WO1996023058A1 - ENHANCING BONE MARROW ENGRAFTMENT USING MSCs - Google Patents

ENHANCING BONE MARROW ENGRAFTMENT USING MSCs Download PDF

Info

Publication number
WO1996023058A1
WO1996023058A1 PCT/US1996/000900 US9600900W WO9623058A1 WO 1996023058 A1 WO1996023058 A1 WO 1996023058A1 US 9600900 W US9600900 W US 9600900W WO 9623058 A1 WO9623058 A1 WO 9623058A1
Authority
WO
WIPO (PCT)
Prior art keywords
bone marrow
cells
mesenchymal stem
marrow
stem cells
Prior art date
Application number
PCT/US1996/000900
Other languages
French (fr)
Inventor
Stephen E. Haynesworth
Arnold I. Caplan
Stanton L. Gerson
Hillard M. Lazarus
Original Assignee
Case Western Reserve University
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 Case Western Reserve University filed Critical Case Western Reserve University
Priority to AU47041/96A priority Critical patent/AU4704196A/en
Publication of WO1996023058A1 publication Critical patent/WO1996023058A1/en

Links

Classifications

    • 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)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/28Bones
    • 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
    • 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
    • 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
    • A61L27/3839Materials 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 the site of application in the body
    • A61L27/3843Connective tissue
    • 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
    • A61L27/3895Materials 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 using specific culture conditions, e.g. stimulating differentiation of stem cells, pulsatile flow conditions
    • 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
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/005Ingredients of undetermined constitution or reaction products thereof
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/02Prostheses implantable into the body
    • A61F2/30Joints
    • A61F2/30756Cartilage endoprostheses
    • 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
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • 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
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/02Materials or treatment for tissue regeneration for reconstruction of bones; weight-bearing implants
    • 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
    • A61L2430/00Materials or treatment for tissue regeneration
    • A61L2430/10Materials or treatment for tissue regeneration for reconstruction of tendons or ligaments
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2301Interleukin-1 (IL-1)
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • 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
    • C12N2533/10Mineral substrates
    • C12N2533/18Calcium salts, e.g. apatite, Mineral components from bones, teeth, shells

Definitions

  • the present invention is directed to various methods and devices for using mesenchymal stem cells (MSCs) to enhance bone marrow engraftment.
  • MSCs mesenchymal stem cells
  • Mesenchymal stem cells are the formative pluripotent blast cells found in the bone that are capable of differentiating into any of the specific types of connective tissues (i.e., the tissues of the adipose, areolar, osseous, cartilaginous, elastic, and fibrous connective tissues) depending upon various environmental influences. Although these cells are normally present at very low frequencies in bone marrow and other mesenchymal tissues, the inventors of the present invention have discovered a process for isolating, purifying, and greatly replicating the mesenchymal stem cells in culture, i.e. in vitro.
  • the methods and devices of the invention utilize such isolated and culture-expanded mesenchymal stem cells (MSCs) . Under selected conditions, they can be induced to differentiate into different types of skeletal -and connective tissues such as bone, cartilage, tendon, ligament, muscle, other connective tissues and marrow stroma.
  • MSCs mesenchymal stem cells
  • the marrow stroma provides the scaffolding as well as soluble factors which direct and support blood cell synthesis, i.e., hematopoiesis.
  • the present invention is directed to a method to improve the process of bone marrow transplantation which is used to regenerate blood cells and marrow tissue in patients where their marrow is depleted or destroyed, such as, for example, during intensive radiation and chemotherapy treatment.
  • culture-expanded MSCs can increase survival and decrease the time of blood and marrow cell regeneration when transplanted with complete bone marrow.
  • the present invention provides a method for enhancing the regeneration of marrow tissue through improved bone marrow transplantation using MSCs.
  • the method for enhancing bone marrow engraftment comprises administering to an individual in need thereof, (i) mesenchymal stem cells and (ii) a bone marrow graft, wherein said mesenchymal stem cells are administered in an amount effective to promote engraftment of the bone marrow in the individual.
  • one embodiment of the invention is directed to a method for using a culture medium comprised of Dulbecco's Modified Essential Medium with low glucose (DMEM- G) or medium 199 plus 1% human albumin as a vehicle or carrier for MSCs which, when administered systemically, will migrate, or home, to the marrow cavity and differentiate into marrow stroma, thereby regenerating the marrow stroma.
  • the MSCs can be administered systemically, e.g., intravenously, into various delivery sites or directly into the bone.
  • a further aspect of the present invention is directed to the timing of injection of the MSCs into the patient relative to the bone marrow transplantation.
  • the MSCs are injected simultaneously with the bone marrow.
  • the MSCs are administered before or after the injection of bone marrow.
  • the present invention is useful to enhance the effectiveness of bone marrow transplantation as a treatment for cancer.
  • the treatment of cancer by x-irradiation or al ylating therapy destroys the bone marrow microenvironment as well as the hematopoietic stem cells.
  • the current treatment is to transplant the patient after marrow ablation with bone marrow which has been previously harvested and cryopreserved.
  • a critical aspect of the present invention is directed to the advantages of transplanting isolated, purified, culture-expanded MSCs to accelerate the process of stromal reconstitution and ultimately marrow engraftmen .
  • Modes of administration of the MSC preparation include but are not limited to systemic intravenous injection and injection directly to the intended site of activity.
  • the preparation can be administered by any convenient route, for example by infusion or bolus injection and can be administered together with other biologically active agents. Administration is preferably systemic.
  • compositions comprise a therapeutically effective amount of the MSCs, and a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutically acceptable carrier includes but is not limited to medium 199 plus 1% serum albumin, saline, buffered saline, dextrose, water, and combinations thereof.
  • the formulation should suit the mode of administration.
  • the MSC preparation or composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a local anesthetic to ameliorate any pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a cryopreserved concentrate in a hermetically sealed container such as an ampoule indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the method of the invention can be altered, particularly by (1) increasing or decreasing the time interval between injecting MSCs and implanting the tissue; (2) increasing or decreasing the amount of MSCs injected; (3) varying the number of MSC injections; (4) varying the method of delivery of MSC; or (5) varying the source of MSCs.
  • MSCs derived from the tissue donor is preferable, the MSCs can be obtained from other individuals or species, or from genetically-engineered inbred donor strains, or from in vitro cell culture.
  • the MSC preparation is used in an amount effective to promote engraftment of bone marrow in the recipient. In general, such amount is at least 1 xlO 4 MSC per kg of body weight and most generally need not be more than 7 x io 5 MSC/kg. Preferably, it is at least about 2 x 10 5 MSC/kg prior to graft introduction and usually need not be more than about 7 x 10 s MSC/kg.
  • the MSC preparation rrray be administered concurrently with the bone marrow transplant or for a period prior to graft introduction of at least about 7 days but generally not to exceed 30 days, with a typical therapeutic treatment period of 7 to 14 days.
  • the MSC preparation preferably is administered either intravenously one to three times per day, and may be adjusted to meet optimal efficacy and pharmacological dosing.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical preparation of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical preparation of the invention.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Marrow in femoral head cancellous bone pieces was obtained from patients with degenerative joint disease during hip or knee joint replacement surgery.
  • marrow was also obtained by iliac aspirate from normal donors and oncology patients who were having marrow harvested for future bone marrow transplantation. All of the oncology patients had malignancies unrelated to the stromal cells and the stromal cells expressed normal karyotype.
  • Marrow for cell culture was prepared from plugs of cancellous bone marrow as follows. Plugs of cancellous bone marrow (0.5-1.5 ml) were transferred to sterile tubes to which 25 ml Dulbecco's Modified Eagles Medium with low glucose (DMEM-LG) (GIBCO, Grand Island, N.Y.) with selected batches of 10% fetal bovine serum (complete medium) was added. The tubes were vortexed to disperse the marrow, then spun at 1000 rpm for 5 minutes to pellet cells and bone pieces. The supernatant and fat layer were removed and the marrow and bone were reconstituted in 5 ml complete medium and vortexed to suspend the marrow cells.
  • DMEM-LG Dulbecco's Modified Eagles Medium with low glucose
  • fetal bovine serum complete medium
  • the suspended cells were collected with a 10 ml syringe fitted with a 16 gauge needle and transferred to separate tubes. Bone pieces were reconstituted in 5 ml of complete medium and the marrow cells were collected as before. Collection of marrow cells was considered complete when a pellet of yellowish-white cancellous bone pieces was all that remained in the original tube. Marrow cells were separated into a single cell suspension by passing them through syringes fitted with 18 and 20 gauge needles. Cells were spun at 1000 x g for 5 minutes after which the fat layer and supernatant were removed. Cells were reconstituted in complete medium, counted with a hemocytometer (red blood cells were lysed prior to counting with 4% acetic acid) , and plated in 100 mm dishes at 50 x 10 6 nucleated cells/dish.
  • a hemocytometer red blood cells were lysed prior to counting with 4% acetic acid
  • Marrow for cell culture was prepared from aspirated bone marrow as follows. Aspirate marrow (5-10 ml) was transferred to sterile 50 ml plastic centrifuge tubes to which 20 ml complete medium was added. The tubes were spun at 1,500 rpm for 5 minutes to pellet the cells. The supernatant and fat layer were removed and the cell pellets were resuspended to 5 ml with complete medium. The cell suspensions then were loaded onto 70% Percoll (Sigma, St. Louis, Mo.) gradients with a 10 ml pipette and spun at 2,000 rpm (460xg) in a GS34 rotor for 15 minutes.
  • Percoll Sigma, St. Louis, Mo.
  • the medium is aspirated off from the top down to the marked line (approximately 12-14 ml) .
  • the collected fraction is transferred to a 50 ml conical plastic tube. 30 ml of complete medium then is added to the tube and centrifuged for 5 minutes at 1,500 rpm. The supernatant then is removed and discarded.
  • the cells then are resuspended in 7 ml of complete medium, and a uniform cell suspension is generated by pipetting cells up and down with a 10 ml pipette.
  • the cells are counted by taking 20 ⁇ l of uniform cell suspension, and adding 20 ⁇ l of 4% acetic acid to the cell suspension.
  • the suspension is mixed and transferred to a hemocytometer, and the cells are counted.
  • Cell concentration then is adjusted with complete medium to 50 x 10 5 cells per 7 ml. 7 ml of cells (50 x 10 6 cells) then are plated per lOOmm culture plate.
  • the marrow mesenchymal stem cells were purified and expanded as follows. Marrow cells from either the femoral head cancellous bone or the iliac aspirate were cultured in complete medium (i.e., DMEM-LG medium with 10% fetal bovine serum) at 37°C in humidified atmosphere containing 95% air and 5% C0 2 . On day 3 after plating, nonadherent cells were removed from the cultures by aspirating the original medium from the plates and replacing the original medium with 7 ml of fresh complete medium. Subsequent medium changes were performed every 3 to 4 days. This process of removing the non-adherent cells during culture media changes results in purification of the mesenchymal stem cells which selectively adhere to the culture plates.
  • complete medium i.e., DMEM-LG medium with 10% fetal bovine serum
  • the cells from each culture then are counted and plated in three new 100 ml plates in 7 ml complete medium.
  • the three new plates of cells are termed first passage cultures.
  • First passage cells then were allowed to expand until they became near confluent, and were then replated as described above to create second passage cultures.
  • aliquots of cells were cryopreserved in 90% fetal bovine serum with 10% DMSO (freezing medium) .
  • EXAMPLE 2 Mesenchymal stem cells were obtained from 10ml of autologous human bone marrow collected either at the time of autologous bone marrow harvest, or during a routine diagnostic bone marrow examination, from the iliac crest. These mesenchymal stem cells were culture expanded and infused into 14 patients who had hematologic malignancies, but had no evidence of active malignancy, and had not received chemotherapy or radiation therapy for at least 4 weeks before collection of mesenchymal stem cells. Five patients each received 1 million autologous mesenchymal stem cells, five patients each were given 10 million mesenchymal stem cells, and four patients each received 50 million mesenchymal stem cells. Toxicity for each patient was determined according to the National Cancer Institute toxicity grading scale.
  • the toxicity grading scale was the same as that based upon the common toxicity criteria for a Phase II trial of high-dose sequential chemotherapy and peripheral blood stem cell autologous transplantation as initial therapy for patients with poor prognosis non- Hodgkin's lymph ⁇ ma. (National Cancer Institute document, draft 4/94: E2493.) Toxicity was graded on a scale from 0 to 4. No Grade 2 or greater toxic effects were observed, and in two patients that received 10 million mesenchymal stem cells, there appeared to be an increase in bone marrow cellularity.
  • a breast cancer patient undergoes a diagnostic posterior iliac crest bone marrow aspiration and biopsy using a local anesthetic.
  • a small portion (2 to 3 ml) of the aliquot (10 to 20 ml) of marrow is submitted for routine histologic testing and determination of tumor content using immunoperoxidase testing of cells grown in semisolid agar as described in Ross, et al., Blood.. Vol. 82, pgs. 2605-2610 (1993) .
  • the remainder of the cells are cultured as hereinabove described .
  • samples of the mesenchymal stem cells are submitted for cell counting, cell viability (Trypan Blue) , and cell surface immunophenotyping (testing with monoclonal antibodies SH2, SH3, and SH4) .
  • SH2 is produced by a hybridoma cell line assigned ATCC Accession No. HB10743.
  • SH3 is produced by a hybridoma cell line assigned ATCC Accession No. HB10744.
  • SH4 is produced by a hybridoma cell line assigned ATCC Accession No. HB10745. Samples also are submitted for detection of occult breast cancer.
  • the patient also undergoes placement of a pheresis central venous catheter, and receives subzutaneous injections of G-CSF (filgrastin) lO ⁇ g/kg/day as described in Peters, et al., Blood. Vol. 81, pgs. 1709-1719 (1993); Chao, et al., Blood. Vol. 81, pgs. 2031-2035 (1993); Sheridan, et al., The Lancet. Vol. 2, pgs. 891-895 (1989); and Winter, et al., Blood. Vol. 82, pg. 293a (1993) .
  • G-CSF filament
  • G-CSF injections begin at least 3 days before the first pheresis is initiated. G-CSF therapy is withheld if the white blood cell count rises above 40,000/ ⁇ L and is resumed once the white blood cell count drops to less than 20,000/ ⁇ L.
  • the patient If the patient is receiving only G-CSF as the vehicle for "mobilization" of peripheral blood progenitor cells, the patient must not have received chemotherapy within 4 weeks of the planned pheresis. If the patient has received both conventional chemotherapy and G-CSF treatment for mobilization, the patient must not have received chemotherapy within 10 days of the planned pheresis, and the white blood cell count must be at least 800/ ⁇ L and the platelet count at least 30,000/ ⁇ L.
  • the patient receives chemotherapy over a period of 96 hours (4 days) , with the following chemotherapy agents: 1.
  • Cyclophosphamide in a total dosage of 6g/m 2 (1.5 g/m 2/day for 4 days) is given via continuous intravenous infusion at 500 mg/m 2 in 1,000 ml normal saline every 8 hours.
  • Thiotepa in a total dosage of 500 mg/m/day for 4 days) is given via continuous intravenous infusion at 125 mg/m 2 in 1,000 ml normal saline every 24 hours.
  • Carboplatin in a total dosage of 1,800 mg/m 2 (200 mg/m 2 /day for 4 days) is given via continuous intravenous infusion at 200 mg/m 2 in 1,000 ml of 5% dextrose in water every 24 hours.
  • the patient also receives 500 mg of mesna in 50 ml normal saline IV over 15 minutes every 4 hours for 6 days
  • the mesenchymal stem cells are harvested from tissue culture flasks.
  • Cells are collected using bovine trypsin (which is deactivated with soybean trypsin inhibitor) , suspended at 10 x 10* cells/ml in TC199 containing 1% human serum albumin (infusion medium) , washed twice in infusion medium, resuspended at approximately 10 6 cells/ml, and injected slowly intravenously over 15 minutes to provide a total dosage of from 10 to about 5x10 6 cells.
  • bovine trypsin which is deactivated with soybean trypsin inhibitor
  • the frozen autologous peripheral blood progenitor cells are removed from the liquid nitrogen refrigerator, transported to the patient in liquid nitrogen, submersed in a 37°C to 40°C sterile water bath, and infused rapidly intravenously without additional filtering or washing steps.
  • GM-CSF in an amount of 250 ⁇ g/m 2 then is given as a daily subcutaneous injection, beginning 3 hours after completion of the autologous blood progenitor cell infusion.
  • the GM-CSF is given daily until the peripheral blood neutrophil count exceeds 1,000/ ⁇ L for three consecutive days.

Abstract

Method and preparations for enhancing bone marrow engraftment in an individual by administering (i) a culturally expanded mesenchymal stem cell preparation and (ii) a bone marrow graft. The mesenchymal stem cells are administered in an amount effective to promote engraftment of the bone marrow.

Description

ENHANCING BONE MARROW ENGRAFTMENT USING MSCs
The present invention is directed to various methods and devices for using mesenchymal stem cells (MSCs) to enhance bone marrow engraftment. Mesenchymal stem cells are the formative pluripotent blast cells found in the bone that are capable of differentiating into any of the specific types of connective tissues (i.e., the tissues of the adipose, areolar, osseous, cartilaginous, elastic, and fibrous connective tissues) depending upon various environmental influences. Although these cells are normally present at very low frequencies in bone marrow and other mesenchymal tissues, the inventors of the present invention have discovered a process for isolating, purifying, and greatly replicating the mesenchymal stem cells in culture, i.e. in vitro. This discovery is the subject of co-pending U.S. patent application Serial No. 193,262, filed February 8, 1994. The methods and devices of the invention utilize such isolated and culture-expanded mesenchymal stem cells (MSCs) . Under selected conditions, they can be induced to differentiate into different types of skeletal -and connective tissues such as bone, cartilage, tendon, ligament, muscle, other connective tissues and marrow stroma.
The marrow stroma provides the scaffolding as well as soluble factors which direct and support blood cell synthesis, i.e., hematopoiesis. The present invention is directed to a method to improve the process of bone marrow transplantation which is used to regenerate blood cells and marrow tissue in patients where their marrow is depleted or destroyed, such as, for example, during intensive radiation and chemotherapy treatment. Along this line, the inventors have discovered that, after lethal doses of radiation, culture-expanded MSCs can increase survival and decrease the time of blood and marrow cell regeneration when transplanted with complete bone marrow.
The present invention provides a method for enhancing the regeneration of marrow tissue through improved bone marrow transplantation using MSCs. The method for enhancing bone marrow engraftment comprises administering to an individual in need thereof, (i) mesenchymal stem cells and (ii) a bone marrow graft, wherein said mesenchymal stem cells are administered in an amount effective to promote engraftment of the bone marrow in the individual. More particularly, one embodiment of the invention is directed to a method for using a culture medium comprised of Dulbecco's Modified Essential Medium with low glucose (DMEM- G) or medium 199 plus 1% human albumin as a vehicle or carrier for MSCs which, when administered systemically, will migrate, or home, to the marrow cavity and differentiate into marrow stroma, thereby regenerating the marrow stroma. The MSCs can be administered systemically, e.g., intravenously, into various delivery sites or directly into the bone.
A further aspect of the present invention is directed to the timing of injection of the MSCs into the patient relative to the bone marrow transplantation. In one embodiment, the MSCs are injected simultaneously with the bone marrow. In another embodiment, the MSCs are administered before or after the injection of bone marrow. The present invention is useful to enhance the effectiveness of bone marrow transplantation as a treatment for cancer. The treatment of cancer by x-irradiation or al ylating therapy destroys the bone marrow microenvironment as well as the hematopoietic stem cells. The current treatment is to transplant the patient after marrow ablation with bone marrow which has been previously harvested and cryopreserved. However, because the bone marrow microenvironment is destroyed, bone marrow engraftment is delayed until the stromal environment is restored. As a result, a critical aspect of the present invention is directed to the advantages of transplanting isolated, purified, culture-expanded MSCs to accelerate the process of stromal reconstitution and ultimately marrow engraftmen .
Modes of administration of the MSC preparation include but are not limited to systemic intravenous injection and injection directly to the intended site of activity. The preparation can be administered by any convenient route, for example by infusion or bolus injection and can be administered together with other biologically active agents. Administration is preferably systemic.
The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of the MSCs, and a pharmaceutically acceptable carrier or excipient. Such a carrier includes but is not limited to medium 199 plus 1% serum albumin, saline, buffered saline, dextrose, water, and combinations thereof. The formulation should suit the mode of administration.
In a preferred embodiment, the MSC preparation or composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a local anesthetic to ameliorate any pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a cryopreserved concentrate in a hermetically sealed container such as an ampoule indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
The method of the invention can be altered, particularly by (1) increasing or decreasing the time interval between injecting MSCs and implanting the tissue; (2) increasing or decreasing the amount of MSCs injected; (3) varying the number of MSC injections; (4) varying the method of delivery of MSC; or (5) varying the source of MSCs. Although MSCs derived from the tissue donor is preferable, the MSCs can be obtained from other individuals or species, or from genetically-engineered inbred donor strains, or from in vitro cell culture.
The MSC preparation is used in an amount effective to promote engraftment of bone marrow in the recipient. In general, such amount is at least 1 xlO4 MSC per kg of body weight and most generally need not be more than 7 x io5 MSC/kg. Preferably, it is at least about 2 x 105 MSC/kg prior to graft introduction and usually need not be more than about 7 x 10s MSC/kg. The MSC preparation rrray be administered concurrently with the bone marrow transplant or for a period prior to graft introduction of at least about 7 days but generally not to exceed 30 days, with a typical therapeutic treatment period of 7 to 14 days. The MSC preparation preferably is administered either intravenously one to three times per day, and may be adjusted to meet optimal efficacy and pharmacological dosing.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical preparation of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
EXAMPLE 1
Isolation, Purification and Cultural
Expansion of Marrow-Derived Mesenchymal Stem Cells
Marrow in femoral head cancellous bone pieces was obtained from patients with degenerative joint disease during hip or knee joint replacement surgery. In addition, marrow was also obtained by iliac aspirate from normal donors and oncology patients who were having marrow harvested for future bone marrow transplantation. All of the oncology patients had malignancies unrelated to the stromal cells and the stromal cells expressed normal karyotype.
Marrow for cell culture was prepared from plugs of cancellous bone marrow as follows. Plugs of cancellous bone marrow (0.5-1.5 ml) were transferred to sterile tubes to which 25 ml Dulbecco's Modified Eagles Medium with low glucose (DMEM-LG) (GIBCO, Grand Island, N.Y.) with selected batches of 10% fetal bovine serum (complete medium) was added. The tubes were vortexed to disperse the marrow, then spun at 1000 rpm for 5 minutes to pellet cells and bone pieces. The supernatant and fat layer were removed and the marrow and bone were reconstituted in 5 ml complete medium and vortexed to suspend the marrow cells. The suspended cells were collected with a 10 ml syringe fitted with a 16 gauge needle and transferred to separate tubes. Bone pieces were reconstituted in 5 ml of complete medium and the marrow cells were collected as before. Collection of marrow cells was considered complete when a pellet of yellowish-white cancellous bone pieces was all that remained in the original tube. Marrow cells were separated into a single cell suspension by passing them through syringes fitted with 18 and 20 gauge needles. Cells were spun at 1000 x g for 5 minutes after which the fat layer and supernatant were removed. Cells were reconstituted in complete medium, counted with a hemocytometer (red blood cells were lysed prior to counting with 4% acetic acid) , and plated in 100 mm dishes at 50 x 106 nucleated cells/dish.
Marrow for cell culture was prepared from aspirated bone marrow as follows. Aspirate marrow (5-10 ml) was transferred to sterile 50 ml plastic centrifuge tubes to which 20 ml complete medium was added. The tubes were spun at 1,500 rpm for 5 minutes to pellet the cells. The supernatant and fat layer were removed and the cell pellets were resuspended to 5 ml with complete medium. The cell suspensions then were loaded onto 70% Percoll (Sigma, St. Louis, Mo.) gradients with a 10 ml pipette and spun at 2,000 rpm (460xg) in a GS34 rotor for 15 minutes. In order to harvest the cells, the tube is marked just below the high concentrated band of platelets, about 25% to 35% of the way down the tube. (Pooled density = 1.03g/ml.) Using a 10 ml pipette, the medium is aspirated off from the top down to the marked line (approximately 12-14 ml) . the collected fraction is transferred to a 50 ml conical plastic tube. 30 ml of complete medium then is added to the tube and centrifuged for 5 minutes at 1,500 rpm. The supernatant then is removed and discarded.
The cells then are resuspended in 7 ml of complete medium, and a uniform cell suspension is generated by pipetting cells up and down with a 10 ml pipette. The cells are counted by taking 20 μl of uniform cell suspension, and adding 20 μl of 4% acetic acid to the cell suspension. The suspension is mixed and transferred to a hemocytometer, and the cells are counted.
Cell concentration then is adjusted with complete medium to 50 x 105 cells per 7 ml. 7 ml of cells (50 x 106 cells) then are plated per lOOmm culture plate.
The marrow mesenchymal stem cells were purified and expanded as follows. Marrow cells from either the femoral head cancellous bone or the iliac aspirate were cultured in complete medium (i.e., DMEM-LG medium with 10% fetal bovine serum) at 37°C in humidified atmosphere containing 95% air and 5% C02. On day 3 after plating, nonadherent cells were removed from the cultures by aspirating the original medium from the plates and replacing the original medium with 7 ml of fresh complete medium. Subsequent medium changes were performed every 3 to 4 days. This process of removing the non-adherent cells during culture media changes results in purification of the mesenchymal stem cells which selectively adhere to the culture plates. When primary culture dishes became nearly confluent, the medium was removed and each plate rinsed with 7 ml of sterile Tyrode's solution. The cells were detached with 4 ml of 0.25% trypsin with 0.1 mM EDTA (GIBCO) for 5 minutes at 37°C. The action of trypsin was stopped with 2 ml of calf serum. The cells then were collected with a 10 ml pipette and centrifuged for 5 minutes at 1,500 rpm. The supernatant then is removed, and the cells resuspended from each plate in 21 ml complete medium, and a uniform cell suspension is generated by gently pipetting the cells up and down in the pipette. The cells from each culture then are counted and plated in three new 100 ml plates in 7 ml complete medium. The three new plates of cells are termed first passage cultures. First passage cells then were allowed to expand until they became near confluent, and were then replated as described above to create second passage cultures. At each passage, aliquots of cells were cryopreserved in 90% fetal bovine serum with 10% DMSO (freezing medium) .
EXAMPLE 2 Mesenchymal stem cells were obtained from 10ml of autologous human bone marrow collected either at the time of autologous bone marrow harvest, or during a routine diagnostic bone marrow examination, from the iliac crest. These mesenchymal stem cells were culture expanded and infused into 14 patients who had hematologic malignancies, but had no evidence of active malignancy, and had not received chemotherapy or radiation therapy for at least 4 weeks before collection of mesenchymal stem cells. Five patients each received 1 million autologous mesenchymal stem cells, five patients each were given 10 million mesenchymal stem cells, and four patients each received 50 million mesenchymal stem cells. Toxicity for each patient was determined according to the National Cancer Institute toxicity grading scale. The toxicity grading scale was the same as that based upon the common toxicity criteria for a Phase II trial of high-dose sequential chemotherapy and peripheral blood stem cell autologous transplantation as initial therapy for patients with poor prognosis non- Hodgkin's lymphσma. (National Cancer Institute document, draft 4/94: E2493.) Toxicity was graded on a scale from 0 to 4. No Grade 2 or greater toxic effects were observed, and in two patients that received 10 million mesenchymal stem cells, there appeared to be an increase in bone marrow cellularity.
EXAMPLE 3
Administration of Bone Marrow Cells and Mesenchymal
Stem Cells to Breast Cancer Patients
Treated with Chemotherapy
A breast cancer patient undergoes a diagnostic posterior iliac crest bone marrow aspiration and biopsy using a local anesthetic. A small portion (2 to 3 ml) of the aliquot (10 to 20 ml) of marrow is submitted for routine histologic testing and determination of tumor content using immunoperoxidase testing of cells grown in semisolid agar as described in Ross, et al., Blood.. Vol. 82, pgs. 2605-2610 (1993) . The remainder of the cells are cultured as hereinabove described . After at least 3 weeks in culture, samples of the mesenchymal stem cells are submitted for cell counting, cell viability (Trypan Blue) , and cell surface immunophenotyping (testing with monoclonal antibodies SH2, SH3, and SH4) . SH2 is produced by a hybridoma cell line assigned ATCC Accession No. HB10743. SH3 is produced by a hybridoma cell line assigned ATCC Accession No. HB10744. SH4 is produced by a hybridoma cell line assigned ATCC Accession No. HB10745. Samples also are submitted for detection of occult breast cancer.
The patient also undergoes placement of a pheresis central venous catheter, and receives subzutaneous injections of G-CSF (filgrastin) lOμg/kg/day as described in Peters, et al., Blood. Vol. 81, pgs. 1709-1719 (1993); Chao, et al., Blood. Vol. 81, pgs. 2031-2035 (1993); Sheridan, et al., The Lancet. Vol. 2, pgs. 891-895 (1989); and Winter, et al., Blood. Vol. 82, pg. 293a (1993) . G-CSF injections begin at least 3 days before the first pheresis is initiated. G-CSF therapy is withheld if the white blood cell count rises above 40,000/μL and is resumed once the white blood cell count drops to less than 20,000/μL.
If the patient is receiving only G-CSF as the vehicle for "mobilization" of peripheral blood progenitor cells, the patient must not have received chemotherapy within 4 weeks of the planned pheresis. If the patient has received both conventional chemotherapy and G-CSF treatment for mobilization, the patient must not have received chemotherapy within 10 days of the planned pheresis, and the white blood cell count must be at least 800/μL and the platelet count at least 30,000/μL.
Daily pheresis procedures are performed using a Cobe Spectra instrument (Cobe, Lakewood, Colorado) , and each cellular collection is cryopreserved using a controlled-rate liquid nitrogen freezer, until at least 15 x 108 mononuclear cells/kg are collected - (Lazarus, et al., Bone Marrow Transplant, Vol. 7, pgs. 241-246 (1991)). Each peripheral blood progenitor cell will be processed and cryopreserved according to previously published techniques. (Lazarus, et al., J. Clin. Oncol.. Vol. 10, pgs, 1682-1689) (1992); Lazarus et al., (1991)).
Eight days before the patient is infused with the autologous peripheral blood progenitor cells, the patient receives chemotherapy over a period of 96 hours (4 days) , with the following chemotherapy agents: 1. Cyclophosphamide in a total dosage of 6g/m2 (1.5 g/m 2/day for 4 days) is given via continuous intravenous infusion at 500 mg/m2 in 1,000 ml normal saline every 8 hours.
2. Thiotepa in a total dosage of 500 mg/m/day for 4 days) is given via continuous intravenous infusion at 125 mg/m2 in 1,000 ml normal saline every 24 hours.
3. Carboplatin in a total dosage of 1,800 mg/m2 (200 mg/m2/day for 4 days) is given via continuous intravenous infusion at 200 mg/m2 in 1,000 ml of 5% dextrose in water every 24 hours.
The patient also receives 500 mg of mesna in 50 ml normal saline IV over 15 minutes every 4 hours for 6 days
(144 hours) , beginning with the first dose of cyclophosphamide.
At least 72 hours after the completion of the chemotherapy, the mesenchymal stem cells are harvested from tissue culture flasks. Cells are collected using bovine trypsin (which is deactivated with soybean trypsin inhibitor) , suspended at 10 x 10* cells/ml in TC199 containing 1% human serum albumin (infusion medium) , washed twice in infusion medium, resuspended at approximately 106 cells/ml, and injected slowly intravenously over 15 minutes to provide a total dosage of from 10 to about 5x106 cells.
The day after the patient receives the mesenchymal stem cells, the frozen autologous peripheral blood progenitor cells are removed from the liquid nitrogen refrigerator, transported to the patient in liquid nitrogen, submersed in a 37°C to 40°C sterile water bath, and infused rapidly intravenously without additional filtering or washing steps. GM-CSF in an amount of 250 μg/m2 then is given as a daily subcutaneous injection, beginning 3 hours after completion of the autologous blood progenitor cell infusion. The GM-CSF is given daily until the peripheral blood neutrophil count exceeds 1,000/μL for three consecutive days.
It is to be understood, however, that the scope of the present invention is not to be limited to the specific embodiments described above. The invention may be practiced other than as particularly described and still be within the scope of the accompanying claims.

Claims

WHAT IS CLAIMED IS:
1. A method for enhancing bone marrow engraftment in an individual in need thereof which comprises administering to said individual (i) mesenchymal stem cells and (ii) a bone marrow graft, wherein said mesenchymal stem cells are administered in an amount effective to promote engraftment of said bone marrow in said individual.
2. The method of claim 1 wherein the mesenchymal stem cells are administered systemically.
3. The method of claim 1 wherein the preparation is administered intravenously.
4. The method of claim l wherein the preparation is injected into said cavity.
5. The method of claim 1 wherein the mesenchymal stem cells are autologous.
6. The method of claim 1 wherein the mesenchymal stem cell preparation is administered concurrently with the bone marrow graft.
7. The method of claim 6 wherein the mesenchymal stem cells are introduced into said individual in a cell suspension also containing bone marrow graft cells.
8. The method of claim 1 wherein the mesenchymal stem cell preparation is administered to the individual prior to administration of the bone marrow graft.
PCT/US1996/000900 1995-01-24 1996-01-22 ENHANCING BONE MARROW ENGRAFTMENT USING MSCs WO1996023058A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU47041/96A AU4704196A (en) 1995-01-24 1996-01-22 Enhancing bone marrow engraftment using mscs

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/377,771 US5733542A (en) 1990-11-16 1995-01-24 Enhancing bone marrow engraftment using MSCS
US08/377,771 1995-01-24

Publications (1)

Publication Number Publication Date
WO1996023058A1 true WO1996023058A1 (en) 1996-08-01

Family

ID=23490468

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/000900 WO1996023058A1 (en) 1995-01-24 1996-01-22 ENHANCING BONE MARROW ENGRAFTMENT USING MSCs

Country Status (3)

Country Link
US (1) US5733542A (en)
AU (1) AU4704196A (en)
WO (1) WO1996023058A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999047163A2 (en) * 1998-03-18 1999-09-23 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
WO2001032189A1 (en) * 1999-10-26 2001-05-10 Osiris Therapeutics, Inc. Supernatant from mesenchymal stem cells for prevention and treatment of immune responses in transplantation
EP1220611A1 (en) * 1999-10-12 2002-07-10 Osiris Therapeutics, Inc. Suppressor cells for prevention and treatment of immune responses in transplantation
WO2007039150A2 (en) * 2005-09-23 2007-04-12 Cellerix, S.L. Cell populations having immunoregulatory activity, method for isolation and uses
EP1841322A1 (en) * 2005-01-11 2007-10-10 Cognate Therapeutics, Inc. Bone marrow stromal cells for immunoprotection of transplanted neural stem cells
US9408876B2 (en) * 2004-09-10 2016-08-09 Cognate Therapeutics, Inc. Liver stromal cells for prevention and treatment of immune responses in transplantation
US10548924B2 (en) 2004-08-25 2020-02-04 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US11273182B2 (en) 2016-03-14 2022-03-15 Takeda Pharmaceutical Company Limited Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in Crohn's disease

Families Citing this family (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6010696A (en) * 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
IL126447A (en) * 1998-10-04 2004-09-27 Vascular Biogenics Ltd Immunological and oral tolerance-inducing composition and use thereof for the prevention and/or for the treatment of atherosclerosis
US20050197283A1 (en) * 1998-10-04 2005-09-08 Vascular Biogenics Ltd. Compositions containing beta 2-glycoprotein I for the prevention and/or treatment of vascular disease
WO2000024261A1 (en) * 1998-10-26 2000-05-04 Philadelphia Health & Education Corporation Stromal cell use
JP2002529508A (en) 1998-11-13 2002-09-10 オシリス セラピューティクス,インコーポレイテッド Method and composition using fibroblasts or supernatant from fibroblasts for suppression of immune response in transplantation
US8297377B2 (en) * 1998-11-20 2012-10-30 Vitruvian Exploration, Llc Method and system for accessing subterranean deposits from the surface and tools therefor
DE60040292D1 (en) * 1999-07-20 2008-10-30 Univ Southern California IDENTIFICATION OF PLURIPOTENTAL PREMESESCHYMAL, PREMHEMOTOPOIETIC TEMPORARY CELLS
US10638734B2 (en) * 2004-01-05 2020-05-05 Abt Holding Company Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US7838289B2 (en) * 2001-02-14 2010-11-23 Abt Holding Company Assay utilizing multipotent adult stem cells
DE20023640U1 (en) * 1999-10-06 2005-06-23 Tigenix N.V. Detecting an embryonic cell surface marker e.g. cartilage-derived morphogenic protein CDMP-1 so as to positively identify viable precursor cells that have entered a post-natal differentiation pathway
US7182781B1 (en) 2000-03-02 2007-02-27 Regeneration Technologies, Inc. Cervical tapered dowel
CA2346362C (en) * 2000-05-26 2010-06-15 Stemcell Technologies Inc. Novel antibody compositions for preparing enriched mesenchymal progenitor preparations
US7442390B2 (en) * 2000-06-05 2008-10-28 University Of South Florida Method for enhancing engraftment of cells using mesenchymal progenitor cells
US7303769B2 (en) * 2000-06-05 2007-12-04 University Of South Florida Method for purifying pluri-differentiated mesenchymal progenitor cells
US7049072B2 (en) * 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
US6936281B2 (en) * 2001-03-21 2005-08-30 University Of South Florida Human mesenchymal progenitor cell
WO2001094541A2 (en) * 2000-06-05 2001-12-13 University Of South Florida Human mesenchymal progenitor cell
CN1449439A (en) 2000-06-26 2003-10-15 株式会社雷诺再生医学研究所 Cell fraction containing cells capable of differentiating into nervous system cells
WO2002016559A2 (en) * 2000-08-22 2002-02-28 The Scripps Research Institute In vivo animal model of human leukemia
US6930222B2 (en) 2000-08-22 2005-08-16 The Scripps Research Institute In vivo animal model of human leukemia
AU2002220209B2 (en) * 2000-12-06 2006-05-25 Robert J. Hariri Method of collecting placental stem cells
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
US20030032179A1 (en) 2000-12-06 2003-02-13 Hariri Robert J. Post-partum mammalian placenta, its use and placental stem cells therefrom
US7311905B2 (en) * 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
KR101012952B1 (en) * 2001-02-14 2011-02-08 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
US6723131B2 (en) 2001-02-28 2004-04-20 The Cleveland Clinic Foundation Composite bone marrow graft material with method and kit
US9969980B2 (en) * 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
WO2003029432A2 (en) * 2001-10-03 2003-04-10 University Of South Florida Human mesenchymal progenitor cell
AU2003278212B2 (en) * 2002-07-31 2009-07-09 Centre National De La Recherche Scientifique Stem cells derived from adipous tissue and differentiated cells derived from said cells
AR047712A1 (en) * 2002-09-07 2006-02-15 Royal Veterinary College TREATMENT METHOD OF A NATURAL SOFT SKELETTIC TISSUE INJURY MANAGING A COMPOSITION OF MESENQUIMATOSE MOTHER CELLS
US9969977B2 (en) * 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
US20040101959A1 (en) * 2002-11-21 2004-05-27 Olga Marko Treatment of tissue with undifferentiated mesenchymal cells
KR101042448B1 (en) * 2002-11-26 2011-06-16 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US7470538B2 (en) * 2002-12-05 2008-12-30 Case Western Reserve University Cell-based therapies for ischemia
NZ566132A (en) * 2003-02-13 2009-09-25 Anthrogenesis Corp Use of umbilical cord blood to treat inflammation, ParkinsonÆs disease or diabetes
ATE482725T1 (en) * 2003-04-01 2010-10-15 Us Dept Of Veteran S Affaires STEM CELL, PROGRESSOR CELL OR TARGET CELL BASED TREATMENT OF MULTI-ORGAN FAILURE.
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
WO2005055929A2 (en) * 2003-12-02 2005-06-23 Celgene Corporation Methods and compositions for the treatment and management of hemoglobinopathy and anemia
CA2512667A1 (en) * 2005-01-07 2006-07-07 Takahiro Ochiya Human hepatocyte-like cells and uses thereof
MX2007010741A (en) * 2005-03-01 2008-03-07 Nat Ct Cell Sciences A composition for creating an artificial bone -marrow like environment and use thereof.
WO2006109312A2 (en) * 2005-04-15 2006-10-19 Vascular Biogenics Ltd. Compositions containing beta 2-glycoprotein i-derived peptides for the prevention and/or treatment of vascular disease
EP1880002A4 (en) * 2005-05-10 2009-03-11 Us Health Therapy of kidney diseases and multiorgan failure with mesenchymal stem cells and mesenchymal stem cell conditioned media
ES2452595T3 (en) 2005-10-13 2014-04-02 Anthrogenesis Corporation Immunomodulation using placental stem cells
KR20210122908A (en) * 2005-12-29 2021-10-12 안트로제네시스 코포레이션 Placental stem cell populations
AU2006332679A1 (en) 2005-12-29 2007-07-12 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US20080286249A1 (en) * 2006-01-12 2008-11-20 Varney Timothy R Use of mesenchymal stem cells for treating genetic diseases and disorders
US20070253931A1 (en) * 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
CN101501185A (en) * 2006-06-09 2009-08-05 人类起源公司 Placental niche and use thereof to culture stem cells
US7993918B2 (en) * 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
DK2084268T3 (en) 2006-10-23 2019-01-21 Celularity Inc METHODS AND COMPOSITIONS FOR TREATING BONE JOIN DEFECTS WITH PLACENTACLE POPULATIONS
WO2008085229A2 (en) * 2006-11-15 2008-07-17 Arteriocyte Inc. Cell-based therapies for treating liver disease
US9387226B2 (en) 2006-11-30 2016-07-12 Medipost Co., Ltd Neural cell proliferation induced through the culture of neural cells with umbilical cord blood-derived mesenchymal stem cells
KR20080068950A (en) * 2007-01-22 2008-07-25 경희대학교 산학협력단 A composition for prevention or treatment of bone marrow damage
DK2120977T3 (en) 2007-02-12 2013-08-12 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
EP2129775A1 (en) * 2007-02-12 2009-12-09 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
AR065584A1 (en) * 2007-03-01 2009-06-17 Cryo Cell Internat Inc OBTAINING INSULATION AND CRIOCONSERVATION OF ENDOMETRIC / MENSTRUAL CELL
US20100172830A1 (en) * 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
US9045735B2 (en) * 2007-04-03 2015-06-02 The Cleveland Clinic Foundation Enrichment of tissue-derived adult stem cells based on retained extracellular matrix material
EP2607477B1 (en) 2007-05-03 2020-09-23 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
TWM322542U (en) * 2007-05-23 2007-11-21 Universal Scient Ind Co Ltd Testing machine
JP2010529987A (en) * 2007-06-15 2010-09-02 ガーネット バイオセラピューティクス インコーポレイテッド Treatment of diseases and disorders using self-replicating colony-forming cells cultured and grown in vitro
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
KR101644659B1 (en) * 2007-09-26 2016-08-01 안트로제네시스 코포레이션 Angiogenic cells from human placental perfusate
KR20210022148A (en) * 2007-09-28 2021-03-02 안트로제네시스 코포레이션 Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
KR20160040739A (en) * 2007-11-07 2016-04-14 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
US20090291061A1 (en) * 2008-05-21 2009-11-26 Riordan Neil H Stem cell therapy for blood vessel degeneration
KR20110050521A (en) * 2008-08-20 2011-05-13 안트로제네시스 코포레이션 Improved cell composition and methods of making the same
KR101903049B1 (en) * 2008-08-20 2018-11-07 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
KR20110050688A (en) * 2008-08-22 2011-05-16 안트로제네시스 코포레이션 Methods and compositions for treatment of bone defects with placental cell populations
EP2204442A1 (en) 2008-10-01 2010-07-07 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method of enriching stem cells in culture
KR20100054711A (en) * 2008-11-14 2010-05-25 메디포스트(주) Composition comprising mesenchymal stem cells or culture solution of mesenchymal stem cells for the prevention or treatment of neural diseases
AU2009316541B2 (en) * 2008-11-19 2015-08-06 Celularity Inc. Amnion derived adherent cells
ES2731340T3 (en) * 2008-11-21 2019-11-15 Celularity Inc Treatment of diseases, disorders or lung conditions using placental cells
CA2767014C (en) * 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
EP3284818B1 (en) * 2010-01-26 2022-03-09 Celularity Inc. Treatment of bone-related cancers using placental stem cells
KR20230054905A (en) 2010-04-07 2023-04-25 셀룰래리티 인코포레이티드 Angiogenesis using placental stem cells
WO2011127113A1 (en) 2010-04-08 2011-10-13 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
ES2666746T3 (en) 2010-07-13 2018-05-07 Anthrogenesis Corporation Methods to generate natural cytolytic lymphocytes
US9725689B2 (en) 2010-10-08 2017-08-08 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
KR101615161B1 (en) 2011-02-02 2016-04-25 메디포스트(주) Use of icam-1 for prevention or treatment of neurological diseases
PL2714059T3 (en) 2011-06-01 2019-04-30 Celularity Inc Treatment of pain using placental stem cells
US9782441B2 (en) * 2011-07-11 2017-10-10 Albert Einstein College Of Medicine, Inc. Stromal cell therapy in treatment of radiation injury
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
JP6562632B2 (en) 2012-02-14 2019-08-21 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Systemic delivery and controlled expression of the paracrine gene for cardiovascular disease and other conditions
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
EP3456817A1 (en) 2013-02-06 2019-03-20 Nc Medical Research Inc. Cell therapy for the treatment of neurodegeneration
WO2015073913A1 (en) 2013-11-16 2015-05-21 Terumo Bct, Inc. Expanding cells in a bioreactor
WO2015148704A1 (en) 2014-03-25 2015-10-01 Terumo Bct, Inc. Passive replacement of media
BR112016022854A2 (en) 2014-04-03 2018-01-16 Univ California method for treating, alleviating or protecting (preventing), slowing down or reversing a disease, method for treating, alleviating or protecting (preventing) a disease, use, and, nucleic acid or gene
WO2016015007A1 (en) 2014-07-25 2016-01-28 Recellerate, Inc. Methods of treating exercise-induced pulmonary hemorrhage
CN106715676A (en) 2014-09-26 2017-05-24 泰尔茂比司特公司 Scheduled feed
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US10463767B2 (en) 2016-04-22 2019-11-05 Vivex Biologics Group, Inc. Moldable bone composition
US11253629B2 (en) 2016-04-22 2022-02-22 Vivex Biologics Group, Inc. Bone gel sheet composition and method of manufacture
US11253630B2 (en) 2016-04-22 2022-02-22 Vivex Biologics Group, Inc. Malleable demineralized bone composition and method of manufacture
US10596298B2 (en) 2016-04-22 2020-03-24 Vivex Biologics Group, Inc. Malleable demineralized bone composition and method of manufacture
US9788950B1 (en) 2016-04-22 2017-10-17 Vivex Biomedical, Inc. Cohesive bone composition
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5226914A (en) * 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5461034A (en) * 1989-02-23 1995-10-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Osteogenic growth polypeptides identified from regenerating bone marrow

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5226914A (en) * 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999047163A3 (en) * 1998-03-18 1999-11-18 Osiris Therapeutics Inc Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
AU755888B2 (en) * 1998-03-18 2003-01-02 Mesoblast International Sarl Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US6875430B2 (en) * 1998-03-18 2005-04-05 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
WO1999047163A2 (en) * 1998-03-18 1999-09-23 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
EP1220611A1 (en) * 1999-10-12 2002-07-10 Osiris Therapeutics, Inc. Suppressor cells for prevention and treatment of immune responses in transplantation
EP1220611A4 (en) * 1999-10-12 2004-12-22 Osiris Therapeutics Inc Suppressor cells for prevention and treatment of immune responses in transplantation
EP2269616A3 (en) * 1999-10-26 2012-06-27 Osiris Therapeutics, Inc. Supernatant from mesenchymal stem cells for prevention and treatment of immune responses in transplantation
WO2001032189A1 (en) * 1999-10-26 2001-05-10 Osiris Therapeutics, Inc. Supernatant from mesenchymal stem cells for prevention and treatment of immune responses in transplantation
AU782064B2 (en) * 1999-10-26 2005-06-30 Mesoblast International Sarl Supernatant from mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US10548924B2 (en) 2004-08-25 2020-02-04 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US10780132B2 (en) 2004-08-25 2020-09-22 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US9408876B2 (en) * 2004-09-10 2016-08-09 Cognate Therapeutics, Inc. Liver stromal cells for prevention and treatment of immune responses in transplantation
EP1841322A4 (en) * 2005-01-11 2009-12-09 Cognate Therapeutics Inc Bone marrow stromal cells for immunoprotection of transplanted neural stem cells
EP1841322A1 (en) * 2005-01-11 2007-10-10 Cognate Therapeutics, Inc. Bone marrow stromal cells for immunoprotection of transplanted neural stem cells
US10758575B2 (en) 2005-06-24 2020-09-01 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US11672831B2 (en) 2005-06-24 2023-06-13 Takeda Pharmaceutical Company Limited Use of adipose tissue-derived stromal stem cells in treating fistula
US11660318B2 (en) 2005-06-24 2023-05-30 Takeda Pharmaceutical Company Limited Use of adipose tissue-derived stromal stem cells in treating fistula
WO2007039150A3 (en) * 2005-09-23 2007-08-23 Cellerix Sl Cell populations having immunoregulatory activity, method for isolation and uses
WO2007039150A2 (en) * 2005-09-23 2007-04-12 Cellerix, S.L. Cell populations having immunoregulatory activity, method for isolation and uses
US9943550B2 (en) 2005-09-23 2018-04-17 Tigenix, S.A.U. Cell populations having immunoregulatory activity, method for isolation and uses
EP2340847A3 (en) * 2005-09-23 2016-11-09 Cellerix, S.A. Cell populations having immunoregulatory activity, method for isolation and uses
JP2014221045A (en) * 2005-09-23 2014-11-27 タイジェニックス、ソシエダッド、アノニマ、ウニペルソナルTigenix S.A.U. Cell population having immunoregulatory activity, isolation method and use thereof
JP2009508507A (en) * 2005-09-23 2009-03-05 セリェリクス、ソシエダッド、リミターダ Cell population having immunomodulatory activity, isolation methods and uses
US11273182B2 (en) 2016-03-14 2022-03-15 Takeda Pharmaceutical Company Limited Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in Crohn's disease

Also Published As

Publication number Publication date
US5733542A (en) 1998-03-31
AU4704196A (en) 1996-08-14

Similar Documents

Publication Publication Date Title
US5733542A (en) Enhancing bone marrow engraftment using MSCS
US6010696A (en) Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
JP4441115B2 (en) Methods and uses of human non-self mesenchymal stem cells
US11236324B2 (en) Isolation of stem cells from adipose tissue by ultrasonic cavitation, and methods of use
Lazarus et al. Human bone marrow-derived mesenchymal (stromal) progenitor cells (MPCs) cannot be recovered from peripheral blood progenitor cell collections
JP3662029B2 (en) Methods for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US7037892B2 (en) Hematopoietic stem cell proliferating agents
US11821005B2 (en) Umbilical cord mesenchymal stem cells (MSCs) and culture method and application thereof
JP2002506082A5 (en)
US11447748B2 (en) Encapsulated adipose-derived stem cells, methods for preparation and theraputic use
MXPA04007732A (en) Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells.
EP1999250B1 (en) Method of cultivation of human mesenchymal stem cells, particularly for the treatment of non-healing fractures, and bioreactor for carrying out this cultivation method
PT1028737E (en) Human mesenchymal stem cells from peripheral blood
WO2014053418A9 (en) Method for obtaining mesenchymal stem cells and use thereof
WO2001094541A2 (en) Human mesenchymal progenitor cell
JP2014529406A (en) Isolation and therapeutic use of perivascular medicinal cells
Fang et al. Cotransplantation of haploidentical mesenchymal stem cells to reduce the risk of graft failure in a patient with refractory severe aplastic anemia.
CN110840914B (en) Method for alleviating or improving vascular disorders using cell therapeutic agent
AU2014211790B2 (en) Use of allogeneic interstitial vessel-layer cell and allogeneic mesenchymal progenitor cell for preventing or treating osteoarthritis
US8383406B2 (en) Method for stimulating the proliferation of differentiated cells belonging to the chondrogenic lineage
KR100641684B1 (en) Agent for engraftment of allogenic hematopoietic stem cells using human adipose stromal cells
Delgado et al. Uses of mesenchymal stem cells
TW202342735A (en) Methods of producing human mesenchymal stem cells from foreskins and uses thereof
US20040076605A1 (en) Method of regenerating human tissue
Blood A New Source of Mesenchymal Stem Cells for Articular Cartilage Repair

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP MX

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase