USRE40811E1 - Peripheralization of hematopoietic stem cells - Google Patents

Peripheralization of hematopoietic stem cells Download PDF

Info

Publication number
USRE40811E1
USRE40811E1 US11/635,821 US63582193A USRE40811E US RE40811 E1 USRE40811 E1 US RE40811E1 US 63582193 A US63582193 A US 63582193A US RE40811 E USRE40811 E US RE40811E
Authority
US
United States
Prior art keywords
cells
stem cells
csf
vla
antibody
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.)
Expired - Lifetime
Application number
US11/635,821
Other languages
English (en)
Inventor
Thalia Papayannopoulou
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.)
University of Washington
Original Assignee
University of Washington
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=25525430&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE40811(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by University of Washington filed Critical University of Washington
Priority to US11/635,821 priority Critical patent/USRE40811E1/en
Application granted granted Critical
Publication of USRE40811E1 publication Critical patent/USRE40811E1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • C07K16/2842Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily against integrin beta1-subunit-containing molecules, e.g. CD29, CD49
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • 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
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Definitions

  • the invention relates to the manipulation of hematopoietic stem cells. More particularly, the invention relates to methods for increasing the number of hematopoietic stem cells in peripheral blood.
  • Hematopoietic stem cells are primitive, uncommitted progenitor cells that give rise to the lymphoid, myeloid and erythroid lineages of cells in blood.
  • the stem cell population constitutes only a small proportion of the total cells in bone marrow and represents even a far more minuscule proportion of the cells in peripheral blood.
  • Stem cells have commonly been characterized by their surface antigenic determinants.
  • Tsukamoto et al. U.S. Pat. No. 5,061,620 (1991)
  • a highly stem cell concentrated cell composition is CD34 + , CD10 ⁇ , CD19 ⁇ and CD33 ⁇ .
  • Leon et al., Blood 77:1218-1227 (1991) teaches that about one per cent of CD34 + cells, or about 0.01% of the total marrow cell population, do not express differentiation antigens, such as CD33 (myeloid lineage), CD71 (erythroid lineage) or CD10 and CD5 (lymphoid B and T lineage), and that reduced expression of CD34 expression during maturation is associated with increased expression of the differentiation antigens.
  • differentiation antigens such as CD33 (myeloid lineage), CD71 (erythroid lineage) or CD10 and CD5 (lymphoid B and T lineage
  • BMT bone marrow transplantation
  • ABMT autologous transplantation
  • M-CSF macrophage colony-stimulating factor
  • G-CSF granulocyte colony-stimulating factor
  • erythropoietin interleukins-1, -2, -3, -4 and -6
  • various interferons and tumor necrosis factors have enormous potential.
  • Another approach to autologous transplantation is to purify stem cells from peripheral blood using immunoaffinity techniques. These techniques hold promise not only for autologous stem cell transplantation in conjunction with chemotherapy, but also for gene therapy, in which purified stem cells are necessary for genetic manipulation to correct defective gene function, then reintroduced into the patient to supply the missing function.
  • immunoaffinity techniques hold promise not only for autologous stem cell transplantation in conjunction with chemotherapy, but also for gene therapy, in which purified stem cells are necessary for genetic manipulation to correct defective gene function, then reintroduced into the patient to supply the missing function.
  • Edgington, Biotechnology 10:1099-1106 (1992) teaches that current procedures require three separate four hour sessions to process enough cells in the absence of peripheralization. DePalma, Genetic Engineering News, Vol. 12, May 1, 1992, teaches that this can be improved by treatment with G-CSF for peripheralization.
  • Integrins are a large family of integral membrane glycoproteins having over 16 heterodimeric members that mediate interactions between cells, interactions between cells and the extracellular matrix, and interactions involved in embryonic development and regulation of T-cell responses.
  • the VLA-5 ( ⁇ 5 ⁇ 1 ) complex is widely distributed and functions as a receptor for fibronectin.
  • the VLA-4 ( ⁇ 4 ⁇ 1 ) complex is expressed at substantial levels on normal peripheral blood B and T cells, thymocytes, monocytes, and some melanoma cells an well as on marrow blast cells and erythroblasts.
  • Ligands for VLA-4 are vascular cell adhesion molecule-1(VCAM-1) and CS-1, an alternately spliced domain within the Hep II region of fibronectin.
  • Another group of integrins CDIIa/CD18, CDIIb/CD18, and CDIIc/CD18
  • CDIIa/CD18, CDIIb/CD18, and CDIIc/CD18 share the common ⁇ 2 chain and are variably expressed on peripheral T cells, monocytes, and mature granulocytes.
  • Ligands for ⁇ 2 -integrins include members of the Ig superfamily (ICAM-1 and ICAM-2) found on activated endothelial cells.
  • TA-2 a monoclonal antibody to rat VLA-4, inhibits the in vivo migration, of small peritoneal exudate lymphocytes and lymphocytes from peripheral lymph nodes, from the blood across the vascular endothelium to sites of inflammation. This document also observes that systemic treatment of rats with TA-2 was accompanied by an increase in total blood lymphocyte count.
  • peripheralizing stem cells both for scientific investigatory purposes for understanding the processes of peripheralization and homing, and for the development of better methods of peripheralization for autologous stem cell transplantation in the course of cancer treatment or gene therapy.
  • such methods should produce even higher levels of stem cells in peripheral blood than existing methods provide.
  • the invention provides a novel method for increasing the number of hematopoietic stem cells and CD34 + cells in peripheral blood, which is also known as “peripheralization” or “mobilization” of hematopoietic stem cells and CD34 + cells.
  • This method comprises the step of administering a blocking agent of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells.
  • anti-VLA-4 or anti-VCAM-1 antibodies which may optionally be single chain, humanized or chimeric, Fab, Fab′, F(ab′) 2 or F(v) fragments thereof, heavy or light chain monomers or dimers thereof, or intermixtures of the same, soluble fibronectin, CS-1 peptides or fibronectin peptides containing the amino acid sequence EILDV or conservatively substituted amino acid sequences, or soluble VCAM-1, bifunctional VCAM-1/Ig fusion proteins or VCAM-1 peptides.
  • the invention provides a novel method for peripheralizing hematopoietic stem cells and CD34 + cells with more predictable greater effectiveness than cytokine treatment alone provides.
  • the method comprises administering a blocking agent of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells, as in the first aspect of the invention, in combination with a stimulating agent of hematopoietic stem cell proliferation.
  • the step of administering a stimulating agent of hematopoietic stem cell proliferation can be carried out by using a cytokine, preferably G-CSF, stem cell factor, totipotent stem cell factor, stem cell proliferation factor or GM-CSF, but alternatively M-CSF, erythropoietin, interleukins-1, -2, -3, -4, -6, or 11.
  • a cytokine preferably G-CSF, stem cell factor, totipotent stem cell factor, stem cell proliferation factor or GM-CSF, but alternatively M-CSF, erythropoietin, interleukins-1, -2, -3, -4, -6, or 11.
  • the invention provides an improved method of transplanting peripheral blood stem cells into a patient who has undergone chemotherapy or radiotherapy for cancer.
  • stem cells prior to the administration of myeloablative chemotherapy or radiotherapy, stem cells are peripheralized from the patient's bone marrow by administration of an agent that mediates blocking of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells.
  • This agent may be administered alone, or preferably in conjunction with an agent that stimulates proliferation of stem cells.
  • the peripheralized stem cells are then collected from peripheral blood by leukapheresis. Stem cells are then enriched from the collected peripheralized blood by immunoadsorption using anti-CD34 antibodies.
  • the invention provides an improved method for carrying out gene therapy in patients having various genetic and acquired diseases.
  • stem cells are peripheralized from the patient's bone marrow by administration of an agent that mediates blocking of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells.
  • this agent may be administered alone or in conjunction with an agent that stimulates proliferation of stem cells.
  • Peripheral blood is then collected by leukapheresis.
  • Stem cells are then enriched from the collected peripheral blood by immunoadsorption using anti-CD34 antibodies.
  • the enriched stem cells are then expanded ex vivo by culturing them in the presence of agents that stimulate proliferation of stem cells.
  • the enriched and optionally expanded stem cells are then transduced with an amphotrophic retroviral vector, or other suitable vectors, that expresses a gene that ameliorates the genetic or acquired disease.
  • the vector may also carry an expressed selectable marker, in which case successfully transduced cells may be selected for the presence of the selectable marker.
  • the transduced and optionally selected stem cells are then returned to the patient's circulating blood and allowed to engraft themselves into the bone marrow.
  • the invention satisfies each of these objects by providing a method for peripheralizing stem cells and CD34 + cells by administering a blocking agent of VLA-4 antigen on the surface of hematopoietic stem cells.
  • This effect can be increased by the use of such blocking agents in conjunction with approaches to amplify stem cells to produce a synergistic effect.
  • FIG. 1A is a profile of total white blood cells and CFU in peripheral blood before treatment of macaques.
  • FIG. 1B is a profile of total white blood cells and CFU in peripheral blood of a baboon after treatment with anti-VLA-4 antibodies.
  • FIG. 1C is a profile of total white blood cells and CFU in peripheral blood of a macaque after treatment with anti-VLA-4 antibodies.
  • FIG. 2 is a profile of total white blood cells and CFU in peripheral blood before treatment of an animal with the anti-CD18 monoclonal antibody 60.3. All symbols are as defined above.
  • FIG. 3B is a profile of the results for a control animal treated with GCSF alone.
  • FIG. 4A shows high proliferative potential (HPP) progenitors (colonies over 0.5 mm in diameter of compact growth) resulting from combined treatment with GCSF and HP1/2 antibody.
  • HPP high proliferative potential
  • FIGS. 5A and 5B are the nucleotide sequences encoding the variable heavy region of the heavy and light chains of anti-VLA-4 murine monoclonal antibody HP 1/2.
  • FIG. 6A is a profile of the results of combined treatment with 5-fluorouracil and anti-VLA-4 murine monoclonal antibody HP1/2. Symbols are as described for FIG. 3 .
  • FIG. 6B is a profile of the results of 5-fluorouracil treatment alone.
  • FIG. 7A is the nucleotide sequences of the V H -encoding regions having CDR-encoding sequences from murine HP1/2 transplanted therein (SEQ ID NO:3).
  • FIG. 7B is the nucleotide sequence of the transplanted V K sequence (SEQ ID NO:4).
  • FIG. 8B is the nucleotide sequence encoding the V K region (SEQ ID NO:6).
  • FIG. 10 is a profile of the results of treatment with murine Fab fragments of anti-VLA-4 antibody HP1/2. Symbols are as described for FIG. 3 .
  • the invention relates to the manipulation of hematopoietic stem cells. More particularly, the invention relates to the peripheralization of hematopoietic stem cells and other CD34 + cells.
  • this invention provides a method for peripheralizing hematopoietic stem cells and CD34 + cells, comprising the step of administering a blocking agent of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells.
  • a blocking agent of VLA-4 antigens is intended to mean an agent that is capable of interfering with interactions between VLA-4 antigens and either VCAM-1 or fibronectin on the surface of stromal cells or in the extracellular matrix (ECM).
  • ECM extracellular matrix
  • any agent that can block VLA-4 antigens can be successfully used in the method of this invention.
  • any agent capable of blocking VLA-4 antigens on the surface of hematopoietic stem cells is considered to be an equivalent of the monoclonal antibody used in the examples herein.
  • this invention contemplates as equivalents at least peptides, peptide mimetics, carbohydrates and small molecules capable of blocking VLA-4 antigens on the surface of CD34 + cells or hematopoietic stem cells.
  • antibody and antibody derivative blocking agents according to this invention include embodiments having binding specificity for VCAM-1 or fibronectin, since these molecules appear to either be important in the adhesion between stem cells and stromal cells or the extracellular matrix or interfere with traffic of stem cells through other tissues and blood.
  • the blocking agents used in the method according to this invention are not antibodies or antibody derivatives, but rather are soluble forms of the natural binding proteins for VLA-4.
  • These blocking agents include soluble VCAM-1, bifunctional VCAM-1/Ig fusion proteins, or VCAM-1 peptides as well as fibronectin, fibronectin having an alternatively spliced non-type III connecting segment and fibronectin peptides containing the amino acid sequence EILDV or a similar conservatively substituted amino acid sequence.
  • These blocking agents will act by competing with the stromal cell- or ECM-bound binding protein for VLA-4 on the surface of stem cells.
  • blocking agents are preferably administered parenterally.
  • the blocking agents are preferably administered as a sterile pharmaceutical composition containing a pharmaceutically acceptable carrier, which may be any of the numerous well known carriers, such as water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, or combinations thereof.
  • a pharmaceutically acceptable carrier such as water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, or combinations thereof.
  • the blocking agent if an antibody or antibody derivative, will be administered at a dose between about 0.1 mg/kg body weight/day and about 10 mg/kg body weight/day.
  • the dose range should preferably be between molar equivalent amounts to these amounts of antibody. Optimization of dosages can be determined by administration of the blocking agents, followed by CFU-GM assay of peripheral blood, or assay of CD34 + cells in peripheral blood. The preferred dosage should produce an increase of at least 10-fold in the
  • the present invention provides a method for peripheralizing hematopoietic stem cells that is far more effective than cytokine treatment alone.
  • the method comprises the step of administering a blocking agent of VLA-4 antigens on the surface of hematopoietic stem cells in combination with the step of administering a stimulating agent of hematopoietic stem cell proliferation in vivo.
  • the step of administering a blocking agent of VLA-4 antigens on the surface of hematopoietic stem cells is carried out in exactly the same fashion that is described for the first apsect of the invention.
  • the step of administering a stimulating agent of hematopoietic stem cell proliferation in vivo is preferably carried out through the administration of cytokines.
  • cytokines for stimulating hematopoietic stem cells to proliferate include granulocyte colony-stimulating factor (G-CSF), stem cell factor, totipotent stem cell factor (TSCF), stem cell proliferation factor (SCPF), granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), erythropoietin, interleukin-1, -2, -3, -4, -6, and -11.
  • G-CSF granulocyte colony-stimulating factor
  • stem cell factor stem cell factor
  • TSCF totipotent stem cell factor
  • SCPF stem cell proliferation factor
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • M-CSF macrophage colony-stimulating factor
  • erythropoietin interleukin-1, -2, -3, -4, -6, and -11.
  • Most preferred are G-
  • G-CSF and GM-CSF to stimulate proliferation of progenitors is well established (see, e.g., Metcalf, Nature 339:27-30 (1989)), as is their ability to cause peripheralization of hematopoietic stem cells (see, e.g., Haas et al., Exp. Hematol. 18:94-98 (1990) and Blood 72:2074 (1988). This ability has also been established for stem cell factor (Andrews et al., Blood 80:920-927 (1992)). In addition, the enormous potential of these other cytokines identified herein has been recognized (see Rowe and Rapoport, J. Clin. Pharmacol. 32:486-501 (1992)).
  • stimulation of hematopoietic stem cells to proliferate can be carried out by any cytokine that is capable of mediating such proliferation in vivo.
  • any cytokine that can stimulate hematopoietic stem cells to proliferate in vivo is considered to be equivalent to G-CSF, stem cell factor and GM-CSF, which are also considered to be equivalent to each other.
  • the use of chemotherapeutic agents alone can lead to the peripheralization of progenitors.
  • Such agents can also be combined with VLA-4 blocking agents in the method according to the present invention.
  • cytokines are preferably administered parenterally.
  • the cytokines are preferably administered as a sterile pharmaceutical composition containing a pharmaceutically acceptable carrier, which may be any of the numerous well known carriers, such as water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, or combinations thereof.
  • a pharmaceutically acceptable carrier such as water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, or combinations thereof.
  • the cytokine if G-CSF, will be administered at a dose between about 1 ⁇ g/kg body weight/day and about 50 ⁇ g/kg body weight/day, most preferably at about 10-15 ⁇ g/kg body weight/day.
  • cytokines will be administered over a course of from about four to about ten days. Optimization of dosages or the combination of cytokines (e.g., G-CSF and kit ligand) can be determined by administration of the cytokine and administration of the blocking agents, followed by CFU-GM assay of peripheral blood. The preferred dosage should produce an increase of at least 5-fold in the CFU-GM counts per milliliter of peripheral blood, compared with cytokines alone.
  • cytokines e.g., G-CSF and kit ligand
  • the step of administering a blocking agent of VLA-4 antigens on the surface of hematopoietic stem cells or CD34 + cells and the step of administering stimulating agents for proliferation of these cells can be carried out concomitantly or sequentially.
  • the steps are carried out sequentially, preferably administering stimulating agents of CD34 + or hematopoietic stem cell proliferation being the first step.
  • this invention provides an improved method of transplanting peripheral blood stem cells into a patient who has undergone chemotherapy or radiotherapy for cancer.
  • stem cells prior to the administration of chemotherapy or radiotherapy, stem cells are peripheralized from the patient's bone marrow by administration of an agent that mediates blocking of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells.
  • the blocking agents used in this method are preferably selected from those blocking agents described in the discussion of the first aspect of the invention. This agent may be administered alone, or in conjunction with an agent that stimulates proliferation of stem cells.
  • the proliferation stimulating agents optionally used in this method are preferably selected from those proliferation stimulating agents described in the discussion of the second aspect of the invention.
  • the peripheralized stem cells are then collected from peripheral blood by leukapheresis.
  • Stem cells are then enriched from the collected peripheralized blood by CD34 affinity chromatography such as immunoadsorption using anti-CD34 antibodies.
  • CD34 affinity chromatography such as immunoadsorption using anti-CD34 antibodies.
  • Such stem cell enrichment is known in the art and has been described, for example, by Berenson, Transplantation Proceedings 24:3032-3034 (1992) and the references cited therein.
  • the enriched stem cells are then expanded ex vivo by culturing them in the presence of agents that stimulate proliferation of stem cells. This ex vivo expansion can be carried out using, alone or in combination, any of the proliferation stimulating agents described in the discussion of the second aspect of the invention.
  • peripheralized stem cells for transplantation after chemotherapy or radiotherapy for cancer
  • the value of using peripheralized stem cells for transplantation after chemotherapy or radiotherapy for cancer is recognized in the art and has been described in numerous references, including Bensinger et al., Blood 81:3158-3163 (1993); Chao et al., 81:2031-2035 (1993); Kessinger and Armitage, Blood 77:211-213 (1991); Gale et al., Bone Marrow Transplantation 9:151-155 (1992); and Siena et al., Blood 74:1904-1914 (1989).
  • the present method according to the invention provides an improvement in the transplantation of stem cells from peripheral blood by increasing the concentration of such stem cells in the peripheral blood, thereby greatly improving the likelihood of success of the transplantation.
  • the present invention provides an improved method of transplanting purified peripheral blood stem cells into a patient who has undergone myeloablative chemotherapy or radiotherapy for AIDS.
  • This method involves the same steps as described for transplanting peripheralized stem cells into a patient who has undergone chemotherapy or radiotherapy for cancer.
  • this method further optionally involves administration to the patient of anti-HIV agents, such as antivirals such as AZT, soluble CD4, and CD4-directed blockers of the AIDS virus or antisense or antigene oligonucleotides, both before and after the return of the enriched and optionally expanded stem cells to the patient's circulating blood.
  • This step serves a “mopping up” function to prevent residual virus from infecting the progeny of the newly returned stem cells.
  • the myeloablative chemotherapy or radiotherapy will generally be expected to destroy any cells in the blood that are infected by HIV.
  • the “mopping up” step thus serves to remove any residual virus that otherwise could possibly infect the progeny of the stem cells transplanted into the patient after such therapy.
  • agents can be useful in such a “mopping up” step.
  • CD4-directed anti-HIV agents and analogs have been shown to prophylactically prevent infection of uninfected CD34 + cells by HIV.
  • anti-HIV oligonucleotides have been shown to prevent HIV infection of uninfected cells, for example in U.S. Pat. No. 4,806,463, the teaching of which are hereby incorporated by reference.
  • this invention provides an improved method for carrying out gene therapy in patients having any of a variety of genetic and acquired diseases.
  • stem cells are peripheralized from the patient's bone marrow by administration of an agent that mediates blocking of VLA-4 antigens on the surface of hematopoietic stem cells and CD34 + cells.
  • the blocking agents used in this method are preferably selected from those blocking agents described in the discussion of the first aspect of the invention.
  • this agent may be administered alone or in conjunction with an agent that stimulates proliferation of stem cells.
  • the proliferation stimulating agent optionally used in this method is preferably selected from those proliferation stimulating agents described in the discussion of the second aspect of the invention. Peripheral blood is then collected by leukapheresis.
  • Stem cells are then enriched from the collected peripheral blood by immunoadsorption using anti-CD34 antibodies.
  • Such stem cell enrichment is known in the art and has been described, for example, by Berenson, Transplantation Proceedings 24:3032-3034 (1992) and the references cited therein.
  • the enriched stem cells are then expanded ex vivo by culturing them in the presence of agents that stimulate proliferation of stem cells. This ex vivo expansion can be carried out using, alone or in combination, any of the proliferation stimulating agents described in the discussion of the second aspect of the invention.
  • Such ex vivo expansion of CD34 + cells from peripheral blood is known in the art and has been described, for example, by Bruggar et al., Blood 81:2579-2584 (1993).
  • the enriched and optionally expanded stem cells are then infected with an amphotrophic retroviral vector, or other appropriate vector, that expresses a gene that ameliorates the genetic or acquired disease.
  • the vector may also carry an expressed selectable marker, in which case successfully transduced cells may be selected for the presence of the selectable marker.
  • the transduced and optionally selected stem cells are then returned to the patient's circulating blood and allowed to engraft themselves into the bone marrow.
  • the present method according to the invention provides an improvement in the transplantation of stem cells from peripheral blood by increasing the concentration of such stem cells in the peripheral blood, thereby greatly improving the likelihood of success of the retroviral transfection and subsequent transplantation and allows for repeated administration of genetically engineered cells in patients with partially ablative regimens and receiving agents that promote proliferation of transduced cells.
  • stem cell enrichment is known in the art and has been described, for example, by Berenson, Transplantation Proceedings 24:3032-3034 (1992) and the references cited therein.
  • the instant invention is useful for many purposes.
  • the methods of peripheralizing hematopoietic stem cells or CD34 + cells is of value in scientific research dedicated to understanding the molecular interactions and molecular signals involved in the homing of these cells to bone marrow, as well as their trafficking in response to certain infections and trauma.
  • This invention also provides sources of peripheral blood that is enriched in CD34 + and hematopoietic stem cells, thus making the methods of the invention useful for therapeutic applications involving autologous transplantation of these cell types following chemotherapy or radiotherapy or in the course of gene therapy.
  • the present invention provides many advantages over the current exclusively cytokine-based techniques.
  • peripheralization can be obtained without risk of cytokine-induced cell differentiation of normal cells or proliferation of contaminating leukemia cells and can be combined with cytotoxic agents.
  • the timing of the peak of progenitors in peripheral blood is consistently between about 24 and about 72 hours from first injection of antibody, thus making the most beneficial timing for leukapheresis more predictable.
  • the well known CFU-GM assay is the most widely used measure of the hematopoietic progenitor viability of a PBSC harvest and correlates well with per cent CD34 + cells present in peripheral blood (see Craig et al., Blood Reviews 6:59-67 (1992)).
  • a blocking agent of VLA-4 antigen on the surface of hematopoietic stem cells and CD34 + cells results in peripheralization of the hematopoietic stem cells and CD34 + cells.
  • monoclonal antibodies against VLA-4 were administered to a macaque after five days of treatment with G-CSF. It is well known that G-CSF can stimulate hematopoietic stem cells and CD34 + cells in vivo (see Metcalf, Nature 339:27-30 (1989)). G-CSF alone caused an increase in CFU-GM present in peripheral blood by days 4 and 5 of treatment. After discontinuation of G-CSF treatment and commencement of treatment with anti-VLA-4 antibodies, the number of CFU-GM in peripheral blood increased even more dramatically.
  • the antigen CD18 is present on stem cells and is widely believed to be important in interactions involving stem cells.
  • Another macaque was treated with a monoclonal antibody against CD18.
  • Antibody was delivered by intravenous injection for three days at a dosage of 2 mg/kg of body weight/day. The results of this control experiment are shown in FIG. 2 .
  • Total white blood cell counts did increase with this treatment, consistent with previous experiments with rabbits.
  • total GFU-GM showed no increase after treatment with anti-CD18 monoclonal antibodies.
  • blocking agents of CD18 do not lead to peripheralization of stem cells or progenitor cells.
  • a baboon was treated with recombinant human G-CSF twice daily for five consecutive days.
  • Each G-CSF treatment consisted of intravenous injection of 15 micrograms G-CSF per kilogram of body weight. After the five days of G-CSF administration, the baboon received two injections, spaced one day apart, of anti-VLA-4 monoclonal antibody (HP1/2). Each injection contained 1 milligram antibody per kilogram body weight.
  • Total white blood cells and CFU-GM were determined as described in Example 1. The results are shown in FIG. 3 . As shown in panel A of that figure, G-CSF resulted in the expected increase in CFU-GM by days 4 and 5 of treatment, along with a marked increase in total white blood cells.
  • HPP cells are cells that give rise to colonies that are macroscopically visible, over 0.5 mm in diameter with dense, compact growth on the analysis grid. Presence of these cells is associated with greater repopulation capacity and such cells are believed to be earlier progenitors.
  • FIG. 4 The observed disparity in peripheral blood HPP cells between G-CSF treatment alone and G-CSF treatment in combination with anti-VLA-4 antibodies is even greater than the disparity observed for CFU-GM.
  • a baboon was treated with the chemotherapeutic agent 5-fluorouracil at a dosage of 100 mg per kilogram body weight. Beginning five days later, the baboon received four injections, spaced one day apart, of anti-VLA-4 monoclonal antibody (HP1/2). Each injection contained one milligram antibody per kilogram body weight.
  • Total white blood cells and CFU-GM were determined as described in Example 1. The results are shown in FIG. 6 . As shown in panel B of that figure, 5-fluorouracil alone produced a modest increase in CFU-GM at days 11 and 12. Administration of anti-VLA-4 antibody after the 5-fluorouracil, however, resulted in a dramatic further increase in CFU-GM, an increase of greater than ten times that produced by 5-fluorouracil alone.
  • CDRs complementarity determining regions
  • the complementarity determining regions (CDRs) of the light and heavy chains of the anti-VLA-4 monoclonal antibody HP1/2 were determined according to the sequence alignment approach of Kabat et al., 1991, 5th Ed., 4 vol., Sequences of Proteins of Immunological Interest, U.S. Department of Health and Human Services, NIH, U.S.A.
  • the CDRs of murine HP1/2 V H correspond to the residues identified in the humanized V H sequences disclosed herein as amino acids 31-35 (CDR1), 50-66 (CDR2) and 99-110 (CDR3), which respectively correspond to amino acids 31-35, 50-65 and 95-102 in the Kabat alignment.
  • the CDRs of murine HP1/2 V K correspond to the residues identified in the humanized V K sequences disclosed herein as amino acids 24-34 (CDR1), 50-56 (CDR2) and 89-97 (CDR3), and to the same residues in the Kabat alignment.
  • the Kabat NEWM framework was chosen to accept the heavy chain CDRs and the Kabat REI framework was chosen to accept the kappa chain CDRs.
  • Transplantation of the CDRs into the human frameworks was achieved by using M13 mutagenesis vectors and synthetic oligonucleotides containing the HP1/2 CDR-encoding sequences flanked by short sequences derived from the frameworks.
  • the V H mutagenesis vector, M13VHPCR1 contains the NEWM framework and has been described by Orlandi et al., Proc. Natl. Acad. Sci U.S.A. 86:3833-3837 (1989).
  • the V K mutagenesis vector, M13VKPCR2 contains essentially the REI framework and is identical to the M13 VKPCR1 vector described by Orlandi et al., except that there is a single amino acid change from Val to Glu in framework 4.
  • Transplanted product was recovered by PCR and cloned into M13mp19 for sequencing.
  • the transplanted V H sequence [SEQ. ID NO:3] is shown in FIG. 7 , panel A.
  • this product encodes the murine amino acids at positions 27-30 and an Arg to Asp change at position 94.
  • the transplanted V K sequence [SEQ. ID NO:4] is shown in FIG. 7 , panel B.
  • the entire V H and V K regions of humanized HP1/2 were cloned into appropriate expression vectors.
  • the appropriate human IgG1, IgG4or kappa constant region was then added to the vector in appropriate reading frame with respect to the murine variable regions.
  • the vectors were then cotransduced into YB2/0 ray myeloma cells (available from ATCC), which were then selected for the presence of both vectors.
  • ELISA analysis of cell supernatants demonstrated that the humanized antibody produced by these cells was at least equipotent with murine HP1/2.
  • the cell line expressing this humanized antibody was deposited with the ATCC on Nov. 3, 1992 and given accession number CRL 11175.
  • Fab fragments from the murine antibody HP1/2 were tested for their ability to peripheralize stem cells and progenitor cells.
  • the experiment was performed by administration of 1 mg/kg of Fab fragment twice daily for three days.
  • a modest effect (compared with humanized or monoclonal antibody) was observed, due to the rapid clearance of Fab fragments.
  • the observed characteristic BFU-e increase validates this result.
  • anti-VLA-4 antibody Fab fragments are capable of causing peripheralization of stem cells and progenitor cells. This suggests that anti-VLA-4 Fab fragments may be capable of acting synergistically in combination with G-CSF for peripheralizing stem cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Communicable Diseases (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US11/635,821 1992-11-13 1993-11-15 Peripheralization of hematopoietic stem cells Expired - Lifetime USRE40811E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/635,821 USRE40811E1 (en) 1992-11-13 1993-11-15 Peripheralization of hematopoietic stem cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US97770292A 1992-11-13 1992-11-13
PCT/US1993/011060 WO1994011027A1 (fr) 1992-11-13 1993-11-15 Peripherisation de cellules souches hematopoietiques
US11/635,821 USRE40811E1 (en) 1992-11-13 1993-11-15 Peripheralization of hematopoietic stem cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08436339 Reissue 1995-07-13

Publications (1)

Publication Number Publication Date
USRE40811E1 true USRE40811E1 (en) 2009-06-30

Family

ID=25525430

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/635,821 Expired - Lifetime USRE40811E1 (en) 1992-11-13 1993-11-15 Peripheralization of hematopoietic stem cells
US08/436,339 Ceased US5843438A (en) 1992-11-13 1993-11-15 Peripheralization of hematopoietic stem cells
US08/463,298 Expired - Lifetime US5824304A (en) 1992-11-13 1995-06-05 Peripheralization of hematopoietic stem cells
US08/463,128 Expired - Lifetime US5695755A (en) 1992-11-13 1995-06-05 Peripheralization of hematopoietic stem cells

Family Applications After (3)

Application Number Title Priority Date Filing Date
US08/436,339 Ceased US5843438A (en) 1992-11-13 1993-11-15 Peripheralization of hematopoietic stem cells
US08/463,298 Expired - Lifetime US5824304A (en) 1992-11-13 1995-06-05 Peripheralization of hematopoietic stem cells
US08/463,128 Expired - Lifetime US5695755A (en) 1992-11-13 1995-06-05 Peripheralization of hematopoietic stem cells

Country Status (11)

Country Link
US (4) USRE40811E1 (fr)
EP (1) EP0670735B1 (fr)
JP (2) JP3731892B2 (fr)
AT (1) ATE150975T1 (fr)
AU (1) AU689454B2 (fr)
CA (1) CA2148712C (fr)
DE (1) DE69309487T2 (fr)
DK (1) DK0670735T3 (fr)
ES (1) ES2102196T3 (fr)
GR (1) GR3023975T3 (fr)
WO (1) WO1994011027A1 (fr)

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994011027A1 (fr) * 1992-11-13 1994-05-26 Board Of Regents Of University Of Washington Peripherisation de cellules souches hematopoietiques
EP0678122B1 (fr) * 1993-01-12 1999-07-28 Biogen, Inc. Molecules d'anticorps anti-vla4 de recombinaison
EP0682529B2 (fr) * 1993-02-09 2005-12-28 Biogen Idec MA, Inc. Anticorps pour le traitement du diabete insulino-dependent
US5677291A (en) * 1993-12-10 1997-10-14 Hoechst Marion Roussel, Inc. Method of lowering serum cholesterol levels with 2,6-di-alkyl-4-silyl-phenols
US5608095A (en) * 1996-04-30 1997-03-04 Hoechst Marion Roussel, Inc. Alkyl-4-silyl-phenols and esters thereof as antiatherosclerotic agents
US5795876A (en) * 1996-04-30 1998-08-18 Hoechst Marion Rousssel, Inc. Method of inhibiting vascular cell adhesion molecule-1 and treating chronic inflammatory diseases with 2, 6-di-alkyl-4-silyl-phenols
US5928638A (en) * 1996-06-17 1999-07-27 Systemix, Inc. Methods for gene transfer
JP2002226377A (ja) * 1996-10-14 2002-08-14 Kirin Brewery Co Ltd 配糖体含有末梢血幹細胞増加剤
US6114572A (en) * 1996-11-20 2000-09-05 Hoechst Marion Roussel, Inc. Substituted phenols and thiophenols useful as antioxidant agents
US6121463A (en) * 1997-06-24 2000-09-19 Hoechst Marion Roussel, Inc. Alkyl-4-silylheterocyclic phenols and thiophenols useful as antioxidant agents
US6133467A (en) * 1997-06-25 2000-10-17 Hoechst Marion Roussel, Inc. 2,6-di-t-butyl-4-[(dimethyl-4-methoxyphenylsilyl)-methyl-oxy]phenol and 2,6-di-t-butyl-4-[(dimethyl-2-methoxy-phenylsilyl)methyloxy]phenol
US20010038834A1 (en) * 1998-01-26 2001-11-08 Gary Balian Bone cancer therapy
AU757873B2 (en) 1998-09-14 2003-03-06 Board Of Regents, The University Of Texas System Methods of treating multiple myeloma and myeloma-induced bone resorption using integrin antagonists
US7618630B2 (en) * 1998-09-14 2009-11-17 Board Of Regents, The University Of Texas System Methods of treating multiple myeloma and myeloma-induced bone resorption using integrin antagonists
US7838539B2 (en) * 1999-07-28 2010-11-23 The Board Of Trustees Of The Leland Stanford Junior University Nicotine receptor agonists in stem cell and progenitor cell recruitment
CA2384948C (fr) * 1999-09-14 2013-07-16 Biogen, Inc. Therapies contre l'insuffisance renale chronique a l'aide d'un ou de plusieurs antagonistes de l'integrine
US20060115473A1 (en) * 2000-12-14 2006-06-01 Biogen Idec Ma Inc., A Massachusetts Corporation Methods of treating central nervous system ischemic or hemorrhagic injury using anti alpha4 integrin antagonists
JP3459609B2 (ja) * 2000-03-17 2003-10-20 三洋電機株式会社 ライトキャッシュ回路、ライトキャッシュ回路を備えた記録装置、およびライトキャッシュ方法
WO2002037968A1 (fr) * 2000-11-10 2002-05-16 The Board Of Trustees Of The Leland Stanford Junior University Methodes de traitement des troubles associes a une insuffisance neuronale avec des cellules derivees de la moelle osseuse
US20040115175A1 (en) * 2000-11-10 2004-06-17 The Board Of Trustees Of The Leland Methods for treating disorders of neuronal deficiency with bone marrow-derived cells
US7994144B2 (en) * 2001-07-10 2011-08-09 Johnson & Johnson Research Pty, Limited Process for the preparation of a composition of genetically modified hematopoietic progenitor cells
BRPI0211570B8 (pt) 2001-07-31 2021-05-25 Anormed Inc uso de 1'-[1,4-fenileno-bis-(metileno)-bis-1,4,8,11- tetrazaciclotetradecano ou um sal farmaceuticamente aceitável do mesmo na preparação de uma composição para coletar células progenitoras e/ou tronco
US20040258673A1 (en) * 2003-04-03 2004-12-23 Hirose Thomas Gordon Elective collection and banking of autologous peripheral blood stem cells
CA2545248A1 (fr) * 2003-09-29 2005-04-14 The Regents Of The University Of California Procedes permettant de modifier l'adhesion, la differenciation et la migration de cellules progenitrices hematopoietiques
JP2007536936A (ja) 2004-05-14 2007-12-20 ベクトン・ディキンソン・アンド・カンパニー 幹細胞集団および使用方法
CN1993460A (zh) * 2004-07-12 2007-07-04 索林集团意大利有限公司 用于培养人细胞的装置和方法
CA2478458A1 (fr) * 2004-08-20 2006-02-20 Michael Panzara Traitement de la sclerose en plaques pediatrique
US20090202527A1 (en) * 2004-11-19 2009-08-13 Biogen Idec Ma Inc. Treatment for multiple sclerosis
MX2011010971A (es) * 2009-04-17 2012-01-19 Biogen Idec Inc Composiciones y metodos para tratar leucemia mielogena aguda.
CA2793959C (fr) 2010-03-25 2019-06-04 Oregon Health & Science University Glycoproteines du cmv et vecteurs recombines
EP3466977B1 (fr) 2010-04-16 2022-01-05 Biogen MA Inc. Anticorps anti-vla-4
WO2012170765A2 (fr) 2011-06-10 2012-12-13 Oregon Health & Science University Glycoprotéines de cmv et vecteurs recombinants cmv
CA2789539A1 (fr) 2011-09-12 2013-03-12 International Aids Vaccine Initiative Immunoselection de virus de la stomatite vesiculeuse recombinant exprimant des proteines hiv-1 en neutralisant largement les anticorps
US9402894B2 (en) 2011-10-27 2016-08-02 International Aids Vaccine Initiative Viral particles derived from an enveloped virus
ES2631608T3 (es) 2012-06-27 2017-09-01 International Aids Vaccine Initiative Variante de la glicoproteína Env del VIH-1
EP2848937A1 (fr) 2013-09-05 2015-03-18 International Aids Vaccine Initiative Procédés d'identification de nouveaux immunogènes du VIH-1
US10058604B2 (en) 2013-10-07 2018-08-28 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
EP3069730A3 (fr) 2015-03-20 2017-03-15 International Aids Vaccine Initiative Trimères de glycoprotéines de l'enveloppe du vih-1 soluble
US9931394B2 (en) 2015-03-23 2018-04-03 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
US9925258B2 (en) 2015-10-02 2018-03-27 International Aids Vaccine Initiative Replication-competent VSV-HIV Env vaccines

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0330506A2 (fr) 1988-02-26 1989-08-30 Dana Farber Cancer Institute Protéines VLA
WO1991003252A1 (fr) 1989-09-01 1991-03-21 Fred Hutchinson Cancer Research Center Inhibition de l'adhesion de lymphocytes sur l'endothelium vasculaire au moyen d'une nouvelle interaction entre le recepteur matriciel extra-cellulaire et son ligand
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
EP0455482A2 (fr) 1990-05-01 1991-11-06 Becton, Dickinson and Company Subliguée de cellules progénitrices humaines
US5206345A (en) 1990-08-02 1993-04-27 Fred Hutchinson Cancer Research Center Il-4 and tnf induce mab 6g10-recognized expression on bone marrow stromal cells
WO1993013798A1 (fr) 1992-01-13 1993-07-22 Biogen, Inc. Traitement de l'asthme
WO1993015764A1 (fr) 1992-02-12 1993-08-19 Biogen, Inc. Traitement de l'inflammation intestinale
WO1994017828A2 (fr) 1993-02-09 1994-08-18 Biogen, Inc. Traitement du diabete insulino-dependant
US5695755A (en) * 1992-11-13 1997-12-09 Papayannopoulou; Thalia Peripheralization of hematopoietic stem cells
US5871734A (en) 1992-01-13 1999-02-16 Biogen, Inc. Treatment for asthma with VLA-4 blocking agents
US5932214A (en) 1994-08-11 1999-08-03 Biogen, Inc. Treatment for inflammatory bowel disease with VLA-4 blockers

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0330506A2 (fr) 1988-02-26 1989-08-30 Dana Farber Cancer Institute Protéines VLA
WO1991003252A1 (fr) 1989-09-01 1991-03-21 Fred Hutchinson Cancer Research Center Inhibition de l'adhesion de lymphocytes sur l'endothelium vasculaire au moyen d'une nouvelle interaction entre le recepteur matriciel extra-cellulaire et son ligand
US5061620A (en) 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
EP0455482A2 (fr) 1990-05-01 1991-11-06 Becton, Dickinson and Company Subliguée de cellules progénitrices humaines
US5206345A (en) 1990-08-02 1993-04-27 Fred Hutchinson Cancer Research Center Il-4 and tnf induce mab 6g10-recognized expression on bone marrow stromal cells
EP0626861B2 (fr) 1992-01-13 2004-04-28 Biogen, Inc. Traitement de l'asthme
WO1993013798A1 (fr) 1992-01-13 1993-07-22 Biogen, Inc. Traitement de l'asthme
US20060013808A1 (en) 1992-01-13 2006-01-19 Biogen Inc., A Massachusetts Corporation Treatment for asthma
US5871734A (en) 1992-01-13 1999-02-16 Biogen, Inc. Treatment for asthma with VLA-4 blocking agents
US7176184B2 (en) 1992-02-12 2007-02-13 Biogen Idec Ma Inc. Treatment for inflammatory bowel disease with a fibronectin polypeptide
EP0625912B1 (fr) 1992-02-12 1997-04-16 Biogen, Inc. Traitement de l'inflammation intestinale
WO1993015764A1 (fr) 1992-02-12 1993-08-19 Biogen, Inc. Traitement de l'inflammation intestinale
US5695755A (en) * 1992-11-13 1997-12-09 Papayannopoulou; Thalia Peripheralization of hematopoietic stem cells
US5824304A (en) * 1992-11-13 1998-10-20 Papayannopoulou; Thalia Peripheralization of hematopoietic stem cells
US5888507A (en) 1993-02-09 1999-03-30 Biogen, Inc. Treatment for insulin dependent diabetes
US20050208053A1 (en) 1993-02-09 2005-09-22 Biogen, Inc., A Massachusetts Corporation Treatment for insulin dependent diabetes
EP0682529B2 (fr) 1993-02-09 2005-12-28 Biogen Idec MA, Inc. Anticorps pour le traitement du diabete insulino-dependent
WO1994017828A2 (fr) 1993-02-09 1994-08-18 Biogen, Inc. Traitement du diabete insulino-dependant
US5932214A (en) 1994-08-11 1999-08-03 Biogen, Inc. Treatment for inflammatory bowel disease with VLA-4 blockers
US6482409B1 (en) 1994-08-11 2002-11-19 Biogen, Inc. Treatment for inflammatory bowel disease with a vcam-1/1gG fusion protein

Non-Patent Citations (53)

* Cited by examiner, † Cited by third party
Title
Bednarczyk, J.L., et al., "Identification of a Combinatorial Epitope Expressed by the Integrin alpha4beta1 Heterodimer Involved in the Regulation of Cell Adhesion," J. Biol. Chem. 269:8348-8354, The American Society for Biochemistry and Molecular Biology, Inc. (Mar. 1994).
Bensinger, W., et al., "Autologous Transplantation with Peripheral Blood Mononuclear Cells Collected After Administration of Recombinant Granulocyte Stimulating Factor," Blood 81:3158-3163, American Society of Hematology (Jun. 1993).
Berenson, R.J., "Transplantation of CD34+ Hematopoietic Precursors: Clinical Rationale," Transplantation Proc. 24:3032-3034, Elsevier Science Inc. (Dec. 1992).
Bochner, B.S., et al., "Adhesion of Human Basophils, Eosinophils, and Neutrophils to Interleukin 1-activated Human Vascular Endothelial Cells: Contributions of Endothelial Cell Adhesion Molecules," J. Exp. Med. 173:1553-1556, The Rockefeller University Press (Jun. 1991).
Bregni,M., et al., "Human Peripheral Blood Hematopoietic Progenitors are Optimal Targets of Retroviral-Mediated Gene Transfer," Blood 80:1418-1422, American Society of Hematology (Sep. 1992).
Bronchud, M.H., et al., "In Vitro and In Vivo Analysis of the Effects of Recombinant Human Granulocyte Colony-Stimulating Factor in Patients," Br. J. Cancer 58:64-69, American Society of Hematology (Apr. 1988).
Brugger, W., et al., "Ex Vivo Expansion of Enriched Peripheral Blood CD34+ Progenitor Cells by Stem Cell Factor, Interleukin-1beta (IL-1beta), IL-6, IL-3, Interferon-gamma, and Erythropoietin," Blood 81:2579-2584, American Society of Hematology (Apr. 1993).
Caux et al (Blood, 1992, 79:2628-2635). *
Chao, N.J., et al., "Granulocyte Colony-Stimulating Factor 'Mobilized' Peripheral Blood Progenitor Cells Accelerate Granulocyte and Platelet Recovery After High-Dose Chemotherapy," Blood 81:2031-2035, American Society of Hematology (Apr. 1993).
Craig, J.I., et al., "Peripheral Blood Stem Cell Transplantation," Blood Rev. 6:59-67, Churchill Livingstone (Jun. 1992).
Denkers, I.A., et al., "VLA Molecule Expression May be Involved in the Release of Acute Myeloid Leukaemic Cells from the Bone Marrow," Leukemia Res. 16:469-476, Elsevier Ltd. (May 1992).
DePalma, "CellPro, Inc. Tests Its Stem Cell-Therapy in Clinic Trials," Genet. Eng. News 12, Mary Ann Liebert, Inc. (May 1992).
Edgington, S.M., "New Horizons for Stem-Cell Bioreactors," Biotechnol.10:1099-1106, Nature Publishing Company (Oct. 1992).
Elices, M.J., et al., "VCAM-1 on Activated Endothelium Interacts with the Leukocyte Integrin VLA-4 at a Site Distinct from the VLA-4/Fibronectin Binding Site," Cell 60:577-584, Cell Press (1990).
European Appeal of EP Patent No. 0 625 912, Decision of the Technical Board of Appeal, 29 pages (Oct. 6, 2004).
European Appeal of EP Patent No. 0 626 861, Decision of the Technical Board of Appeal, 4 pages (Oct. 23, 2003).
European Opposition of EP Patent 0 625 912 , Interlocutory Decision in Opposition Proceedings (Articles 102(3) and 106(3) EPC), 17 pages (Jun. 6, 2001).
European Opposition of EP Patent 0 625 912 document, Declaration of Dr. Roy Lobb, 2 pages (Mar. 13, 1999).
European Opposition of EP Patent No. 0 626 861, Interlocutory Decision in Opposition Proceedings (Articles 102(3) and 106(3) EPC), 17 pages (Feb. 13, 2003).
European Opposition of EP Patent No. 0 682 529, Interlocutory Decision in Opposition Proceedings (Articles 102(3) and 106(3) EPC), 16 pages (May 28, 2001).
Gale, R.P., et al., "Blood Stem Cell Transplants Come of Age," Bone Marrow Transplant. 9:151-155, Nature Publishing Group (Mar. 1992).
Gerhartz, H., "Zukunftspersperktiven von Knochenmarkund Stammzellaktivierung fur die autologe Transplantation," Beitr. Infusionther. 28:254-258, Karger (1991).
Gillis, "Chap. 21. T-Cell-Derived Lymphokines" in : Fundamental Immunol., Coleman, R.M., ed., McGraw-Hill Professional Publishing, New York, NY, pp. 621-638 (1989).
Haas, R., "Successful Autologous transplantation of Blood Stem Cells Mobilized with Recombinant Human Granulocyte-Macrophage Colony-Stimulating Factor," Exp. Hematol. 18:94-98, Elsevier Science Inc. (Feb. 1990).
Hemler, M.E., "VLA Proteins in the Integrin Family: Structures, Functions, and Their Role on Leukocytes," Annu. Rev. Immunol.8:365-400, Annual Reviews, Inc. (1990).
Issekutz (J of Immunology, Dec. 1991, 147:4178-4184). *
Kessinger, A. and Armitage, J.O., "The Evolving Role of Autologous Peripheral Stem Cell Transplantation Following High-Dose Therapy for Malignancies," Blood 77:211-213, American Society of Hematology (Jan. 1991).
Korbling, M., "Die Tolleder Stammzell-Mobilisation im Rahmen der Autologen Blutstammzell-Transplantation,"Beitr. Infusionther. 28:233-241, Karger (1991).
Liesveld, J.L., et al., "Expression of Integrins and Examination of Their Adhesive Function in Normal and Leukemic Hematopoietic Cells," Blood 81:112-121, American Society of Hematology (Jan. 1993).
Lobo, F., et al., "Addition of Peripheral Blood Stem Cells Collected Without Mobilization Techniques to Transplanted Autologous Bone Marrow Did Not Hasten Marrow Recovery Following Myeloablative Therapy," Bone Marrow Transplant. 8:389-392, Nature Publishing Group (Nov. 1991).
Magrin, et al., "Collection, Processing and Storage of Peripheral Blood Stem Cells (PBSC)," Haematologica, Suppl 1, 76:55-57, Ferrata Storti Foundation (Mar. 1991).
Papayannoulou, T. and Nakamoto, B., "Peripheralization of hemopoietic Progenitors in Primates Treated with Anti-VLA4 Integrin," Proc. Natl. Acad. Sci. USA 90L9374-9378, National Academy of Sciences (Oct. 1993).
Pulido, R., et al., "Functional Evidence for Three Distinct and Independently Inhibitable Adhesion Activities Mediated by the Human Integrin VLA-4. Correlation with Distinct alpha4 Epitopes," J. Biol. Chem. 266:10241-10245, American Society for Biochemistry and Molecular Biology (Jun. 1991).
Rowe, J.M. and Rapoport, A.P., "Hemapoietic Growth Factors: A Review," J. Clin. Pharmacol. 32:486-501, Sage Science Press (Jun. 1992).
Ryan, D.H., et al., "Inhibition of Human Bone Marrow Lymphoid Progenitor Colonies by Antibodies to VLA Integrins," J. Immunol. 149:3759-3764, American Association of Immunologists (Dec. 1992).
Siena, S., et al., Circulation of CD34+ Hematopoietic Stem Cells in the Peripheral Blood of High-Dose Cyclophosphamide-Treated Patients: Enhancement by Intravenous Recombinant Human Granulocyte-Macrophage Colony-Stimulating Factor, Blood 74:1905-1914, American Society of Hematology 1989.
Simmons, P.J., et al., "Vascular Cell Adhesion Molecule-1 Expressed by Bone Marrow Stromal Cells Mediates the Binding of Hematopoietic Progenitor Cells," Blood 80:388-395, American Society of Hematology (Jul. 1992).
Strauch, U.G., et al., "Distinct binding specifities of integrins alpha4beta7 (LPAM-1) alpha4beta1 (VLA-4), and IELbeta7," Intl. Immunol. 6:263-275, Oxford University Press (Feb. 1994).
Teixido et al., "Human CD34 + Progenitor Cell Adhesion to Marrow Stroma is Mediated by VLA-4/VCAM and VLA5/Fibronectin", Blood, 78, Suppl. 1, p. 302a, abstract 1200 (1991). *
Teixido, J., et al., "Human CD+Progenitor Cell Adhesion to Marrow Stroma is Mediated by VLA-4/VCAM and VLA5/Fibronectin," Blood Suppl. 1, 78:302a, abstract no. 1200, American Society for Clinical Investigation (Aug. 1992).
Teixido, J., et al., "Role of beta1 and beta2 Integrins in the Adhesion of Human CD34hi Stem Cells to Bone Marrow Stroma," J. Clin. Invest. 90:358-367, American Society for Clinical Investigation (Aug. 1992).
U.S. Interference Document, Decision on Motions, Patent Interference No. 105,378, Papayannopoulouv. Masinovsky, 2 pages (Oct. 20, 2006).
U.S. Interference Document, Exhibit 2014: Declaration of Timothy A. Springer, Ph.D., submitted by Party Papayannopoulou in Patent Interference No. 105,378 Papayannopoulou v. Masinovsky, 10 pages (Mar. 17, 2006).
U.S. Interference Document, Judgment-Request for Adverse -Bd. R. 127(b), Patent Interference No. 105,378, Papayannopoulouv. Masinovsky, 3 pages (Nov. 29, 2006).
U.S. Interference Document, Order-Motion Times-Bd. R. 104(c), Patent Interference No. 105,378, Papayannopoulouv. Masinovsky, 13 pages (Dec. 14, 2005).
U.S. Interference Document, Order-Termination of Proceedings-Bd.R. 8, Patent Interference No. 105,378, Papayannoulou v. Masinovsky, 3 pages (Jan. 22, 2007).
U.S. Interference Document, Papayannopoulou Request for Adverse Judgment, submitted in Patent Interference No. 105,378, Papayannopulouv. Masinovsky, 3 pages (Nov. 9, 2006).
U.S. Interference Document, Papayannopoulou Submission of Transcript of Oral Argument on Aug. 23, 2006, submitted in Patent Interference No. 105,378, Papayannopoulou v. Masinovsky, 88 pages (Oct. 13, 2006).
U.S. Interference Document, Papayannopoulou Substantive Motion 1 and Appendices 1-2, submitted in Patent Interference No. 105,378, Papayannopoulou v. Masinovsky, 38 pages (Mar. 17, 2006).
U.S. Interference Document, Redeclaration-Bd.R. 203(c), Patent Interference No. 105,378, Papayannopoulouv. Masinovsky, 4 pages (Aug. 25, 2006).
Weller, P.F., et al., "Human eosinophil adherence to vascular endothelium mediated by binding to vascular cell adhesion molecule 1 and endothelial leukocyte adhesion molecule 1," Proc. Natl. Acad. Sci. USA 88:7430-7433, National Academy of Sciences (Aug. 1991).
Williams et al (Nature, Aug. 1991, 352:438-441). *
Williams et al., "Fibronectin and VLA-4 in hematopoietic Stem Cells-Microenvironment Interactions", Nature, 352, pp. 438-441 (1991). *

Also Published As

Publication number Publication date
US5695755A (en) 1997-12-09
US5843438A (en) 1998-12-01
GR3023975T3 (en) 1997-10-31
DE69309487D1 (de) 1997-05-07
CA2148712C (fr) 2012-01-17
EP0670735A1 (fr) 1995-09-13
ATE150975T1 (de) 1997-04-15
AU5727294A (en) 1994-06-08
DK0670735T3 (da) 1997-07-28
ES2102196T3 (es) 1997-07-16
AU689454B2 (en) 1998-04-02
JP2003277294A (ja) 2003-10-02
JP3731892B2 (ja) 2006-01-05
JPH08506091A (ja) 1996-07-02
DE69309487T2 (de) 1997-10-23
US5824304A (en) 1998-10-20
CA2148712A1 (fr) 1994-05-26
WO1994011027A1 (fr) 1994-05-26
EP0670735B1 (fr) 1997-04-02

Similar Documents

Publication Publication Date Title
USRE40811E1 (en) Peripheralization of hematopoietic stem cells
US11673953B2 (en) DLL3 targeting chimeric antigen receptors and binding agents
JP2883201B2 (ja) Cd28刺激関連免疫療法
Kupiec-Weglinski et al. Interleukin 2 receptor-targeted therapy—rationale and applications in organ transplantation
AU2009224690B2 (en) Agent for treating disease
US5817311A (en) Methods of inhibiting T-cell medicated immune responses with LO-CD2a-specific antibodies
JPH04316600A (ja) Cd25結合分子
US5730979A (en) LO-CD2a antibody and uses thereof for inhibiting T cell activation and proliferation
EP3475301B1 (fr) Prévention ou traitement de rechute de malignité hématologique à l'aide d'un antagoniste tnfr2
AU657255B2 (en) Monoclonal antibodies for inducing tolerance
KR20050093759A (ko) 혈소판 및 조혈 줄기세포의 생산을 증가시키는 방법
Hakimi et al. Development of Zenapax®: a humanized anti-Tac antibody
WO1995013093A1 (fr) Traitement d'un patient avant une transplantation
JPH03505211A (ja) 好酸球増加症の予防または鎮静用薬剤
Egan et al. Treatment of epidermolysis bullosa acquisita with the humanized anti-Tac mAb daclizumab
Huang et al. Immunotherapy of multiple myeloma
AU737602B2 (en) LO-CD2a antibody and uses thereof for inhibiting T cell activation and proliferation
AU655459B2 (en) Method for inhibiting pulmonary oxygen toxicity
Williams et al. T cell activation antigens: therapeutic implications
US7592006B1 (en) Composition comprising the LO-CD2a antibody
Waldmann et al. The interleukin-2 receptor: a target for immunotherapy
Chatenoud Treatment of autoimmune disease: Biological and molecular therapies
Hakimi et al. DEVELOPMENT OF ZENAPAX*: A HUMANIZED ANTI-TAC
IE922028A1 (en) Antibody composition acting synergistically
Williams et al. De Novo Expression of Receptors on T Cells

Legal Events

Date Code Title Description
FPAY Fee payment

Year of fee payment: 12