of cells stained with a fluorescein-conjugated anti-Bcl-2 antibody and subtracting fluorescence of cells exposed to fluorescein-conjugated IgG. Values shown are mean ⁇ SD for three separate donors of MSCs and SB623 cells; * indicates p ⁇ 0.05 compared to control group.
  • FIG. 3 shows measurement of the fraction of cells expressing Ki67 in cultures of HUVECs that have been starved for serum and growth factors.
  • Left-most bar shows results obtained from control serum/growth factor-starved HUVECs; center bar shows results for serum/growth factor-starved HUVECs cultured for seven days in the presence of conditioned medium from MSCs, and the right-most bar shows results for serum/growth factor-starved HUVECs cultured for seven days in the presence of conditioned medium from SB623 cells.
  • Results were obtained by measuring fluorescence of cells stained with a fluorescein-conjugated anti-Ki67 antibody and subtracting fluorescence of cells exposed to fluorescein-conjugated IgG. Values shown are mean ⁇ SD for three separate donors of MSCs and SB623 cells; * indicates p ⁇ 0.05 compared to control group.
  • FIG. 4 shows phase-contrast photomicrographs of HUVECs following culture for 16 hours in conditioned media from MSCs or SB623 cells.
  • the left-most photograph shows cells cultured in conditioned medium from MSCs; the center photograph shows cells cultured in conditioned medium from SB623 cells; and the right-most photograph shows cells cultured in commercial culture medium without added conditioned medium.
  • FIG. 5 shows measurement of the effect of conditioned medium on tube formation by HUVECs.
  • Left-most bar shows results obtained from control serum/growth factor-starved HUVECs; center bar shows results for serum/growth factor-starved HUVECs cultured for 16 hours in the presence of conditioned medium from MSCs, and the right-most bar shows results for serum/growth factor-starved HUVECs cultured for 16 hours in the presence of conditioned medium from SB623 cells. Values shown are mean ⁇ SEM for three separate donors of MSCs and SB623 cells.
  • FIGS. 6A-6C show photographs of aortic rings after culture for 10 days in unconditioned medium (A), MSC conditioned medium (B), or SB623 cell conditioned medium (C).
  • FIGS. 7A and 7B show measurements of vessel sprouting and branching in an aortic ring assay.
  • FIG. 7A shows counts of new vessels and of branchpoints in the new vessels.
  • the left bar shows measurements of new vessel formation and the right bar shows measurements of vessel branching.
  • the left-most pair of bars (“Control”) shows results obtained from control aortic rings; the center pair of bars (“MSC CM”) shows results obtained from aortic rings cultured for 10 days in MSC conditioned medium; and the right-most pair of bars (“SB623 CM”) shows results obtained from aortic rings cultured for 10 days in SB623 cell conditioned medium.
  • FIG. 7B show ratios of branchpoints to new vessels for control aortic rings (left bar), rings cultured 10 days in MSC conditioned medium (center bar) and rings cultured 10 days in SB623 cell conditioned medium (right bar).
  • FIG. 8 shows levels of four different trophic factors in conditioned medium from MSCs (light bars) and SB623 cells (dark bars). Protein levels are expressed as picograms per ml of conditioned medium per 10 6 cells. Conditioned media from MSCs (and SB623 cells derived therefrom) from four different human donors were tested, as indicated in the figure. Levels of angiogenin, angiopoietin-2, heparin-binding epidermal growth factor-like growth factor (HB-EGF), and placental growth factor (PIGF) are shown.
  • angiogenin angiopoietin-2
  • HB-EGF heparin-binding epidermal growth factor-like growth factor
  • PIGF placental growth factor
  • FIG. 9 shows levels of ten different cytokines in conditioned medium from MSCs and SB623 cells.
  • Cells for production of conditioned medium were obtained from four different donors (D1, D2, D3 and D4), as indicated in the figure.
  • This figure highlights the vast amounts of VEGF produced by MSCs and SB623 cells, compared to the levels of the other trophic factors tested. Abbreviations are given in the legend to Table 1 (Example 6).
  • FIGS. 10A and 10B show the effects of a VEGF receptor inhibitor on improvements in HUVEC viability promoted by SB623 cell-conditioned medium.
  • FIG. 10A shows the fraction of cells permeable to propidium iodide in cultures of HUVECs that had been starved for serum and growth factors. Left-most bar shows results obtained from control serum/growth factor-starved HUVECs; center bar shows results for serum/growth factor-starved HUVECs cultured for five days in the presence of conditioned medium from SB623 cells, and the right-most bar shows results for serum/growth factor-starved HUVECs cultured for five days in the presence of conditioned medium from SB623 cells and 50 nM SU5416. Results were averaged from two donors; “*” indicates p ⁇ 0.05 with respect to control cultures; “#” indicates p ⁇ 0.05 with respect to cultures exposed to SB623 cell conditioned medium and SU5416.
  • FIG. 10B shows measurement of the fraction of cells expressing Bcl-2 in a culture of HUVECs that had been starved for serum and growth factors.
  • Left-most bar shows results obtained from control serum/growth factor-starved HUVECs; center bar shows results for serum/growth factor-starved HUVECs cultured for five days in the presence of conditioned medium from SB623 cells, and the right-most bar shows results for serum/growth factor-starved HUVECs cultured for five days in the presence of conditioned medium from SB623 cells and 50 nM SU5416.
  • Results averaged from duplicate donors, were obtained by measuring fluorescence of cells stained with a fluorescein-conjugated anti-Bcl-2 antibody and subtracting fluorescence of cells exposed to fluorescein-conjugated IgG.
  • FIG. 11 shows measurement of the fraction of cells expressing Ki67 in HUVEC cultures exposed to SB623 cell conditioned medium in the presence and absence of the VEGFR2 inhibitor SU5416, and by control cells cultured in the absence of CM.
  • the left-most (clear) bar shows results obtained from control serum/growth factor-starved HUVECs; the center (black) bar shows results for serum/growth factor-starved HUVECs cultured in the presence of conditioned medium from SB623 cells, and the right-most (gray) bar shows results for serum/growth factor-starved HUVECs cultured in the presence of conditioned medium from SB623 cells and 50 nM SU5416. Values shown are mean ⁇ SEM for two separate donors of SB623 cells. “*” indicates p ⁇ 0.05 with respect to the negative control cultures (no conditioned medium); “#” indicates p ⁇ 0.05 with respect to SU5416-treated cultures.
  • FIG. 12 shows the effects of a VEGF receptor inhibitor on the enhancement of tube formation by HUVECs promoted by MSC- and SB623 cell-conditioned media.
  • the top row shows cells cultured in the absence of the inhibitor.
  • the left-most panel of the top row (“neg”) shows a phase-contrast photomicrograph of control HUVECs following culture for 16 hours in Opti-MEM Medium.
  • the second panel from the left (“+10 ng VEGF”) shows a phase-contrast photomicrograph of HUVECs following culture for 16 hours in Opti-MEM Medium to which 10 ng/ml VEGF was added.
  • the third panel from the left (“+MSC-CM”) shows a phase-contrast photomicrograph of HUVECs following culture for 16 hours in MSC-conditioned medium.
  • the rightmost panel (“+SB623-CM”) shows a phase-contrast photomicrograph of HUVECs following culture for 16 hours in SB623 cell-conditioned medium.
  • Panels in the bottom row show photomicrographs of HUVECs under the same conditions as in the top row but with the addition of 50 nM SU5416.
  • FIG. 13 shows quantitation of tube formation by HUVECs exposed to SB623 cell conditioned medium in the presence and absence of the VEGFR2 inhibitor SU5416, and by control cells cultured in the absence of CM.
  • the left-most (clear) bar shows results obtained from control serum/growth factor-starved HUVECs; the center (black) bar shows results for serum/growth factor-starved HUVECs cultured in the presence of conditioned medium from SB623 cells, and the right-most (gray) bar shows results for serum/growth factor-starved HUVECs cultured in the presence of conditioned medium from SB623 cells and 50 nM SU5416. Values shown are mean ⁇ SEM for three separate donors of SB623 cells. “*” indicates p ⁇ 0.05 with respect to the negative control cultures (no conditioned medium); “#” indicates p ⁇ 0.05 with respect to SU5416-treated cultures.
  • FIG. 14 shows the effects of a VEGF receptor inhibitor on enhancement of vessel outgrowth and branching promoted by SB623 cell-conditioned medium in an aortic ring assay.
  • the left panel shows a photomicrograph of an aortic ring after culture for 10 days on a RGF-basement gel in OptiMEM medium (“Negative control”).
  • the center panel shows a photomicrograph of an aortic ring after culture for 10 days in SB623 cell conditioned medium (“+SB623-CM”).
  • the right panel shows a photomicrograph of an aortic ring after culture for 10 days in SB623 cell conditioned medium containing 50 nM SU5416 (“+SB623-CM+SU5416”). Enlargements of certain regions of each photomicrograph are shown in the lower row.
  • SB623 cells cells descended from MSCs that have been transfected with a vector containing sequences encoding a Notch intracellular domain
  • SB623 cells promote survival and proliferation of endothelial cells in vitro under serum- and growth factor-deprived conditions, and stimulate vascular tube formation by human umbilical vein endothelial cells.
  • conditioned medium from SB623 cells promoted endothelial sprouting and branching in a rodent aortic ring assay.
  • angiogenesis refers to the formation of new vasculature (e.g., blood vessels; e.g., veins, arteries, venules, arterioles, capillaries). Angiogenesis can occur by sprouting of new vessels from an existing vessel, and/or by branching of a vessel. Angiogenesis also includes the attendant processes of matrix remodeling and cell recruitment (e.g., recruitment of smooth muscle cells, monocytes and/or pericytes).
  • MSCs refer to adherent, non-hematopoietic stem cells obtained from bone marrow. These cells are variously known as mesenchymal stem cells, mesenchymal stromal cells, marrow adherent stromal cells, marrow adherent stem cells and bone marrow stromal cells.
  • Stroke is the name given to conditions resulting from impairment of blood flow in the brain. Such cerebrovascular impairment can result, for example, from intracranial hemorrhage, or from reduction or blockage of blood flow in the brain (i.e., cerebral ischemia). Ischemic blockages can result from thrombosis (i.e., formation of a clot in situ in a cranial vessel or a vessel supplying the brain) or from a cerebral embolism (i.e., migration of a clot to a site in the brain). The damage resulting from ischemic or hemorrhagic stroke usually results in impairment of neurological function. Additional information relating to different types of stroke, and their characteristics, is found in co-owned U.S. Pat. No. 8,092,792; the disclosure of which is incorporated by reference in its entirety herein for the purpose of describing different types of stroke and their characteristics.
  • MSCs Mesenchymal Stem Cells
  • SB623 cells are obtained from marrow adherent stromal cells, also known as mesenchymal stem cells (MSCs), by expressing the intracellular domain of the Notch protein in the MSCs.
  • MSCs are obtained by selecting adherent cells (i.e., cells that adhere to tissue culture plastic) from bone marrow.
  • MSCs Exemplary disclosures of MSCs are provided in U.S. patent application publication No. 2003/0003090; Prockop (1997) Science 276:71-74 and Jiang (2002) Nature 418:41-49. Methods for the isolation and purification of MSCs can be found, for example, in U.S. Pat. No. 5,486,359; Pittenger et al. (1999) Science 284:143-147 and Dezawa et al. (2001) Eur. J. Neurosci. 14:1771-1776.
  • Human MSCs are commercially available (e.g., BioWhittaker, Walkersville, Md.) or can be obtained from donors by, e.g., bone marrow aspiration, followed by selection for adherent bone marrow cells. See, e.g., WO 2005/100552.
  • MSCs can also be isolated from umbilical cord blood. See, for example, Campagnoli et al. (2001) Blood 98:2396-2402; Erices et al. (2000) Br. J. Haematol. 109:235-242 and Hou et al. (2003) Int. J. Hematol. 78:256-261.
  • the Notch protein is a transmembrane receptor, found in all metazoans, that influences cell differentiation through intracellular signaling.
  • a Notch ligand e.g., Delta, Serrate, Jagged
  • NBD Notch intracellular domain
  • the NICD translocates to the nucleus, where it acts as a transcription factor, recruiting additional transcriptional regulatory proteins (e.g., MAM, histone acetylases) to relieve transcriptional repression of various target genes (e.g., Hes 1).
  • additional transcriptional regulatory proteins e.g., MAM, histone acetylases
  • Notch signaling is found, for example in Artavanis-Tsakonas et al. (1995) Science 268:225-232; Mumm and Kopan (2000) Develop. Biol. 228:151-165 and Ehebauer et al. (2006) Sci. STKE 2006 (364), cm7. [DOI: 10.1126/stke.3642006 cm7].
  • a culture of MSCs is contacted with a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD); e.g., by transfection; followed by enrichment of transfected cells by drug selection and further culture.
  • a polynucleotide comprising sequences encoding a Notch intracellular domain (NICD); e.g., by transfection; followed by enrichment of transfected cells by drug selection and further culture.
  • any polynucleotide encoding a Notch intracellular domain can be used, and any method for the selection and enrichment of transfected cells can be used.
  • MSCs are transfected with a vector containing sequences encoding a Notch intracellular domain and also containing sequences encoding a drug resistance marker (e.g. resistance to G418).
  • a drug resistance marker e.g. resistance to G418
  • two vectors one containing sequences encoding a Notch intracellular domain and the other containing sequences encoding a drug resistance marker, are used for transfection of MSCs.
  • selection is achieved, after transfection of a cell culture with the vector or vectors, by adding a selective agent (e.g., G418) to the cell culture in an amount sufficient to kill cells that do not comprise the vector but spare cells that do. Absence of selection entails removal of said selective agent or reduction of its concentration to a level that does not kill cells that do not comprise the vector. Following selection (e.g., for seven days) the selective agent is removed and the cells are further cultured (e.g., for two passages).
  • a selective agent e.g., G4108
  • Preparation of SB623 cells thus involves transient expression of an exogenous Notch intracellular domain in a MSC.
  • MSCs can be transfected with a vector comprising sequences encoding a Notch intracellular domain wherein said sequences do not encode a full-length Notch protein. All such sequences are well known and readily available to those of skill in the art. For example, Del Amo et al. (1993) Genomics 15:259-264 present the complete amino acid sequences of the mouse Notch protein; while Mumm and Kopan (2000) Devel. Biol. 228:151-165 provide the amino acid sequence, from mouse Notch protein, surrounding the so-called S3 cleavage site which releases the intracellular domain.
  • SB623 cells are prepared by introducing, into MSCs, a nucleic acid comprising sequences encoding a Notch intracellular domain such that the MSCs do not express exogenous Notch extracellular domain. Such can be accomplished, for example, by transfecting MSCs with a vector comprising sequences encoding a Notch intracellular domain wherein said sequences do not encode a full-length Notch protein.
  • SB623 cells mesenchymal stem cells in which a Notch intracellular domain has been transiently expressed
  • SB623 cells descendants of mesenchymal stem cells in which a Notch intracellular domain has been transiently expressed
  • SB623 cells have angiogenic activity; and that said cells synthesize and secrete angiogenic factors. Accordingly, transplantation of SB623 cells is useful for treatment of disorders in which a therapeutic benefit can be achieved by increasing angiogenesis in a subject.
  • Such disorders include, but are not limited to, cerebral ischemia (e.g., stroke), cardiac ischemia (e.g., ischemic heart disease), ischemia of the bowel (e.g., ischemic colitis, mesenteric ischemia), ischemia of the limb, cutaneous ischemia, ocular ischemic syndrome (e.g., retinal ischemia) and cerebral palsy.
  • cerebral ischemia e.g., stroke
  • cardiac ischemia e.g., ischemic heart disease
  • ischemia of the bowel e.g., ischemic colitis, mesenteric ischemia
  • ischemia of the limb e.g., ischemic colitis, mesenteric ischemia
  • ocular ischemic syndrome e.g., retinal ischemia
  • cerebral palsy e.g., cerebral palsy.
  • SB623 cells as described herein can be used in a number of methods related to stimulation of angiogenesis. These include, but are not limited to, treatment of any of the disorders mentioned in the previous paragraph, augmentation of angiogenesis, repair of ischemic damage, preventing death of endothelial cells, enhancing survival of endothelial cells, stimulating proliferation of endothelial cells, and/or enhancing the branching of blood vessels,
  • Such methods can be performed in vitro or in a subject.
  • the subject can be a mammal, preferably a human.
  • Stimulation of angiogenesis by SB623 cells, and the attendant effects of such stimulation as disclosed herein, can occur, for example, in the central nervous system (e.g., in the brain).
  • Transplantation of SB623 cells can also be used in methods for providing one or more angiogenic trophic factors to a subject.
  • angiogenic trophic factors include, but are not limited to, angiogenin, angiopoietin-2, epidermal growth factor, basic fibroblast growth factor, heparin-binding epithelial growth factor-like growth factor, hepatocyte growth factor, leptin, platelet-derived growth factor-BB, placental growth factor and vascular endothelial growth factor.
  • SB623 cells can be used in combination with a second pro-angiogenic agent, in combination therapies for increasing angiogenesis in a subject.
  • Said combination therapies can be used for all of the purposes set forth above.
  • the second pro-angiogenic agent can be, e.g., a small molecule drug, a nucleic acid or a polypeptide (e.g., antibody, transcription factor).
  • nucleic acids are triplex-forming nucleic acids, ribozymes and siRNAs that activate expression of angiogenic proteins and/or block expression of anti-angiogenic proteins.
  • Exemplary antibodies are those that bind to and/or inhibit the activity of angiogenic proteins (or other angiogenic agents).
  • Exemplary transcription factors are those that inhibit transcription of a gene encoding one or more anti-angiogenic protein(s), as well as those that activate the transcription of one or more pro-angiogenic protein(s).
  • Anti-angiogenic and pro-angiogenic proteins are known in the art.
  • Exemplary anti-angiogenic proteins include pigment epithelium derived factor (PEDF) and placental growth factor (PlGF).
  • Exemplary pro-angiogenic proteins include vascular endothelial growth factor (VEGF) angiopoietin, and hepatocyte growth factor (HGF).
  • transcription factors as disclosed above are non-naturally-occurring (engineered) transcription factors.
  • An example of such a non-naturally-occurring transcription factor is a non-naturally-occurring zinc finger protein that has been engineered to bind to a DNA sequence in cellular chromatin that regulates transcription of a target gene (e.g., a VEGF gene).
  • Said engineered zinc finger transcription factors comprise, in addition to an engineered zinc finger DNA-binding domain, a transcriptional regulatory domain (e.g., a transcriptional activation domain or a transcriptional repression domain), as are known in the art.
  • Zinc finger binding domain are engineered to have a novel binding specificity, compared to a naturally-occurring zinc finger protein.
  • Engineering methods include, but are not limited to, rational design and various types of empirical selection methods. Rational design includes, for example, using databases comprising triplet (or quadruplet) nucleotide sequences and individual zinc finger amino acid sequences, in which each triplet or quadruplet nucleotide sequence is associated with one or more amino acid sequences of zinc fingers which bind the particular triplet or quadruplet sequence. See, for example, U.S. Pat. Nos. 6,140,081; 6,453,242; 6,534,261; 6,610,512; 6,746,838; 6,866,997; 7,067,617; U.S. Patent Application Publication Nos. 2002/0165356; 2004/0197892; 2007/0154989; 2007/0213269; and International Patent Application Publication Nos. WO 98/53059 and WO 2003/016496.
  • Exemplary selection methods including phage display, interaction trap, hybrid selection and two-hybrid systems, are disclosed in U.S. Pat. Nos. 5,789,538; 5,925,523; 6,007,988; 6,013,453; 6,140,466; 6,200,759; 6,242,568; 6,410,248; 6,733,970; 6,790,941; 7,029,847 and 7,297,491; as well as U.S. Patent Application Publication Nos. 2007/0009948 and 2007/0009962; WO 98/37186; WO 01/60970 and GB 2,338,237.
  • Transcriptional activation and repression domain are known in the art. See, e.g., Science 269:630 (1995).
  • Exemplary transcriptional activation domains include p65, VP16 and VP64.
  • Exemplary transcriptional repression domains include KRAB, KAP-1, MAD, FKHR, ERD and SID.
  • Functional domains from nuclear hormone receptors can act as either activators or repressors, depending upon the presence of a ligand. See also U.S. Pat. No. 7,985,887.
  • compositions comprising SB623 cells as disclosed herein are also provided. Such compositions typically comprise the SB623 cells and a pharmaceutically acceptable carrier. Supplementary active compounds can also be incorporated into SB623 cell compositions (see below).
  • a “therapeutically effective amount” of a composition comprising SB623 cells can be any amount that stimulates angiogenesis.
  • dosage amounts can vary from about 100; 500; 1,000; 2,500; 5,000; 10,000; 20,000; 50,000; 100,000; 500,000; 1,000,000; 5,000,000 to 10,000,000 cells or more (or any integral value therebetween); with a frequency of administration of, e.g., once per day, twice per week, once per week, twice per month, once per month, depending upon, e.g., body weight, route of administration, severity of disease, etc.
  • compositions and techniques for their preparation and use are known to those of skill in the art in light of the present disclosure.
  • suitable pharmacological compositions and techniques for their administration one may refer to texts such as Remington's Pharmaceutical Sciences, 17th ed. 1985; Brunton et al., “Goodman and Gilman's The Pharmacological Basis of Therapeutics,” McGraw-Hill, 2005; University of the Sciences in Philadelphia (eds.), “Remington: The Science and Practice of Pharmacy,” Lippincott Williams & Wilkins, 2005; and University of the Sciences in Philadelphia (eds.), “Remington: The Principles of Pharmacy Practice,” Lippincott Williams & Wilkins, 2008.
  • physiologically compatible carrier refers to a carrier that is compatible with the SB623 cells and with any other ingredients of the formulation, and is not deleterious to the recipient thereof.
  • suitable carriers include cell culture medium (e.g., Eagle's minimal essential medium), phosphate buffered saline, Hank's balanced salt solution+/ ⁇ glucose (HBSS), and multiple electrolyte solutions such as Plasma-LyteTM A (Baxter).
  • the volume of a SB623 cell suspension administered to a patient will vary depending on the site of implantation, treatment goal and number of cells in solution. Typically the amount of cells administered to a patient will be a therapeutically effective amount.
  • a “therapeutically effective amount” or “effective amount” refers to the number of transplanted cells which are required to effect treatment of the particular disorder; i.e., to produce a reduction in the amount and/or severity of the symptoms associated with that disorder.
  • transplantation of a therapeutically effective amount of SB623 cells results in new vessel growth, vessel sprouting and vessel branching, e.g., in an area that has been damaged by ischemia.
  • Therapeutically effective amounts vary with the type and extent of ischemic damage, and can also vary depending on the overall condition of the subject.
  • compositions or vehicles can also include pharmaceutically acceptable materials, compositions or vehicles, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, i.e., carriers.
  • carriers can, for example, stabilize the SB623 cells and/or facilitate the survival of the SB623 cells in the body.
  • Each carrier should be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject.
  • materials which can serve as pharmaceutically-acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • Exemplary formulations include, but are not limited to, those suitable for parenteral administration, e.g., intrapulmonary, intravenous, intra-arterial, intra-ocular, intra-cranial, sub-meningial, or subcutaneous administration, including formulations encapsulated in micelles, liposomes or drug-release capsules (active agents incorporated within a biocompatible coating designed for slow-release); ingestible formulations; formulations for topical use, such as eye drops, creams, ointments and gels; and other formulations such as inhalants, aerosols and sprays.
  • the dosage of the compositions of the disclosure will vary according to the extent and severity of the need for treatment, the activity of the administered composition, the general health of the subject, and other considerations well known to the skilled artisan.
  • compositions described herein are delivered locally to a site of ischemic damage.
  • Localized delivery allows for the delivery of the composition non-systemically, thereby reducing the body burden of the composition as compared to systemic delivery.
  • Such local delivery can be achieved, for example, by intra-cranial injection, or through the use of various medically implanted devices including, but not limited to, stents and catheters, or can be achieved by inhalation, phlebotomy, or surgery.
  • Methods for coating, implanting, embedding, and otherwise attaching desired agents to medical devices such as stents and catheters are established in the art and contemplated herein.
  • kits for carrying out the administration of SB623 cells optionally in combination with another therapeutic agent, to a subject.
  • a kit comprises a composition of SB623 cells, formulated in a pharmaceutical carrier, optionally containing, e.g., a pro-angiogenic agent (see below), formulated as appropriate, in one or more separate pharmaceutical preparations.
  • SB623 cell compositions can be used in combination with other compositions comprising substances that stimulate angiogenesis (“pro-angiogenic agents”), e.g., for treatment of stroke.
  • the compositions can be administered sequentially in any order or concurrently.
  • therapeutic compositions as disclosed herein can contain both SB623 cells and a pro-angiogenic agent.
  • separate therapeutic compositions one comprising SB623 cells and the other comprising a pro-angiogenic agent, can be administered to the subject, either separately or together.
  • a pro-angiogenic agent is a protein (e.g., fibroblast growth factor, platelet-derived growth factor, transforming growth factor alpha, hepatocyte growth factor, vascular endothelial growth factor, sonic hedgehog, MAGP-2, HIF-1, PR-39, RTEF-1, c-Myc, TFII, Egr-1, ETS-1) or a nucleic acid encoding such a protein.
  • a protein e.g., fibroblast growth factor, platelet-derived growth factor, transforming growth factor alpha, hepatocyte growth factor, vascular endothelial growth factor, sonic hedgehog, MAGP-2, HIF-1, PR-39, RTEF-1, c-Myc, TFII, Egr-1, ETS-1
  • a nucleic acid encoding such a protein See, for example, Vincent et al. (2007) Gene Therapy 14:781-789.
  • a pro-angiogenic agent can be a small RNA molecule (e.g., siRNA, shRNA, microRNA) or a ribozyme that targets a nucleic acid encoding an inhibitor of angiogenesis.
  • a pro-angiogenic agent can be a triplex-forming nucleic acid that binds to DNA sequences regulating the expression of a protein that inhibits angiogenesis, such as to block transcription of the gene encoding the protein.
  • a pro-angiogenic agent is a transcription factor that activates expression of a pro-angiogenic molecule (e.g., protein).
  • a pro-angiogenic molecule e.g., protein
  • Naturally-occurring transcription factors such as, for example, HIF-1alpha
  • synthetic transcriptional regulatory proteins can be constructed by genetic engineering. For example, methods for the design of zinc finger DNA-binding domains that bind to a sequence of interest, and methods for the fusion of such zinc finger DNA-binding domains to transcriptional activation and repression domains, have been described. See, for example, U.S. Pat. Nos.
  • a non-naturally-occurring (i.e., synthetic) zinc finger protein that activates transcription of the VEGF gene can be used, in combination with SB623 cells, for augmenting angiogenesis, e.g., in the treatment of stroke.
  • a natural or synthetic transcriptional regulatory protein e.g., a synthetic zinc finger transcriptional regulatory protein
  • a pro-angiogenic agent e.g., a synthetic zinc finger transcriptional regulatory protein
  • MSCs and SB623 cells were obtained and/or prepared as described. See, for example, U.S. Pat. No. 7,682,825 (Mar. 23, 2010) and U.S. Patent Application Publications Nos. 2010/0266554 (Oct. 21, 2010), 2010/0310529 (Dec. 9, 2010), 2011/0229442 (Sep. 22, 2011), and 2011/0306137 (Dec. 15, 2011); the disclosures of which are incorporated by reference in their entireties for the purposes of describing the preparation of SB623 cells (variously referred to as “neural precursor cells” and “neural regenerating cells” in those documents).
  • MSCs and SB623 cells were cultured in growth medium, which contained alph ⁇ -MEM (Mediatech, Herndon, Va.) supplemented with 10% fetal bovine serum (FBS, Hyclone, Logan, Utah), 2 mM L-glutamine and 1% penicillin/streptomycin (both from Invitrogen, Carlsbad, Calif.).
  • MSCs and SB623 cells typically expressed CD29, CD90 and CD105; and did not express CD31, CD34, or CD45, as determined by flow cytometry.
  • frozen MSCs and SB623 cells from the same human donor were thawed, re-plated in growth medium, and allowed to recover for approximately one week.
  • conditioned medium cells were grown to approximately 90% confluence (15,000 cells/cm 2 ), the plates were rinsed once with phosphate buffered saline (PBS) and the medium was then replaced with OptiMEM® medium (Invitrogen, Carlsbad, Calif.), maintaining the same cell density.
  • PBS phosphate buffered saline
  • OptiMEM® medium Invitrogen, Carlsbad, Calif.
  • HUVECs human umbilical vein endothelial cells
  • CM conditioned medium
  • EBM-2/ECGS medium Endothelial Basal Medium-2/Endothelial Cell Growth Supplements; Lonza, Walkersville, Md.
  • the HUVEC monolayers were rinsed twice with warm PBS and incubated with 12 ml of fresh EBM-2 medium overnight at 37° C., 5% CO 2 .
  • CM Effects of CM were then assessed by withdrawing 6 ml of medium from each flask, and replacing it with 6 ml fresh OptiMEM (control), 6 ml MSC conditioned medium, or 6 ml SB623 cell conditioned medium (conditioned media prepared as described in Example 1). After 7 days, non-adherent and adherent cells were collected, centrifuged at 1400 rpm for 5 min, and divided into three fractions for subsequent staining analyses (PI, Bcl-2 and Ki67).
  • Bcl-2 is an anti-apoptotic protein originally identified as being overexpressed in certain B-cell lymphomas. Accordingly, the fraction of cells expressing the Bcl-2 protein was measured in serum/growth factor-starved HUVECs as an indicator of their apoptotic potential.
  • Bcl-2 measurement cells were fixed in 4% paraformaldehyde and permeabilized with 0.1% Triton-X100 for one hour. Following permeabilization, cells were stained for one hour, on ice, with fluorescein-conjugated anti-Bcl-2 antibody, then samples were washed, acquired, and analyzed on the FL-1 channel of a BD FACSCalibur. Cells exposed to fluorescein-conjugated IgG were used as a negative control. For these assays, 3 different human donor pairs were tested.
  • results, shown in FIG. 2 indicate that presence of either MSC conditioned medium or SB623 cell conditioned medium significantly increased the fraction of Bcl-2-positive cells in cultures of serum-starved endothelial cells.
  • conditioned medium from MSCs or from SB623 cells decreased the number of dead (PI-positive) cells and increased of the number of cells expressing the anti-apoptotic Bcl-2 protein shows that both MSCs and SB623 cells secrete factors that enhance endothelial cell survival.
  • Ki67 is a protein present in cells exiting from the GO (quiescent) phase of the cell cycle; therefore Ki67 levels can be used as a measure of cell proliferation.
  • the fraction of cells expressing Ki67 protein was measured in HUVECs that had been starved for serum and growth factors, then cultured with conditioned medium from either MSCs or SB623 cells.
  • HUVECs were cultured and exposed to CM as described in Example 2. Cells were fixed in 4% paraformaldehyde and permeabilized with 0.1% Triton-X100 for one hour. Following permeabilization, cells were stained for one hour on ice with fluorescein-conjugated anti-KI67 antibody, then samples were washed, acquired, and analyzed on the FL-1 channel of a BD FACSCalibur. Cells exposed to fluorescein-conjugated IgG were used as a negative control. For these assays, 3 different human donor pairs were tested.
  • FIG. 3 shows that culture of starved HUVECs in the presence of conditioned medium from either MSCs or SB623 cells resulted in an increased fraction of cells expressing Ki67, compared to control HUVECs not exposed to conditioned medium.
  • conditioned medium from MSCs or from SB623 cells increased the number of cells expressing the proliferation-associated Ki67 protein shows that both MSCs and SB623 cells secrete factors that enhance endothelial cell proliferation.
  • a HUVEC tube formation assay was used to test the ability of MSCs and SB623 cells to elaborate factors that stimulate vessel formation. See, for example, E J Smith & CA Staton, “Tubule formation assays,” in Angiogenesis Assays—A Critical Appraisal of Current Techniques , (Staton, Lewis & Bicknell, eds.). John Wiley & Sons, Ltd., West Wales, UK, pp. 65-87, 2006; and Goodwin (2007) Microvasc. Res. 74:172-183.
  • HUVECs were passed five times in EBM-2/ECGS medium, then transferred to alph ⁇ -MEM/0.5% FBS/2 mM glutamine/pen-strep, at a density of 1 ⁇ 10 5 cells/ml. After 24 hours, HUVECs were harvested using 0.25% trypsin-EDTA, rinsed, and resuspended in ⁇ -MEM/2 mM glutamine/pen-strep at a density of 1 ⁇ 10 5 cells/ml.
  • RGF Reduced Growth Factor
  • vasculature after ischemic injury requires that surviving endothelial cells receive signals that prompt their migration and invasion. Such signals may arise from vascular smooth muscle cells, monocytes, and/or macrophages, among others.
  • the aortic ring assay was used. See, for example, Nicosia & Ottinetti (1990) Lab. Invest. 63:115-122 and Nicosia (2009) J. Cell. Mol. Med. 13:4113-4136.
  • aortic rings For preparation of aortic rings, adult Sprague-Dawley rats were euthanized prior to dissection. After clamping off its two ends, the aorta was removed and placed in ice-cold ⁇ -MEM/pen-strep medium prior to removal of the external adipose layer. Adipose-free aorta was rinsed twice with ice-cold EBM-2/pen-strep medium before being sectioned into rings of 1.0 mm thickness.
  • the aortic rings were then transferred to plates containing EBM-2/pen-strep medium and incubated at 37° C., 5% CO 2 for 6 days, with the medium replaced with fresh EBM-2/pen-strep medium on day 3, to deplete any endogenous rat angiogenic factors. At that point, the medium was replaced with alpha-MEM/pen-strep medium and culture was continued for 24 hours.
  • phase contrast photographs were taken on Day 10, and results were quantified by an experimenter blinded to the group, by counting vessel outgrowth and branching. Growth of new vessels was quantitated by measuring the number of vessels growing out from the ring; and vessel branching was quantitated by measuring the number of branchpoints present in vessels growing out from the aortic ring. For this assay, 7 different human donor pairs were tested.
  • FIG. 7A shows that conditioned medium from both MSCs and SB623 cells stimulated an increase in the number of newly-sprouted vessels and in the degree of branching, compared to control aortic rings.
  • FIG. 6C shows that significant increases in vessel branching were observed in rings cultured in SB623 cell-conditioned medium
  • FIG. 6B shows that rings cultured in MSC-conditioned medium
  • FIG. 7A shows that rings cultured in unconditioned medium.
  • MSCs and SB623 cells The levels of certain cytokines and trophic factors in conditioned medium from MSCs and SB623 cells were measured.
  • MSCs or SB623 cells were cultured in growth medium to ⁇ 90% confluence (15,000 cells/cm 2 ), at which point medium was removed, the cells were rinsed in PBS, and Opti-MEM® medium (Invitrogen, Carlsbad, Calif.) was added to give a concentration of ⁇ 150,000 cells/ml.
  • the conditioned medium was collected 72 hours later and assayed using a Quantibody®Human Angiogenesis Array 1 (RayBiotech, Norcross, Ga.) according to the manufacturer's instructions.
  • a culture of MSCs from a particular donor and a culture of SB623 cells made from those MSCs are referred to as a matched “donor pair.”
  • donor pair a culture of MSCs from a particular donor and a culture of SB623 cells made from those MSCs.
  • four donor pairs were assayed.
  • Results, expressed as protein concentration, were normalized to the number of cells present in the culture when the conditioned medium was collected.
  • FIG. 8 shows results, by donor, for angiogenin, ANG-2, HB-EGF and P1GF.
  • FIG. 9 shows results for these four factors, and six others, also by donor, and highlights the large amounts of VEGF produced by MSCs and SB623 cells.
  • Table 1 shows protein levels averaged among the four donor pairs for the ten factors tested. Although levels of trophic factors secreted were variable among the different donors (as shown, for example, in FIGS. 8 and 9 ), levels of four of the factors (angiogenin, angiopoietin-2, HB-EGF and P1GF) were consistently different between MSCs and SB623 cells. Angiogenin, ANG-2 and HB-EGF were more highly expressed by SB623 cells, while higher concentrations of P1GF were produced by MSCs.
  • ANG-2 angiopoietin-2
  • EGF epidermal growth factor
  • bFGF basic fibroblast growth factor/fibroblast growth factor 2
  • HB-EGF heparin-binding epidermal growth factor-like growth factor
  • HGF hepatocyte growth factor
  • PDGF-BB platelet-derived growth factor-BB
  • PIGF placental growth factor
  • VEGF vascular endothelial growth factor. Numbers refer to cytokine levels expressed as pg/ml/10 6 cells. “AVG” refers to the average value from 4 sources of MSCs and 4 sources of SB623 cells from which conditioned medium was obtained; “SD” refers to standard deviation. “—” indicates that levels, if any, were below the limit of detection in the assay; “n/a” indicates “not applicable”
  • VEGF receptor 2 Flk-1
  • Flt-1 VEGF receptor 1
  • HUVEC viability assays (propidium iodide uptake and Bcl-2 expression) were conducted as described in Example 2 on two batches of SB623 cell-conditioned medium, in the presence and absence of 50 nM SU5416; except that cells were cultured for five days, instead of seven, before assay. The inhibitor was added to cultures 30 minutes before addition of CM. Since higher concentrations of SU5416 can inhibit receptor tyrosine kinases other than VEGFR2, this SU5416 concentration was chosen so that VEGFR2 signaling (but not signaling by, e.g., PDGF receptor, EGF receptors, or Flt3) was inhibited. The results, shown in FIGS.
  • FIG. 10A and 10B indicate that more cells take up PI ( FIG. 10A ) and fewer cells express the anti-apoptotic Bcl-2 protein ( FIG. 10B ) when HUVECs are cultured in SB623 conditioned medium and SU5416, than when they are cultured in SB623 cell-conditioned medium alone.
  • inhibition of VEGF receptor activity partially reduces the positive effect of SB623 cell-conditioned medium on viability of HUVECs, pointing to a role of the VEGF protein in these effects.
  • VEGF receptor inhibitor The effect of the VEGF receptor inhibitor on stimulation of HUVEC proliferation by SB623 cell factors was also assessed. Assays for expression of Ki67 were conducted as described in Example 3, except that 50 nM SU5416 was added to cultures 30 minutes before addition of conditioned medium, and cells were cultured for five days, instead of seven, before assay. The results, shown in FIG. 11 and averaged from two donors, indicate that the enhancement of HUVEC proliferation observed in the presence of conditioned medium from SB623 cells was partially reversed by inhibition of VEGFR2.
  • HUVEC tube formation assays with conditioned medium from MSCs and SB623 cells were conducted as described in Example 4, in the presence and absence of the VEGF2 receptor inhibitor SU5416.
  • Cells cultured in the absence of conditioned medium were used as negative controls; and cells cultured in the presence of VEGF (10 ng/ml were used as positive controls.
  • the results, shown in FIG. 12 indicate that VEGF, MSC-conditioned medium and SB623 cell-conditioned medium all promote tube formation; while the VEGFR2 inhibitor SU5416 reduces the stimulation of tube formation by all of these agents.
  • Aortic ring angiogenesis assays were conducted as described in Example 5 on one batch of SB623 cell-conditioned medium, in the presence and absence of 50 nM SU5416. The inhibitor was added to cultures 30 minutes before addition of CM and rings were assayed after 10 days of culture. The results indicate that the vessel outgrowth and branching resulting from culture of aortic rings in SB623 cell-conditioned medium ( FIG. 14 , compare left and center panels) was reduced in the presence of the VEGF receptor inhibitor SU5416 ( FIG. 14 , compare center and right panels). These results provide further evidence for the role of VEGF in the pro-angiogenic activities of SB623 cell-conditioned medium.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Vascular Medicine (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Cardiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Botany (AREA)
  • Dermatology (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Transplantation (AREA)
US13/750,772 2012-01-27 2013-01-25 Methods and compositions for modulating angiogenesis and vasculogenesis Abandoned US20130195817A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/750,772 US20130195817A1 (en) 2012-01-27 2013-01-25 Methods and compositions for modulating angiogenesis and vasculogenesis
US15/063,290 US20160263159A1 (en) 2012-01-27 2016-03-07 Methods and compositions for modulating angiogenesis and vasculogenesis
US15/608,656 US10245286B2 (en) 2012-01-27 2017-05-30 Methods and compositions for modulating angiogenesis and vasculogenesis
US16/269,000 US11304982B2 (en) 2012-01-27 2019-02-06 Methods and compositions for modulating angiogenesis and vasculogenesis
US17/654,778 US20220193144A1 (en) 2012-01-27 2022-03-14 Methods and compositions for modulating angiogenesis and vasculogenesis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261591486P 2012-01-27 2012-01-27
US201261637740P 2012-04-24 2012-04-24
US201261709619P 2012-10-04 2012-10-04
US13/750,772 US20130195817A1 (en) 2012-01-27 2013-01-25 Methods and compositions for modulating angiogenesis and vasculogenesis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/063,290 Continuation US20160263159A1 (en) 2012-01-27 2016-03-07 Methods and compositions for modulating angiogenesis and vasculogenesis

Publications (1)

Publication Number Publication Date
US20130195817A1 true US20130195817A1 (en) 2013-08-01

Family

ID=47710337

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/750,772 Abandoned US20130195817A1 (en) 2012-01-27 2013-01-25 Methods and compositions for modulating angiogenesis and vasculogenesis
US15/063,290 Abandoned US20160263159A1 (en) 2012-01-27 2016-03-07 Methods and compositions for modulating angiogenesis and vasculogenesis
US15/608,656 Active US10245286B2 (en) 2012-01-27 2017-05-30 Methods and compositions for modulating angiogenesis and vasculogenesis
US16/269,000 Active 2034-02-12 US11304982B2 (en) 2012-01-27 2019-02-06 Methods and compositions for modulating angiogenesis and vasculogenesis
US17/654,778 Pending US20220193144A1 (en) 2012-01-27 2022-03-14 Methods and compositions for modulating angiogenesis and vasculogenesis

Family Applications After (4)

Application Number Title Priority Date Filing Date
US15/063,290 Abandoned US20160263159A1 (en) 2012-01-27 2016-03-07 Methods and compositions for modulating angiogenesis and vasculogenesis
US15/608,656 Active US10245286B2 (en) 2012-01-27 2017-05-30 Methods and compositions for modulating angiogenesis and vasculogenesis
US16/269,000 Active 2034-02-12 US11304982B2 (en) 2012-01-27 2019-02-06 Methods and compositions for modulating angiogenesis and vasculogenesis
US17/654,778 Pending US20220193144A1 (en) 2012-01-27 2022-03-14 Methods and compositions for modulating angiogenesis and vasculogenesis

Country Status (8)

Country Link
US (5) US20130195817A1 (ja)
EP (2) EP2806881A1 (ja)
JP (2) JP6023218B2 (ja)
CN (2) CN104254337A (ja)
AU (5) AU2013211934B2 (ja)
CA (1) CA2862309A1 (ja)
HK (1) HK1199200A1 (ja)
WO (1) WO2013112917A1 (ja)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9326999B2 (en) 2012-10-09 2016-05-03 Sanbio, Inc. Methods and compositions for treatment of retinal degeneration
US9828593B2 (en) 2012-05-16 2017-11-28 Sanbio, Inc. Methods and compositions for treatment of traumatic brain injury and for modulation of migration of neurogenic cells
US10245286B2 (en) 2012-01-27 2019-04-02 Sanbio, Inc. Methods and compositions for modulating angiogenesis and vasculogenesis
US20220387506A1 (en) * 2021-05-27 2022-12-08 Sanbio, Inc. Cell therapies and methods of treatment for small-volume stroke

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016161290A1 (en) * 2015-04-01 2016-10-06 Sanbio, Inc. Methods and compositions for stimulation of cell proliferation and provision of biologically active mixtures of fgf2 isoforms
WO2018125829A1 (en) * 2016-12-28 2018-07-05 Sanbio, Inc. Cell delivery system and methods of operation thereof
JP2020523027A (ja) * 2017-06-16 2020-08-06 イーエムベーアー−インスティテュート フュール モレクラレ バイオテクノロジー ゲゼルシャフト ミット ベシュレンクテル ハフツング 血管オルガノイド、オルガノイドの製造及び使用方法
CN108130326A (zh) * 2017-12-27 2018-06-08 南昌大学第附属医院 miRNA反义核苷酸的用途及修饰的间充质干细胞及修饰方法
WO2024121818A1 (en) * 2022-12-09 2024-06-13 Mesoblast International Sarl Method of treating heart failure in subjects with persistent inflammation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060051334A1 (en) * 1999-03-30 2006-03-09 Myocardial Therapeutics, Inc. Injection of bone marrow-derived conditioned medium for angiogenesis
US20060216276A1 (en) * 2005-03-07 2006-09-28 Mari Dezawa Use of materials for treatment of central nervous system lesions
US7605140B2 (en) * 2000-12-07 2009-10-20 Sangamo Biosciences, Inc. Regulation of angiogenesis with zinc finger proteins

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US20040071637A1 (en) 1993-04-27 2004-04-15 Elia James P. Method for repairing a damaged portion of a human organ
US6140466A (en) 1994-01-18 2000-10-31 The Scripps Research Institute Zinc finger protein derivatives and methods therefor
US6242568B1 (en) 1994-01-18 2001-06-05 The Scripps Research Institute Zinc finger protein derivatives and methods therefor
US20030044396A1 (en) 1998-04-21 2003-03-06 Elia James P. Methods for treating diseases and increasing longevity
ATE407205T1 (de) 1994-08-20 2008-09-15 Gendaq Ltd Verbesserung in bezug auf bindungsproteine bei der erkennung von dna
GB9824544D0 (en) 1998-11-09 1999-01-06 Medical Res Council Screening system
US5789538A (en) 1995-02-03 1998-08-04 Massachusetts Institute Of Technology Zinc finger proteins with high affinity new DNA binding specificities
US5925523A (en) 1996-08-23 1999-07-20 President & Fellows Of Harvard College Intraction trap assay, reagents and uses thereof
GB2338237B (en) 1997-02-18 2001-02-28 Actinova Ltd In vitro peptide or protein expression library
GB9703369D0 (en) 1997-02-18 1997-04-09 Lindqvist Bjorn H Process
GB9710807D0 (en) 1997-05-23 1997-07-23 Medical Res Council Nucleic acid binding proteins
GB9710809D0 (en) 1997-05-23 1997-07-23 Medical Res Council Nucleic acid binding proteins
US6410248B1 (en) 1998-01-30 2002-06-25 Massachusetts Institute Of Technology General strategy for selecting high-affinity zinc finger proteins for diverse DNA target sites
WO1999045132A1 (en) 1998-03-02 1999-09-10 Massachusetts Institute Of Technology Poly zinc finger proteins with improved linkers
US6140081A (en) 1998-10-16 2000-10-31 The Scripps Research Institute Zinc finger binding domains for GNN
US6453242B1 (en) 1999-01-12 2002-09-17 Sangamo Biosciences, Inc. Selection of sites for targeting by zinc finger proteins and methods of designing zinc finger proteins to bind to preselected sites
US6534261B1 (en) 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US7013219B2 (en) 1999-01-12 2006-03-14 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US6794136B1 (en) 2000-11-20 2004-09-21 Sangamo Biosciences, Inc. Iterative optimization in the design of binding proteins
AU2001226935B2 (en) 2000-01-24 2006-06-22 Gendaq Limited Nucleic acid binding polypeptides characterized by flexible linkers connected nucleic acid binding modules
US20020061512A1 (en) 2000-02-18 2002-05-23 Kim Jin-Soo Zinc finger domains and methods of identifying same
US7029847B2 (en) 2000-05-16 2006-04-18 Massachusetts Institute Of Technology Methods and compositions for interaction trap assays
US7067317B2 (en) 2000-12-07 2006-06-27 Sangamo Biosciences, Inc. Regulation of angiogenesis with zinc finger proteins
US7067617B2 (en) 2001-02-21 2006-06-27 The Scripps Research Institute Zinc finger binding domains for nucleotide sequence ANN
US20040197892A1 (en) 2001-04-04 2004-10-07 Michael Moore Composition binding polypeptides
US20030003090A1 (en) 2001-05-31 2003-01-02 Prockop Darwin J. Directed in vitro differentiation of marrow stromal cells into neural cell progenitors
JP3886346B2 (ja) 2001-06-22 2007-02-28 サンバイオ,インコーポレイティド 骨髄間質細胞由来Schwann細胞を含む神経再生用医薬組成物
EP1421177A4 (en) 2001-08-20 2006-06-07 Scripps Research Inst ZINC FINGER FASTENING DOMAINS FOR CNN
EP2270146A1 (en) 2002-02-06 2011-01-05 Sanbio, Inc. Method of inducing differentiation of bone marrow stromal cells to neural cells or skeletal muscle cells by introduction of notch gene
CN1973031B (zh) 2004-04-12 2011-11-02 桑比欧公司 具有神经元祖细胞特性的细胞
US20060083734A1 (en) * 2004-10-18 2006-04-20 Henrich Cheng Composition and method for repairing nerve damage and enhancing functional recovery of nerve
JP2009520463A (ja) 2005-11-28 2009-05-28 ザ スクリプス リサーチ インスティテュート Tnnのための亜鉛フィンガー結合ドメイン
US20070154989A1 (en) 2006-01-03 2007-07-05 The Scripps Research Institute Zinc finger domains specifically binding agc
WO2008010246A1 (en) 2006-07-20 2008-01-24 SAFILO SOCIETÀ AZIONARIA FABBRICA ITALIANA LAVORAZIONE OCCHIALI S.p.A. Resilient hinge device for spectacles, and spectacles comprising said device
WO2008102460A1 (en) 2007-02-23 2008-08-28 Sanbio Inc. Transplantation of bone marrow stromal cell-induced neural stem cells for promoting fuctional recovery after spinal cord injury
US8945919B2 (en) 2007-08-15 2015-02-03 San Bio, Inc. Methods and composition for treating neural degeneration
EP2215216B1 (en) 2007-12-03 2018-05-30 SanBio, Inc. Extracellular matrix from pluripotent cells
DK2222839T3 (en) 2007-12-03 2015-12-21 Sanbio Inc Methods and compositions for modulating differentiation of pluripotent cells
EP3851520A1 (en) * 2008-04-30 2021-07-21 SanBio, Inc. Neural regenerating cells with alterations in dna methylation
CN106692193B (zh) 2011-04-06 2021-01-05 桑比欧公司 用于调节外周免疫功能的方法和组合物
US20120263681A1 (en) 2011-04-12 2012-10-18 Fujifilm Corporation Composition comprising cell and biocompatible polymer
CN107513551A (zh) 2011-08-19 2017-12-26 桑比欧公司 神经源性因子和胶质生成性因子及其测定
EP2806881A1 (en) 2012-01-27 2014-12-03 SanBio, Inc. Methods and compositions for modulating angiogenesis and vasculogenesis

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060051334A1 (en) * 1999-03-30 2006-03-09 Myocardial Therapeutics, Inc. Injection of bone marrow-derived conditioned medium for angiogenesis
US7605140B2 (en) * 2000-12-07 2009-10-20 Sangamo Biosciences, Inc. Regulation of angiogenesis with zinc finger proteins
US20060216276A1 (en) * 2005-03-07 2006-09-28 Mari Dezawa Use of materials for treatment of central nervous system lesions

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Wu et al., Stem Cells 25(10):2648-2659, October 2007 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10245286B2 (en) 2012-01-27 2019-04-02 Sanbio, Inc. Methods and compositions for modulating angiogenesis and vasculogenesis
US11304982B2 (en) 2012-01-27 2022-04-19 Atricure, Inc. Methods and compositions for modulating angiogenesis and vasculogenesis
US9828593B2 (en) 2012-05-16 2017-11-28 Sanbio, Inc. Methods and compositions for treatment of traumatic brain injury and for modulation of migration of neurogenic cells
US10626389B2 (en) 2012-05-16 2020-04-21 Sanbio, Inc. Methods and compositions for modulation of migration of neurogenic cells
US9326999B2 (en) 2012-10-09 2016-05-03 Sanbio, Inc. Methods and compositions for treatment of retinal degeneration
US9855299B2 (en) 2012-10-09 2018-01-02 Sanbio, Inc. Methods and compositions for treatment of retinal degeneration
US20220387506A1 (en) * 2021-05-27 2022-12-08 Sanbio, Inc. Cell therapies and methods of treatment for small-volume stroke

Also Published As

Publication number Publication date
AU2021203467B2 (en) 2024-06-06
US10245286B2 (en) 2019-04-02
US11304982B2 (en) 2022-04-19
CN104254337A (zh) 2014-12-31
AU2021203467A1 (en) 2021-06-24
US20170258850A1 (en) 2017-09-14
AU2017272177B2 (en) 2019-05-16
CN112386681A (zh) 2021-02-23
EP4218727A2 (en) 2023-08-02
WO2013112917A1 (en) 2013-08-01
AU2019204305B2 (en) 2021-03-04
AU2013211934A1 (en) 2014-07-31
CA2862309A1 (en) 2013-08-01
EP2806881A1 (en) 2014-12-03
EP4218727A3 (en) 2023-08-30
WO2013112917A9 (en) 2014-11-13
US20160263159A1 (en) 2016-09-15
AU2024200552A1 (en) 2024-02-15
HK1199200A1 (en) 2015-06-26
JP2015504922A (ja) 2015-02-16
AU2019204305A1 (en) 2019-07-04
US20190167730A1 (en) 2019-06-06
US20220193144A1 (en) 2022-06-23
AU2017272177A1 (en) 2017-12-21
JP2016222739A (ja) 2016-12-28
AU2013211934B2 (en) 2017-09-14
JP6023218B2 (ja) 2016-11-09
JP6280180B2 (ja) 2018-02-14

Similar Documents

Publication Publication Date Title
US20220193144A1 (en) Methods and compositions for modulating angiogenesis and vasculogenesis
US10363285B2 (en) Methods and compositions for stimulation of cell proliferation and provision of biologically active mixtures of FGF2 isoforms
CN109718250B (zh) 用于治疗创伤性脑损伤和用于调节神经原性细胞迁移的方法和组合物
Perrucci et al. Cyclophilin A modulates bone marrow-derived CD117+ cells and enhances ischemia-induced angiogenesis via the SDF-1/CXCR4 axis
US20150297643A1 (en) Inhibition of rho and or rock and cell transplantation
WO2019027299A2 (ko) 간세포성장인자를 발현하는 중간엽줄기세포를 유효성분으로 포함하는 혈관계 질환 예방 또는 치료용 약학 조성물
KR102088767B1 (ko) 혼합물 4f를 이용한 줄기세포 생물학적 활성 증가용 조성물
KR101625901B1 (ko) 활성이 촉진된 줄기세포를 유효성분으로 포함하는 폐고혈압 치료용 약학적 조성물
EP4393499A1 (en) Small extracellular vesicles with antifibrotic properties
KR20240012562A (ko) 소규모 뇌졸중에 대한 세포 치료법 및 치료 방법

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANBIO, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DAO, MONIQUE;TATE, CIARA;CASE, CASEY;SIGNING DATES FROM 20130213 TO 20130226;REEL/FRAME:029926/0661

STCB Information on status: application discontinuation

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