US20100172885A1 - Multipotent Adult Stem Cells And Uses of Multipotent Adult Stem Cells To Treat Inflammation - Google Patents

Multipotent Adult Stem Cells And Uses of Multipotent Adult Stem Cells To Treat Inflammation Download PDF

Info

Publication number
US20100172885A1
US20100172885A1 US12/727,971 US72797110A US2010172885A1 US 20100172885 A1 US20100172885 A1 US 20100172885A1 US 72797110 A US72797110 A US 72797110A US 2010172885 A1 US2010172885 A1 US 2010172885A1
Authority
US
United States
Prior art keywords
stem cells
integrin
cells
mesenchymal stem
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/727,971
Inventor
Mark F. Pittenger
Sudeepta Aggarwal
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.)
Mesoblast International SARL
Original Assignee
Osiris Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39772836&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100172885(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Osiris Therapeutics Inc filed Critical Osiris Therapeutics Inc
Priority to US12/727,971 priority Critical patent/US20100172885A1/en
Assigned to OSIRIS THERAPEUTICS, INC. reassignment OSIRIS THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AGGARWAL, SUDEEPTA, PITTENGER, MARK F.
Publication of US20100172885A1 publication Critical patent/US20100172885A1/en
Assigned to MESOBLAST INTERNATIONAL SÀRL reassignment MESOBLAST INTERNATIONAL SÀRL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OSIRIS THERAPEUTICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • 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
    • A61K38/20Interleukins [IL]
    • A61K38/2026IL-4
    • 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
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0664Dental pulp stem cells, Dental follicle stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0665Blood-borne mesenchymal stem cells, e.g. from umbilical cord blood
    • 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
    • C12N5/0666Mesenchymal stem cells from hair follicles
    • 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
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • MSCs Mesenchymal stem cells
  • MSCs Mesenchymal stem cells
  • adipocytes a kind of multipotent stem cell that can differentiate readily into lineages including osteoblasts, myocytes, chondrocytes, and adipocytes.
  • neuronal-like precursors Woodbury, et al., J. Neurosci. Res ., Vol. 69, pg. 908 (2002); Sanchez-Ramos, et al., Exp. Neurol ., Vol. 171, pg. 109 (2001)), cardiomyocytes (Toma, et al., Circulation , Vol. 105, pg. 93 (2002); Fakuda, Artif. Organs , Vol. 25, pg. 187 (2001)) and possibly other cell types.
  • MSCs have been shown to provide effective feeder layers for expansion of hematopoietic and embryonic stem cells (Eaves, et al., Ann.
  • MSCs can be useful in the repair and/or regeneration of damaged bone, cartilage, meniscus or myocardial tissues (DeKok, et al., Clin. Oral Implants Res ., Vol. 14, pg. 481 (2003)); Wu, et al., Transplantation , Vol. 75, pg. 679 (2003); Noel, et al., Curr. Opin. Investig. Drugs , Vol. 3, pg.
  • MSCs express major histocompatibility complex (MHC) class I antigen on their surface, but do not express MHC class II (Le Blanc, et al., Exp. Hematol ., Vol. 31, pg. 890 (2003); Potian, et al., J. Immunol ., Vol. 171, pg. 3426 (2003)) and do not express B7 or CD40 co-stimulatory molecules (Majumdar, et al., J. Biomed. Sci ., Vol. 10, pg. 228 (2003)), suggesting that these cells have a low-immunogenic phenotype (Tse, et al., Transplantation , Vol. 75, pg. 389 (2003)).
  • MHC major histocompatibility complex
  • MSCs also inhibit T-cell proliferative responses in an MHC-independent manner (Bartholomew, et al., Exp. Hematol ., Vol. 30, pg. 42 (2002); Devine, et al., Cancer J ., Vol. 7, pg. 576 (2001); DiNicola, et al., Blood , Vol. 99, pg. 3838 (2002)).
  • These immunological properties of MSCs can enhance their transplant engraftment and limit the ability of the recipient's immune system to recognize and reject allogeneic cells following transplantation.
  • MSCs that modulate the immune response and support hematopoiesis together with their ability to differentiate into appropriate cell types under local stimuli make them desirable stem cells for cellular transplantation studies (Majumdar, et al., Hematother. Stem Cell Res ., Vol. 9, pg. 841 (2000); Haynesworth, et al., J. Cell. Physiol ., Vol. 166, pg. 585 (1996).
  • the present technology generally relates to multipotent adult stem cells, such as adult bone marrow-derived stem cells. More particularly, the present technology relates to new and heretofore unappreciated uses for multipotent adult stem cells, such as mesenchymal stem cells, including, but not limited to promoting angiogenesis in various tissues and organs, treating autoimmune diseases, treating allergic responses, treating cancer, treating inflammatory diseases and disorders, promoting wound healing, treating inflammation, and repairing epithelial damage.
  • multipotent adult stem cells can be used, for example, to treat an autoimmune disease, treat an inflammatory response, treat an allergic disease, treat a pulmonary disease having fibrotic and/or inflammatory components, repair epithelial damage, and promote wound healing in subjects, including a human subject.
  • Autoimmune diseases that can be treated with multipotent adult stem cells include, for example, Type 1 Diabetes, inflammatory bowel disease, Crohn's disease, and uveitis.
  • Pulmonary diseases that can be treated with multipotent adult stem cells include, but are not limited to, Acute Respiratory Distress Syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD), and asthma.
  • Inflammatory responses including those associated with autoimmune diseases or pulmonary diseases, can be reduced with multipotent adult stem cells.
  • An inflammatory response can be reduced by, for example, reducing the production or expression of pro-inflammatory mediators, increasing the production or expression of anti-inflammatory mediators, or a combination thereof.
  • Multipotent adult stem cells of the present technology have the capacity to differentiate into at least one cell type of each of the mesodermal, ectodermal, and endodermal lineages.
  • the cells can be induced to differentiate into cells of at least osteoblast, chondrocyte, adipocyte, fibroblast, marrow stroma, skeletal muscle, smooth muscle, cardiac muscle, endothelial, epithelial, hematopoietic, glial, neuronal or oligodendrocyte cell types, among others.
  • Multipotent adult stem cells of the present technology are capable of differentiating into at least one cell type of at least one of the endodermal, ectodermal, or mesodermal embryonic lineages.
  • Bone marrow is an accessible and renewable source of adult multipotent stem cells that can be greatly expanded in culture.
  • bone marrow-derived mesenchymal stem cells are multipotent cells that have been identified and cultured from various avian and mammalian species.
  • the present technology provides one or more cell preparations comprising adult bone marrow-derived stem cells in one or more doses effective to treat an inflammatory response in a subject.
  • the subject can be a human having, for example, inflammatory bowel disease.
  • the stem cells can be capable of differentiating into at least one cell type of each of the endodermal, ectodermal, and mesodermal embryonic lineages.
  • the effective dose of the cell preparation can contain a sufficient number of stem cells to provide about 1 ⁇ 10 5 to about 1 ⁇ 10 7 cells per kilogram of the subject.
  • the subject can be a mammal.
  • the mammal can be, for example, a primate, including a human and a non-human primate.
  • Administration of the cell preparation can, for example, elevate interferon-beta levels in the subject.
  • the present technology also provides a method of treating inflammatory bowel disease in a subject, comprising administering to the subject allogeneic multipotent adult stem cells in an amount effective to treat inflammatory bowel disease.
  • the subject can be an animal.
  • the animal can be, for example, a primate, including a human and a non-human primate.
  • the multipotent adult stem cells can be administered systemically, such as intravenously, intraarterially, or intraperitoneally.
  • the multipotent adult stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, such as a pharmaceutically acceptable liquid medium.
  • the multipotent adult stem cells can be administered as a suspension of cells.
  • the amount effective to treat inflammatory bowel disease can be about 1 ⁇ 10 5 to about 1 ⁇ 10 7 cells per kilogram of the subject.
  • the present technology further provides a method of treating a gastrointestinal autoimmune disease in a subject, comprising administering to the subject allogeneic multipotent adult stem cells in an amount effective to repair or regenerate intestinal tissue.
  • the autoimmune disease can be selected from the group consisting of Crohn's disease, inflammatory bowel disease, and autoimmune gastritis.
  • the subject can be an animal.
  • the animal can be, for example, a primate, including a human and a non-human primate.
  • the multipotent adult stem cells can be administered systemically, such as intravenously, intraarterially, or intraperitoneally.
  • the multipotent adult stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, such as a pharmaceutically acceptable liquid medium.
  • the multipotent adult stem cells can be administered as a suspension of cells.
  • the amount effective to repair or regenerate intestinal tissue can be about 1 ⁇ 10 5 to about 1 ⁇ 10 7 cells per kilogram of the subject.
  • the multipotent adult stem cells of the present technology can be positive for one or more cell surface markers selected from, for example, integrin al (CD49a); integrin ⁇ 2 (CD49b); integrin ⁇ 3 (CD49c); integrin ⁇ 5 (CD49e); integrin ⁇ V (CD51); integrin ⁇ 1 (CD29); integrin ⁇ 3 (CD61); integrin ⁇ 4 (CD104); IL-1R (CD121a); IL-3R ⁇ (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFN ⁇ R (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGF ⁇ 1R; TGF ⁇ IIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD
  • the stem cells can also be positive for each of CD49b, CD49e, and CD140a. Further, the stem cells can be negative for one or more cell surface markers such as integrin ⁇ 4 (CD49d); integrin ⁇ L (CD11a); integrin C ⁇ 2 (CD18); CD4; CD14; CD34; CD45; IL-2R (CD25); EGFR-3; Fas ligand; ICAM-3 (CD50); E-Selectin (CD62E); P-Selectin (CD62P); vW Factor; cadherin 5; or Lewis x (CD15).
  • the multipotent adult stem cells of the present technology can be adult bone marrow-derived stem cells such as, for example, mesenchymal stem cells.
  • the present technology also provides a method of repairing and/or regenerating intestinal tissue in a subject, comprising treating the subject with allogeneic mesenchymal stem cells.
  • the subject can be an animal.
  • the animal can be, for example, a primate, including a human and a non-human primate.
  • the stem cells can be from an adult.
  • the mesenchymal stem cells can be administered to the subject as part of, for example, a pharmaceutical formulation having a pharmaceutically acceptable carrier, such as a liquid injectable carrier, a liquid topical carrier, a gel injectable carrier, a gel topical carrier, a solid matrix, and combinations thereof.
  • the mesenchymal stem cell pharmaceutical formulation can be administered via intravenous, intraarterial, or intraperitoneal injection.
  • Mesenchymal stem cells can be administered in an amount effective to repair or regenerate intestinal tissue in a subject.
  • the amount effective to repair or regenerate intestinal tissue can be about 1 ⁇ 10 5 to about 1 ⁇ 10 7 cells per kilogram of the subject
  • multipotent adult stem cells regulate the production of various factors that affect several steps in the inflammatory and/or immune response process.
  • the multipotent adult stem cells of the present technology can be employed in the treatment of disease conditions and disorders involving the immune system, or diseases, conditions, or disorders involving inflammation, epithelial damage, or allergic responses.
  • Such diseases, conditions, and disorders include, but are not limited to, autoimmune diseases, allergies, arthritis, inflamed wounds, alopecia araeta (baldness), periodontal diseases including gingivitis and periodontitis, and other diseases, conditions or disorders involving an immune response.
  • multipotent adult stem cells including mesenchymal stem cells, express and secrete vascular endothelial growth factor, or VEGF, which promotes angiogenesis by stimulating the formation of new blood vessels.
  • Mesenchymal stem cells also stimulate peripheral blood mononuclear cells (PBMCs) to produce VEGF.
  • PBMCs peripheral blood mononuclear cells
  • DCs dendritic cells
  • IFN- ⁇ Interferon-Beta
  • FIG. 1 MSCs modulate dendritic cell functions.
  • A Flow cytometric analysis of mature monocytic DC1 cells using antibodies against HLA-DR and CD11c and of plasmacytoid DC2 cells using antibodies against HLA-DR and CD123 (IL-3 receptor).
  • B MSCs inhibit TNF- ⁇ secretion (primary y-axis) and increase IL-10 secretion (secondary y-axis) from activated DC1 and DC2 respectively.
  • C MSCs cultured with mature DC1 cells inhibit IFN- ⁇ secretion (primary y-axis) by T cells and increase IL-4 levels (secondary y-axis) as compared to MSC or DC alone.
  • FIG. 2 MSCs inhibit pro-inflammatory effector T cell function.
  • A Flow cytometric analysis of T reg cell numbers (in %) by staining PBMCs or the non-adherent fraction in MSC+PBMC culture (MSC+PBMC) with FITC-conjugated CD4 (x-axis) and PE conjugated CD25 (y-axis) antibodies. Gates were set based on isotype control antibodies as background. Graphs are representative of 5 independent experiments.
  • B Th1 cells generated in presence of MSCs secreted reduced levels of IFN- ⁇ (primary Y-axis) and Th2 cells generated in presence of MSCs secreted increased amounts of IL-4 (secondary y-axis) in cell culture supernatants.
  • C MSCs inhibit IFN- ⁇ secretion from purified NK cells cultured for 0, 24, or 48 hours in a 24-well plate. Data shown are mean ⁇ SD cytokine secretion in one experiment and are representative of 3 independent experiments.
  • FIG. 3 MSCs lead to increased numbers of T reg cell population and increased GITR expression.
  • MLR mixed lymphocyte reaction
  • PHA phytohemagglutinin
  • T reg population generated in the presence of MSCs (lane 3) was similar functionally to the T reg cells generated in the absence of MSCs (lane 2).
  • B PBMCs were cultured for 3 days in the absence (top plot) or presence (bottom plot) of MSCs (MSC to PBMC ratio 1:10), following which the non-adherent fraction was harvested and immunostained with FITC-labeled GITR and PE-labeled CD4. Results show a greater than twofold increase in GITR expression in cells cultured in the presence of MSCs.
  • FIG. 4 MSCs produce PGE 2 and blocking PGE 2 reverses MSC-mediated immuno-modulatory effects.
  • A PGE 2 secretion (mean ⁇ SD) in culture supernatants obtained from MSCs cultured in the presence or absence of PGE 2 blockers NS-398 or indomethacin (Indometh.) at various concentrations. Inhibitor concentrations are in ⁇ M and data presented are values obtained after 24 hour culture
  • B COX-1 and COX-2 expression in MSCs and PBMCs using real-time RT-PCR.
  • MSCs expressed significantly higher levels of COX-2 as compared to PBMCs, and when MSCs were cultured in presence of PBMCs, there was a >3-fold increase in COX-2 expression in MSCs. Representative data from 1 of 3 independent experiments are shown. The MSC+PBMC cultures were setup in a trans-well chamber plate where MSCs were plated onto the bottom chamber and PBMCs onto the top chamber.
  • C Presence of PGE 2 blockers indomethacin (Ind.) or NS-398 increases TNF- ⁇ secretion from activated DCs (open bars) and IFN- ⁇ secretion from Th1 cells (hatched bars) as compared to controls.
  • FIG. 5 Constituitive MSC cytokine secretion is elevated in the presence of allogeneic PBMCs.
  • cytokines IL-6 and VEGF, lipid mediator PGE 2 , and matrix metalloproteinase 1 (pro MMP-1) were analyzed.
  • the MSCs produced IL-6, VEGF, and PGE 2 constituitively, and the levels of these factors increased upon co-culture with PBMCs, thereby suggesting that MSCs can play a role in modulating immune functions in an inflammatory setting.
  • FIG. 6 MSCs inhibit mitogen-induced T-cell proliferation in a dose-dependent manner. Increasing numbers of allogeneic PBMCs were incubated with constant numbers of MSCs (2,000 cells/well) plated on a 96-well plate in the presence or absence of PHA (2.5 mg/ml) for 72 hours, and 3H thymidine incorporation determined (in counts per minute, or cpm). There was a dose-dependent inhibition of the proliferation of PHA-treated PBMCs in the presence of MSCs. Representative results from 1 of 3 independent experiments are shown. Similar results were reported by LeBlanc, et al., Scand J. Immunol., Vol. 57, pg. 11 (2003).
  • FIG. 7 Schematic diagram of proposed MSC mechanism of action.
  • MSCs mediate their immuno-modulatory effects by affecting cells from both the innate (DC-pathways 2-4; and NK-pathway 6) and adaptive (T-pathways 1 and 5 and B-pathway 7) immune systems.
  • immature DCs migrate to the site of potential entry, mature and acquire an ability to prime na ⁇ ve T cells (by means of antigen specific and co-stimulatory signals) to become protective effector T cells (cell-mediated Th1 or humoral Th2 immunity).
  • MSCs can alter the outcome of immune response by limiting the ability of DCs to mount a cell-mediated response (pathway 2) or by promoting the ability to mount a humoral response (pathway 4). Also, when mature effector T cells are present, MSCs can interact with them to skew the balance of Th1 (pathway 1) responses towards TH2 responses (pathway 5), and probably towards an increased IgE producing B cell activity (pathway 7), desirable outcomes for suppression of GvHD and autoimmune disease symptoms.
  • Th1 pathway 1
  • TH2 responses pathway 5
  • IgE producing B cell activity pathway 7
  • MSCs in their ability to result in an increased generation of T reg population can result in a tolerant phenotype and can aid a recipient host by dampening bystander inflammation in their local micro-environment.
  • Dashed line (----) represents proposed mechanism.
  • a method of treating a disease selected from the group consisting of autoimmune diseases and graft-versus-host disease in an animal including, for example, a human.
  • the method comprises at least the step of administering to the animal mesenchymal stem cells in an amount effective to treat the disease in the animal.
  • IL-10 Interleukin-10
  • T reg cells regulatory T-cells
  • DC dendritic cells
  • Autoimmune diseases which can be treated in accordance with the present technology include, but are not limited to, multiple sclerosis, Type 1 diabetes, rheumatoid arthritis, uveitis, autoimmune thyroid disease, inflammatory bowel disease, scleroderma, Graves' Disease, lupus, Crohn's disease, autoimmune lymphoproliferative disease (ALPS), demyelinating disease, autoimmune encephalomyelitis, autoimmune gastritis (AIG), and autoimmune glomerular diseases.
  • graft-versus-host disease can be treated. It is to be understood, however, that the scope of the present technology is not to be limited to the treatment of the specific diseases mentioned herein.
  • the mesenchymal stem cells are administered to a mammal.
  • the mammal can be a primate, including human and non-human primates.
  • the multipotent adult stem cell therapy is based, for example, on the following sequence: harvest of stem cell-containing tissue such as, for example, bone marrow; isolation and/or expansion of stem cells; and administration of the stem cells, with or without biochemical or genetic manipulation, to the animal.
  • the mesenchymal stem cells that are administered can be, for example, a homogeneous composition or a mixed cell population enriched in mesenchymal stem cells.
  • the mesenchymal stem cell compositions can be obtained, for example, by culturing adherent bone marrow or periosteal cells.
  • Adult bone marrow-derived stem cells, such as bone marrow-derived mesenchymal stem cells, can be identified by specific cell surface markers, which are capable of being bound by unique monoclonal antibodies.
  • a method for obtaining a cell population enriched in mesenchymal stem cells is described in, for example, U.S. Pat. No. 5,486,359, the contents of which are hereby incorporated by reference in its entirety.
  • Adult bone marrow contains multipotent stem cells, including mesenchymal stem cells.
  • Alternative sources for multipotent adult stem cells such as mesenchymal stem cells include, but are not limited to, blood, skin, cord blood, muscle, fat, bone, and perichondrium.
  • Multipotent adult stem cells can be obtained from a variety of sources, including bone marrow.
  • multipotent adult stem cells such as human mesenchymal stem cells can be obtained from bone marrow from a number of different sources, including plugs of femoral head cancellous bone pieces, patients with degenerative joint disease during hip or knee replacement surgery, and aspirated marrow from normal donors or oncology patients who have marrow harvested for future bone marrow transplantation.
  • Harvested marrow can be prepared for cell culture by a number of different mechanical isolation processes depending upon the source of the harvested marrow (i.e., the presence of bone chips, peripheral blood, etc.) that are well known in the art.
  • Exemplary culture media and culture conditions are identified in, for example, U.S. Pat. No. 5,486,359 and include media and conditions that allow for expansion, growth, and isolation of mesenchymal stem cells, without differentiation.
  • Multipotent stem cells isolated from human adult bone marrow can be surface antigen positive for integrin al (CD49a); integrin ⁇ 2 (CD49b); integrin ⁇ 3 (CD49c); integrin ⁇ 5 (CD49e); integrin ⁇ V (CD51); integrin ⁇ 1 (CD29); integrin ⁇ 3 (CD61); integrin ⁇ 4 (CD104); IL-1R (CD121a); IL-3R ⁇ (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFN ⁇ R (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGF ⁇ 1R; TGF ⁇ IIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD166); hyalur
  • Cell isolated from human adult bone marrow can be surface antigen negative for integrin ⁇ 4 (CD49d); integrin ⁇ L (CD11a); integrin C ⁇ 2 (CD18); CD4; CD14; CD34; CD45; IL-2R (CD25); EGFR-3; Fas ligand, ICAM-3 (CD50); E-Selectin (CD62E); P-Selectin (CD62P); vW Factor; cadherin 5; Lewis x (CD15); and combinations thereof.
  • integrin ⁇ 4 CD49d
  • integrin ⁇ L CD11a
  • integrin C ⁇ 2 CD18
  • CD4 CD14
  • CD34 CD45
  • IL-2R CD25
  • EGFR-3 Fas ligand, ICAM-3
  • CD50 E-Selectin
  • CD62P P-Selectin
  • vW Factor cadherin 5; Lewis x (CD15); and combinations thereof.
  • Cell preparations having greater than about 95%, usually greater than about 98%, of multipotent adult human stem cells can be achieved using techniques for isolation, purification, and culture expansion of stem cells.
  • isolated, cultured adult bone marrow-derived stem cells such as mesenchymal stem cells can comprise a single phenotypic population (about 95% or about 98% homogeneous) by flow cytometric analysis of expressed surface antigens.
  • the desired cells in such composition can be identified, for example, by expression of a cell surface marker (e.g., CD73 or CD105) specifically bound by an antibody produced from hybridoma cell line SH2, ATCC accession number HB 10743, an antibody produced from hybridoma cell line SH3, ATCC accession number HB 10744, or an antibody produced from hybridoma cell line SH4, ATCC accession number HB 10745.
  • a cell surface marker e.g., CD73 or CD105
  • Such antibodies selectively bind bone marrow-derived mesenchymal stem cells and, therefore, can be used to identify, quantify, isolate, or purify mesenchymal stem cells from bone marrow samples.
  • the mesenchymal stem cells can be administered by a variety of procedures.
  • the mesenchymal stem cells can be administered systemically, such as by intravenous, intraarterial, or intraperitoneal administration.
  • the mesenchymal stem cells can be from a spectrum of sources including autologous, allogeneic, or xenogeneic.
  • the mesenchymal stem cells are administered in an amount effective to treat an autoimmune disease or graft-versus-host disease in an animal.
  • the mesenchymal stem cells can be administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 2 ⁇ 10 6 cells/kg to about 4 ⁇ 10 6 cells/kg. In still other embodiments, the mesenchymal stem cells are administered in an amount of about 3 ⁇ 10 6 cells/kg.
  • the amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the autoimmune disease to be treated, and the extent and severity thereof.
  • the mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier.
  • the mesenchymal stem cells can be administered as a cell suspension in a pharmaceutically acceptable liquid medium or gel for injection or topical application.
  • a method of treating an inflammatory response in an animal comprises administering to the animal mesenchymal stem cells in an amount effective to treat the inflammatory response in the animal.
  • the mesenchymal stem cells prevent or reverse an inflammatory response by, for example, increasing expression, production, and/or secretion of pro-inflammatory cytokines such as, for example, tumor necrosis factor-alpha (TNF- ⁇ ) and Interferon- ⁇ (IFN- ⁇ ); decreasing expression, production, and/or secretion of anti-inflammatory cytokines such as, for example, IL-10 and IL-4; and/or combinations thereof.
  • the mesenchymal stem cells promote T-cell maturation to regulatory T-cells (T reg ), thereby controlling inflammatory responses. It is also believed that the mesenchymal stem cells inhibit T helper 1 cells (Th1 cells), thereby decreasing the expression of the IFN- ⁇ in certain inflammatory reactions, such as those associated with psoriasis, for example.
  • the inflammatory responses which can be treated are those associated with psoriasis.
  • the mesenchymal stem cells can be administered to an animal such that the mesenchymal stem cells prevent or reduce inflammation in the brain by, for example, contacting or secreting factors that affect, microglia and/or astrocytes in the brain.
  • the mesenchymal stem cells limit neurodegeneration caused by activated glial cells in diseases or disorders such as Alzheimer's disease, Parkinson's disease, stroke, or brain cell injuries.
  • the mesenchymal stem cells can be administered to an animal such that the mesenchymal stem cells reduce skin inflammation as can occur in psoriasis, chronic dermatitis, and contact dermatitis by, for example, contacting or secreting factors that affect, keratinocytes and Langerhans cells in the epidermis of the skin.
  • this embodiment is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells can contact the keratinocytes and Langerhans cells in the epidermis, and alter the expression of T-cell receptors and cytokine secretion profiles, leading to decreased expression of TNF- ⁇ and increased regulatory T-cell (T reg cell) population.
  • the mesenchymal stem cells can be used to reduce inflammation in the bone, as occurs in arthritis and arthritis-like conditions, including but not limited to, osteoarthritis and rheumatoid arthritis, and other arthritic diseases such as ankylosing spondylitis, avascular necrosis (osteonecrosis), fibromyalgia, juvenile dermatomyositis, juvenile rheumatoid arthritis, juvenile spondyloarthopathy, lyme disease, marfan syndrome, myositis, osteogenesis imperfecta, osteoporosis, Paget's disease, Raynaud's Phenomenon, scleroderma, Sjorgren's Syndrome, and systemic lupus erythematosus.
  • osteoarthritis and rheumatoid arthritis and other arthritic diseases such as ankylosing spondylitis, avascular necrosis (osteonecrosis), fibromyalgia, juvenile derm
  • the mesenchymal stem cells can be used to limit inflammation in the gut and liver during inflammatory bowel disease and chronic hepatitis, respectively.
  • the scope of this aspect of the present technology is not intended to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells promote increased secretion of IL-10 and the generation of T reg cells.
  • the mesenchymal stem cells can be used to inhibit excessive neutrophil and macrophage activation in pathological conditions such as sepsis and trauma, including burn injury, surgery, and transplants.
  • pathological conditions such as sepsis and trauma, including burn injury, surgery, and transplants.
  • the mesenchymal stem cells promote secretion of suppressive cytokines such as IL-10, and inhibit macrophage migration inhibitory factor (MIF).
  • MIF macrophage migration inhibitory factor
  • the mesenchymal stem cells can be used to control inflammation in immune privileged sites such as the eye, including the cornea, lens, pigment epithelium, and retina, brain, spinal cord, pregnant uterus and placenta, ovary, testes, adrenal cortex, liver, and hair follicles.
  • immune privileged sites such as the eye, including the cornea, lens, pigment epithelium, and retina, brain, spinal cord, pregnant uterus and placenta, ovary, testes, adrenal cortex, liver, and hair follicles.
  • the mesenchymal stem cells can be used to treat tissue damage associated with end-stage renal disease (ESRD) infections during dialysis and/or glomerulonephritis.
  • ESRD end-stage renal disease
  • mesenchymal stem cells can promote renal repair.
  • Mesenchymal stem cells also express and secrete vascular endothelial growth factor, or VEGF, which stimulates new blood vessel formation, which should aid in the repair of damaged kidney tissue.
  • VEGF vascular endothelial growth factor
  • the mesenchymal stem cells can be used to control viral infections such as influenza, hepatitis C, Herpes Simplex Virus, vaccinia virus infections, and Epstein-Barr virus.
  • viral infections such as influenza, hepatitis C, Herpes Simplex Virus, vaccinia virus infections, and Epstein-Barr virus.
  • IFN- ⁇ Interferon-Beta
  • the mesenchymal stem cells can be used to control parasitic infections such as Leishmania infections and Helicobacter infections.
  • parasitic infections such as Leishmania infections and Helicobacter infections.
  • the scope of this embodiment is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells mediate responses by T helper 2 (Th2) cells, and thereby promote increased production of Immunoglobulin E (IgE) by B-cells.
  • Th2 T helper 2
  • the mesenchymal stem cells can be administered to a mammal, including human and non-human primates, as described herein.
  • the mesenchymal stem cells also can be administered systemically, as described herein.
  • the mesenchymal stem cells can be administered directly to an arthritic joint.
  • the mesenchymal stem cells are administered in an amount effective to treat an inflammatory response in an animal.
  • the mesenchymal stem cells can be administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 2 ⁇ 10 6 cells/kg to about 4 ⁇ 10 6 cells/kg. In still other embodiments, the mesenchymal stem cells are administered in an amount of about 3 ⁇ 10 6 cells/kg.
  • the exact dosage of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the inflammatory response being treated, and the extent and severity thereof.
  • the mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein.
  • a method of treating inflammation and/or repairing epithelial damage in an animal comprises administering to the animal mesenchymal stem cells in an amount effective to treat the inflammation and/or epithelial damage in the animal.
  • the mesenchymal stem cells cause a decrease in the secretion of the pro-inflammatory cytokines TNF- ⁇ and IFN- ⁇ by T-cells, and an increase in the secretion of the anti-inflammatory cytokines IL-10 and Interleukin-4 (IL-4) by T-cells. It is also believed that the mesenchymal stem cells cause a decrease in IFN- ⁇ secretion by natural killer (NK) cells.
  • NK natural killer
  • the inflammation and/or epithelial damage which can be treated in accordance with at least this aspect of the present technology includes, but is not limited to, inflammation and/or epithelial damage caused by a variety of diseases and disorders, including, but not limited to, autoimmune disease, rejection of transplanted organs, burns, cuts, lacerations, and ulcerations, including skin ulcerations and diabetic ulcerations.
  • diseases and disorders including, but not limited to, autoimmune disease, rejection of transplanted organs, burns, cuts, lacerations, and ulcerations, including skin ulcerations and diabetic ulcerations.
  • the mesenchymal stem cells are administered to an animal at least in order to repair epithelial damage resulting from autoimmune diseases, including, but not limited to, rheumatoid arthritis, Crohn's Disease, Type 1 diabetes, multiple sclerosis, scleroderma, Graves' Disease, lupus, inflammatory bowel disease, autoimmune gastritis (AIG), and autoimmune glomerular disease.
  • autoimmune diseases including, but not limited to, rheumatoid arthritis, Crohn's Disease, Type 1 diabetes, multiple sclerosis, scleroderma, Graves' Disease, lupus, inflammatory bowel disease, autoimmune gastritis (AIG), and autoimmune glomerular disease.
  • the mesenchymal stem cells also can repair epithelial damage resulting from graft-versus-host disease (GvHD).
  • This aspect of the present technology is applicable particularly to the repair of epithelial damage resulting from graft-versus-host disease, and more particularly, to the repair of epithelial damage resulting from severe graft-versus-host disease, including Grades III and IV GvHD affecting, for example, the skin and/or the gastrointestinal system.
  • Applicants have discovered, in particular, that mesenchymal stem cells, when administered to a patient suffering from severe graft-versus-host disease, and in particular, Grades III and IV GvHD, the administration of the mesenchymal stem cells resulted in repair of skin and/or ulcerated intestinal epithelial tissue in the subject and/or patient.
  • the mesenchymal stem cells are administered to an animal in order to repair epithelial damage to a transplanted organ or tissue including, but not limited to, kidney, heart, and lung, caused by rejection of the transplanted organ or tissue.
  • the mesenchymal stem cells are administered to an animal to repair epithelial damage caused by burns, cuts, lacerations, and ulcerations, including, but not limited to, skin ulcerations and diabetic ulcerations.
  • the mesenchymal stem cells can be administered to an animal such as a mammal, including human and non-human primates, as described herein.
  • the mesenchymal stem cells also can be administered systemically, as described herein.
  • the mesenchymal stem cells are administered in an amount effective to repair epithelial damage in an animal.
  • the mesenchymal stem cells can be administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 2 ⁇ 10 6 cells/kg to about 4 ⁇ 10 6 cells/kg. In still other embodiments, the mesenchymal stem cells are administered in an amount of about 3 ⁇ 10 6 cells/kg.
  • the exact dosage of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the type of epithelial damage being repaired, and the extent and severity thereof.
  • a method of treating cancer in an animal comprises administering to the animal mesenchymal stem cells in an amount effective to treat cancer in the animal.
  • the scope of this aspect of the present technology is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells interact with dendritic cells, which leads to IFN- ⁇ secretion, which in turn acts as a tumor suppressor.
  • Cancers which can be treated include, but are not limited to, hepatocellular carcinoma, cervical cancer, pancreatic cancer, prostate cancer, fibrosarcoma, medullablastoma, and astrocytoma. It is to be understood, however, that the scope of the present technology is not to be limited to any specific type of cancer.
  • animal includes a mammal, such as a human or a non-human primate.
  • the mesenchymal stem cells are administered to the animal in an amount effective to treat cancer in the animal.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg.
  • the exact amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the type of cancer being treated, and the extent and severity thereof.
  • the mesenchymal stem cells are administered in conjunction with an acceptable pharmaceutical carrier, and can be administered systemically, as described herein. Alternatively, the mesenchymal stem cells can be administered directly to the cancer being treated.
  • a method of treating an allergic disease or disorder in an animal comprises administering to the animal mesenchymal stem cells in an amount effective to treat the allergic disease or disorder in the animal.
  • mesenchymal stem cells when administered after an acute allergic response, provide for inhibition of mast cell activation and degranulation. Also, it is believed that the mesenchymal stem cells downregulate basophil activation and inhibit cytokines such as TNF- ⁇ , chemokines such as Interleukin-8 and monocyte chemoattractant protein, or MCP-1, lipid mediators such as leukotrienes, and inhibit main mediators such as histamine, heparin, chondroitin sulfates, and cathepsin.
  • cytokines such as TNF- ⁇
  • chemokines such as Interleukin-8 and monocyte chemoattractant protein, or MCP-1
  • lipid mediators such as leukotrienes
  • main mediators such as histamine, heparin, chondroitin sulfates, and cathepsin.
  • Allergic diseases or disorders which can be treated include, but are not limited to, asthma, allergic rhinitis, atopic dermatitis, and contact dermatitis. It is to be understood, however, that the scope of the present technology is not to be limited to any specific allergic disease or disorder.
  • the mesenchymal stem cells are administered to the animal in an amount effective to treat the allergic disease or disorder in the animal.
  • the animal can be a mammal.
  • the mammal can be a primate, including human and non-human primates.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg.
  • the exact dosage is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the allergic disease or disorder being treated, and the extent and severity thereof.
  • the mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein.
  • the mesenchymal stem cells can be administered systemically, such as by intravenous or intraarterial administration, for example.
  • a method of promoting wound healing in an animal comprises administering to the animal mesenchymal stem cells in an amount effective to promote wound healing in the animal.
  • the mesenchymal stem cells cause T reg cells and dendritic cells to release IL-10, which limits or controls inflammation in a wound, thereby promoting healing of a wound.
  • the mesenchymal stem cells can promote wound healing and fracture healing by inducing secretion factors by other cell types.
  • the mesenchymal stem cells can induce prostaglandin E 2 (PGE 2 )-mediated release of vascular endothelial growth factor (VEGF) by peripheral blood mononuclear cells (PBMCs), as well as PGE 2 -mediated release of growth hormone, insulin, insulin-like growth factor 1 (IGF-1) insulin-like growth factor binding protein-3 (IGFBP-3), and endothelin-1.
  • PGE 2 prostaglandin E 2
  • IGF-1 insulin-like growth factor 1
  • IGFBP-3 insulin-like growth factor binding protein-3
  • Wounds which can be healed include, but are not limited to, those resulting from cuts, lacerations, burns, and skin ulcerations.
  • the mesenchymal stem cells are administered to the animal in an amount effective to promote wound healing in the animal.
  • the animal can be a mammal, and the mammal can be a primate, including human and non-human primates.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg.
  • the exact amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, and the extent and severity of the wound being treated.
  • the mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein.
  • the mesenchymal stem cells can be administered systemically, as described herein.
  • the mesenchymal stem cells can be administered directly to a wound, such as in a fluid on a dressing or reservoir containing the mesenchymal stem cells.
  • a method of treating or preventing fibrosis or fibrotic disorder in an animal comprises administering to the animal mesenchymal stem cells in an amount effective to treat or prevent fibrosis or a fibrotic disorder in an animal.
  • the mesenchymal stem cells can be administered to the animal in order to treat or prevent any type of fibrosis or fibrotic disorder and in the animal, including, but not limited to, cirrhosis of the liver, fibrosis of the kidneys associated with end-stage renal disease, and lung disorders or diseases having fibrotic and can include in addition, inflammatory components, including, but not limited to, Acute Respiratory Distress Syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD). It is to be understood that the scope of the present technology is not to be limited to any specific type of fibrosis or fibrotic disorder.
  • ARDS Acute Respiratory Distress Syndrome
  • COPD Chronic Obstructive Pulmonary Disease
  • the mesenchymal stem cells are administered to the animal in an amount effective to treat or prevent fibrosis or a fibrotic disorder in the animal.
  • the animal can be a mammal, and the mammal can be a primate, including human and non-human primates.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg.
  • the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg.
  • the exact amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, and the extent and severity of the fibrosis or fibrotic disorder being treated or prevented.
  • the mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein.
  • the mesenchymal stem cells can be administered systemically, also as described herein.
  • angiogenesis i.e., the formation of new blood vessels from a pre-existing microvascular bed
  • a method of promoting angiogenesis in an organ or tissue of an animal comprises administering to the animal mesenchymal stem cells in an amount effective to promote angiogenesis in an organ or tissue of the animal.
  • angiogenesis can be used to treat coronary and peripheral artery insufficiency, and thus can be a noninvasive and curative approach to the treatment of coronary artery disease, ischemic heart disease, and peripheral artery disease.
  • Angiogenesis can play a role in the treatment of diseases and disorders in tissue and organs other than the heart, as well as in the development and/or maintenance of organs other than the heart.
  • Angiogenesis can provide a role in the treatment of internal and external wounds, as well as dermal ulcers.
  • Angiogenesis also plays a role in embryo implantation, and placental growth, as well as the development of the embryonic vasculature.
  • Angiogenesis also is essential for the coupling of cartilage resorption with bone formation, and is essential for correct growth plate morphogenesis.
  • angiogenesis is necessary for the successful engineering and maintenance of highly metabolic organs, such as the liver, where a dense vascular network is necessary to provide sufficient nutrient and gas transport.
  • the mesenchymal stem cells can be administered to the tissue or organ in need of angiogenesis by a variety of procedures.
  • the mesenchymal stem cells can be administered systemically, such as by intravenous, intraarterial, or intraperitoneal administration, or the mesenchymal stem cells can be administered directly to the tissue or organ in need of angiogenesis, such as by direct injection into the tissue or organ in need of angiogenesis.
  • the mesenchymal stem cells can be from a spectrum of sources including autologous, allogeneic, or xenogeneic.
  • PBMCs peripheral blood mononuclear cells
  • VEGF vascular endothelial growth factor
  • the animal is a mammal.
  • the mammal can be a primate, including human and non-human primates.
  • the mesenchymal stem cells in accordance with the present technology, can be employed in the treatment, alleviation, or prevention of any disease or disorder which can be alleviated, treated, or prevented through angiogenesis.
  • the mesenchymal stem cells can be administered to an animal to treat blocked arteries, including those in the extremities, such as arms, legs, hands, and feet, as well as the neck or in various organs.
  • the mesenchymal stem cells can be used to treat blocked arteries which supply the brain, thereby treating or preventing stroke.
  • the mesenchymal stem cells can be used to treat blood vessels in embryonic and post-natal corneas and can be used to provide glomerular structuring.
  • the mesenchymal stem cells can be employed in the treatment of wounds, both internal and external, as well as the treatment of dermal ulcers found in the feet, hands, legs or arms, including, but not limited to, dermal ulcers caused by diseases such as diabetes and sickle cell anemia.
  • the mesenchymal stem sells can be employed to promote embryo implantation and prevent miscarriage.
  • mesenchymal stem cells can be administered to an unborn animal, including humans, to promote the development of the vasculature in the unborn animal.
  • the mesenchymal stem cells can be administered to an animal, born or unborn, in order to promote cartilage resorption and bone formation, as well as promote correct growth plate morphogenesis.
  • the mesenchymal stem cells are administered in an amount effective in promoting angiogenesis in an animal.
  • the mesenchymal stem cells can be administered in an amount of from about 1 ⁇ 10 5 cells/kg to about 1 ⁇ 10 7 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1 ⁇ 10 6 cells/kg to about 5 ⁇ 10 6 cells/kg.
  • the amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the disease or disorder to be treated, alleviated, or prevented, and the extent and severity thereof.
  • the mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier.
  • the mesenchymal stem cells can be administered as a cell suspension in a pharmaceutically acceptable liquid medium for injection. Injection can be local, such as by administration directly into the tissue or organ in need of angiogenesis, or systemic, such as intravenously or intraarterially.
  • the mesenchymal stem cells can be genetically engineered with one or more polynucleotides encoding a therapeutic agent.
  • the polynucleotides can be delivered to the mesenchymal stem cells via an appropriate expression vehicle.
  • Expression vehicles which can be employed to genetically engineer the mesenchymal stem cells include, but are not limited to, retroviral vectors, adenoviral vectors, and adeno-associated virus vectors.
  • polynucleotide encoding a therapeutic agent is dependent upon various factors, including the disease or disorder being treated, and the extent and severity thereof.
  • Polynucleotides encoding therapeutic agents, and appropriate expression vehicles are described further in U.S. Pat. No. 6,355,239, the contents of which are hereby incorporated by reference in its entirety.
  • mesenchymal stem cells when employed in the above-mentioned therapies and treatments, can be employed in combination with other therapeutic agents known to those skilled in the art, including, but not limited to, growth factors, cytokines, drugs such as anti-inflammatory drugs, and cells other than mesenchymal stem cells, such as dendritic cells, and can be administered with soluble carriers for cells such as hyaluronic acid, or in combination with solid matrices, such collagen, gelatin, or other biocompatible polymers, as appropriate.
  • MSCs Materials and Methods Culture of human MSCs.
  • Human MSCs were cultured as described by Pittenger et al., Science , Vol. 284, pg. 143 (1999). Briefly, marrow samples were collected from the iliac crest of anonymous donors following informed consent by Poietics Technologies, Div of Cambrex Biosciences. MSCs were cultured in complete Dulbecco's Modified Eagle's Medium-Low Glucose (Life Technologies, Carlsbad, Calif.) containing 1% antibiotic-antimyotic solution (Invitrogen, Carlsbad, Calif.) and 10% fetal bovine serum (FBS, JRH BioSciences, Lenexa, Kans.).
  • MSCs grew as an adherent monolayer and were detached with trypsin/EDTA (0.05% trypsin at 37° C. for 3 minutes). All MSCs used were previously characterized for multilineage potential and retained the capacity to differentiate into mesenchymal lineages (chondrocytic, adipogenic, and osteogenic) (Pittenger, et al., Science , Vol. 284, pg. 143 (1999)).
  • PBMCs Peripheral blood mononuclear cells
  • DCs dendritic cells
  • CD1c+ monocytic lineage
  • CD1c expressing B cells were magnetically depleted of CD19+ cells using magnetic beads, followed by labeling the B-cell depleted fraction with biotin-labeled CD1c (BDCA1+) and anti-biotin antibodies and separating them from the unlabeled cell fraction utilizing magnetic columns according to the manufacturer's instructions (Miltenyi Biotech, Auburn, Calif.).
  • Precursors of DCs of plasmacytoid lineage were isolated from PBMCs by immuno-magnetic sorting of positively labeled antibody coated cells (BDCA2+) (Miltenyi Biotech, Auburn, Calif.).
  • MSC-DC culture In most experiments, human MSCs and DCs were cultured in equal numbers for various time periods and cell culture supernatant collected and stored at ⁇ 80° C. until further evaluation. In selected experiments, MSCs were cultured with mature DC1 or DC2 cells (1:1 MSC:DC ratio) for 3 days, and then the combined cultures (MSCs and DCs) were irradiated to prevent any proliferation. Next, antibody purified, na ⁇ ve, allogeneic T cells (CD4+,CD45RA+) were added to the irradiated MSCs/DCs and cultured for an additional 6 days.
  • the non-adherent cell fraction (purified T cells) was then collected from the cultures, washed twice and re-stimulated with PHA for another 24 hours, following which cell culture supernatants were harvested and analyzed for secreted IFN- ⁇ and IL-4 by ELISA.
  • NK cells Isolation of NK cells.
  • Purified populations of NK cells were obtained by depleting non-NK cells that are magnetically labeled with a cocktail of biotin-conjugated monoclonal antibodies (anti-CD3, -CD14, -CD19, -CD36 and anti-IgE antibodies) as a primary reagent and anti-biotin monoclonal antibodies conjugated to Microbeads as secondary labeling reagent.
  • the magnetically labeled non-NK cells were retained in MACS (Miltenyi Biotech, Auburn, Calif.) columns in a magnetic field, while NK cells passed through and were collected.
  • T Reg cell population was isolated using a 2-step isolation procedure.
  • First non-CD4 + T cells were indirectly magnetically labeled with a cocktail of biotin labeled antibodies and anti-biotin microbeads. The labeled cells were then depleted by separation over a MACS column (Miltenyi Biotech, Auburn, Calif.).
  • CD4 + CD25 + cells were directly labeled with CD25 microbeads and isolated by positive selection from the pre-enriched CD4 + . T cell fraction.
  • the magnetically labeled CD4 + CD25 + T cells were retained on the column and eluted after removal of the column from the magnetic field.
  • CD4+CD25+T reg cell populations were isolated from PBMC or MSC+PBMC (MSC to PBMC ratio 1:10) cultures (cultured without any further stimulation for 3 days) using a 2-step magnetic isolation procedure. These cells were irradiated to block any further proliferation and used as stimulators in a mixed lymphocyte reaction (MLR), where responders were allogeneic PBMCs (stimulator to responder ratio 1:100) in the presence of PHA (2.5 ⁇ g/ml). The culture was carried out for 48 hours, following which 3 H thymidine was added. Incorporated radioactivity was counted after 24 hours.
  • MLR mixed lymphocyte reaction
  • PBMCs were cultured in the absence or presence of MSCs (MSC to PBMC ratio 1:10), following which the non-adherent fraction was harvested and immunostained with FITC-labeled glucocorticoid-induced TNF receptor, or GITR, and PE-labeled CD4.
  • Th1/Th2 cells Peripheral blood mononuclear cells (PBMCs) were plated at 2 ⁇ 10 6 cells/ml for 45 min. at 37° C. in order to remove monocytes. Non-adherent fraction was incubated in the presence of plate-bound anti-CD3 (5 ⁇ g/ml) and anti-CD28 (1 ⁇ g/ml) antibodies under Th1 (IL-2 (4 ng/ml) IL-12 (5 ng/ml)+anti-IL-4 (1 ⁇ g/ml)) or Th2 (IL-2 (4 ng/ml)+IL-4 (4 ng/ml)+anti-IFN- ⁇ (1 ⁇ g/ml)) conditions for 3 days in the presence or absence of MSCs.
  • IL-2 (4 ng/ml) IL-12
  • Th2 IL-2 (4 ng/ml)+IL-4 (4 ng/ml)+anti-IFN- ⁇ (1 ⁇ g/ml)
  • the cells were washed and then re-stimulated with PHA (2.5 ⁇ g/ml) for another 24 or 48 hours, following which levels of IFN- ⁇ and IL-4 were measured in culture supernatants by ELISA (R&D Systems, Minneapolis, Minn.).
  • VEGF vascular endothelial growth factor
  • PGE 2 vascular endothelial growth factor
  • pro-MMP-1 matrix metalloproteinase 1
  • PBMCs Proliferation of PBMCs.
  • Purified PBMCs were prepared by centrifuging leukopack (Cambrex, Walkersville, Md.) on Ficoll-Hypaque (Lymphoprep, Oslo, Norway). Separated cells were cultured (in triplicates) in the presence or absence of MSCs (plated 3-4 hours prior to PBMC addition to allow them to settle) for 48 hours in presence of the mitogen PHA (Sigma Chemicals, St. Louis, Mo.).
  • PBMCs were resuspended in medium containing PGE 2 inhibitors Indomethacin (Sigma Chemicals, St. Louis, Mo.) or NS-938 (Cayman Chemicals, Ann Arbor, Mich.).
  • Quantitative RT-PCR Total RNA from cell pellets were prepared using a commercially available kit (Qiagen, Valencia, Calif.) and according to the manufacturer's instructions. Contaminating genomic DNA was removed using the DNA-free kit (Ambion, Austin, Tex.). Quantitative RT-PCR was performed on a MJ Research Opticon detection system (South San Francisco, Calif.) using QuantiTect SYBR Green RT-PCR kit (Qiagen, Valencia, Calif.) with primers at concentration of 0.5 ⁇ M. Relative changes in expression levels in cells cultured under different conditions were calculated by the difference in Ct values (crossing point) using ⁇ -actin as internal control.
  • COX-1 and COX-2 specific primers were: COX-1: 5′-CCG GAT GCC AGT CAG GAT GAT G-3′ (forward) (SEQ ID NO:1), 5′-CTA GAC AGC CAG ATG CTG ACA G-3′ (reverse) (SEQ ID NO:2); COX-2: 5′-ATC TAC CCT CCT CAA GTC CC-3′ (forward) (SEQ ID NO:3), 5′-TAC CAG AAG GGC AGG ATA CAG-3′ (reverse) (SEQ ID NO:4).
  • DC1 and DC2 precursor dendritic cells were isolated by immuno-magnetic sorting of BDCA1 + and BDCA2 + cells respectively and matured by incubation with GM-CSF and IL-4 (1 ⁇ 10 3 IU/ml and 1 ⁇ 10 3 IU/ml, respectively) for DC1 cells, or IL-3 (10 ng/ml) for DC2 cells.
  • GM-CSF and IL-4 (1 ⁇ 10 3 IU/ml and 1 ⁇ 10 3 IU/ml, respectively
  • IL-3 10 ng/ml
  • DC1 cells In the presence of the inflammatory agent bacterial lipopolysaccharide (LPS, 1 ng/ml), DC1 cells produced moderate levels of TNF- ⁇ but when MSCs were present (ratios examined 1:1 and 1:10), there was >50% reduction in TNF- ⁇ secretion ( FIG. 1B ).
  • DC2 cells produced IL-10 in the presence of LPS and its levels were increased greater than 2-fold upon MSC:DC2 co-culture (1:1) ( FIG. 1B ). Therefore, the MSCs modified the cytokine profile of activated DCs in culture towards a more tolerogenic phenotype.
  • activated DCs when cultured with MSCs, were able to reduce IFN- ⁇ and increase IL-4 levels secreted by na ⁇ ve CD4 + T cells ( FIG. 1C ) suggesting a MSC-mediated shift from pro-inflammatory to anti-inflammatory T cell phenotype.
  • T Reg T-regulatory cells
  • the CD4 + CD25 + T Reg cell population generated in presence of MSCs expressed increased levels of gluocorticoid-induced TNF receptor (GITR), a cell surface receptor expressed on T Reg cell populations, and was suppressive in nature as it suppressed allogeneic T cell proliferation (FIG. 3 A,B).
  • GITR gluocorticoid-induced TNF receptor
  • T Reg cell populations a cell surface receptor expressed on T Reg cell populations, and was suppressive in nature as it suppressed allogeneic T cell proliferation
  • MSCs were investigated as to their direct ability to affect T cell differentiation. Using antibody selected purified T cells (CD4 + Th cells), IFN- ⁇ producing Th1 and IL-4 producing T H 2 cells were generated in presence or absence of MSCs. When MSCs were present during differentiation, there was reduced IFN- ⁇ secretion by Th1 cells and increased IL-4 secretion by Th2 cells ( FIG. 2B ).
  • MSCs modify T-cell functions by soluble factor(s) (LeBlanc, et al., Exp. Hematol ., Vol. 31, pg. 890 (2003); Tse, et al., Transplantation , Vol. 75, pg. 389 (2003). It was observed that the MSCs secreted several factors, including IL-6, prostaglandin E 2 , VEGF and proMMP-lconstitutively, and the levels of each increased upon culture with PBMCs ( FIG. 5 ).
  • MSCs were cultured with activated dendritic cells (DC1) or Th1 cells in the presence of PGE 2 inhibitors NS-398 or indomethacin.
  • DC1s dendritic cells
  • Th1 cells Th1 cells
  • PGE 2 inhibitors NS-398 or indomethacin increased TNF- ⁇ secretion by DC1s, and IFN- ⁇ secretion from Th1 cells ( FIG. 4C ), respectively, suggesting that MSC effects on immune cell types can be mediated by secreted PGE 2 .
  • Recent studies have shown that MSCs inhibit T-cell proliferation induced by various stimuli (DeNicola, et al., Blood , Vol. 99, pg.
  • MSCs inhibit mitogen-induced T cell proliferation in a dose-dependent manner ( FIG. 6 ) and when PGE 2 inhibitors NS-398 (5 ⁇ M) or indomethacin (4 ⁇ M) were present, there was a >70% increase in ( 3 H) thymidine incorporation by PHA-treated PBMCs in MSC containing cultures as compared to controls without inhibitors ( FIG. 4D ).
  • FIG. 7 a model of MSC interaction with other immune cell types ( FIG. 7 ) is proposed.
  • MSCs can interact with them directly and inhibit the pro-inflammatory IFN- ⁇ production (pathway 1) and promote regulatory T cell phenotype (pathway 3) and anti-inflammatory T H 2 cells (pathway 5).
  • MSCs can alter the outcome of the T cell immune response through DCs by secreting PGE 2 , inhibiting pro-inflammatory DC1 cells (pathway 2) and promoting anti-inflammatory DC2 cells (pathway 4) or regulatory DCs (pathway 3).
  • a shift towards T H 2 immunity suggests a change in B cell activity towards increased generation of IgE/IgG1 subtype antibodies (pathway 7).
  • MSCs by their ability to inhibit IFN- ⁇ secretion from NK cells likely modify NK cell function (pathway 6).
  • This model of MSC:Immune cell interactions is consistent with the experimentation performed in several other laboratories (LeBlanc, et al., Exp. Hematol ., Vol. 31, pg. 890 (2003); Tse, et al., Transplantation , Vol. 75, pg. 389 (2003); DiNicola, et al., Blood , Vol. 99, pg. 3838 (2002)). Further examination of the proposed mechanisms is underway and animal studies are now necessary to examine the in vivo effects of MSC administration.
  • GVHD gastrointestinal graft-versus-host disease
  • the patient was given an intravenous infusion of allogeneic mesenchymal stem cells in 50 ml of Plasma Lyte A (Baxter) in an amount of 3 ⁇ 10 6 cells per kilogram of body weight.
  • Plasma Lyte A Plasma Lyte A
  • the patient was evaluated at two weeks post-infusion. At two weeks post-infusion, an endoscopic view of the patient's colon showed that the areas of inflammation and ulceration visible prior to treatment were resolved. In addition, a biopsy of the patient's colon showed significant regeneration of intestinal crypts.
  • the administration of the mesenchymal stem cells to the patient resulted in a significant reduction in the inflammatory component of gastrointestinal graft-versus-host disease, and resulted in the regeneration of new functional intestinal tissue.

Abstract

Disclosed are cell preparations comprising multipotent adult stem cells and methods for using multipotent adult stem cells to treat autoimmune diseases, treat allergic responses, treat cancer, treat inflammatory diseases, treat fibrotic disorders, reduce inflammation and/or fibrosis, promote would healing, repair epithelial damage, and/or promote angiogenesis.

Description

    RELATED APPLICATIONS
  • This application is a continuation of application Ser. No. 11/541,853, filed Oct. 2, 2006, which is a continuation-in-part of application Ser. No. 11/080,298, filed Mar. 15, 2005, which claims priority based on provisional application Ser. No. 60/555,118, filed Mar. 22, 2004; the contents of each application are hereby incorporated by reference in their entireties.
  • FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • The present technology was made with Government support under Contract No. N66001-02-C-8068 awarded by the Department of the Navy. The Government has certain rights in this technology
  • BACKGROUND OF THE INVENTION
  • Mesenchymal stem cells (MSCs), which are present in adult bone marrow, are a kind of multipotent stem cell that can differentiate readily into lineages including osteoblasts, myocytes, chondrocytes, and adipocytes. (Pittenger, et al., Science, Vol. 284, pg. 143 (1999); Haynesworth, et al., Bone, Vol. 13, pg. 69 (1992); Prockop, Science, Vol. 276, pg. 71 (1997)). In vitro studies have demonstrated the capability of MSCs to differentiate into muscle (Wakitani, et al., Muscle Nerve, Vol. 18, pg. 1417 (1995)), neuronal-like precursors (Woodbury, et al., J. Neurosci. Res., Vol. 69, pg. 908 (2002); Sanchez-Ramos, et al., Exp. Neurol., Vol. 171, pg. 109 (2001)), cardiomyocytes (Toma, et al., Circulation, Vol. 105, pg. 93 (2002); Fakuda, Artif. Organs, Vol. 25, pg. 187 (2001)) and possibly other cell types. In addition, MSCs have been shown to provide effective feeder layers for expansion of hematopoietic and embryonic stem cells (Eaves, et al., Ann. N.Y. Acad. Sci., Vol. 938, pg. 63 (2001); Wagers, et al., Gene Therapy, Vol. 9, pg. 606 (2002)). Recent studies with a variety of animal models have shown that MSCs can be useful in the repair and/or regeneration of damaged bone, cartilage, meniscus or myocardial tissues (DeKok, et al., Clin. Oral Implants Res., Vol. 14, pg. 481 (2003)); Wu, et al., Transplantation, Vol. 75, pg. 679 (2003); Noel, et al., Curr. Opin. Investig. Drugs, Vol. 3, pg. 1000 (2002); Ballas, et al., J. Cell. Biochem. Suppl., Vol. 38, pg. 20 (2002); Mackenzie, et al., Blood Cells Mol. Dis., Vol. 27 (2002)). Several investigators have used MSCs with encouraging results for transplantation in animal disease models including osteogenesis imperfecta (Pereira, et al., Proc. Nat. Acad. Sci., Vol. 95, pg. 1142 (1998)), parkinsonism (Schwartz, et al., Hum. Gene Ther., Vol. 10, pg. 2539 (1999)), spinal cord injury (Chopp, et al., Neuroreport, Vol. 11, pg. 3001 (2000); Wu, et al., J. Neurosci. Res., Vol. 72, pg. 393 (2003)) and cardiac disorders (Tomita, et al., Circulation, Vol. 100, pg. 247 (1999). Shake, et al., Ann. Thorac. Surg., Vol. 73, pg. 1919 (2002)). Importantly, promising results also have been reported in clinical trials for osteogenesis imperfecta (Horwitz, et al., Blood, Vol. 97, pg. 1227 (2001); Horowitz, et al. Proc. Nat. Acad. Sci., Vol. 99, pg. 8932 (2002)) and enhanced engraftment of heterologous bone marrow transplants (Frassoni, et al., Int. Society for Cell Therapy, SA006 (abstract) (2002); Koc, et al., J. Clin. Oncol., Vol. 18, pg. 307 (2000)).
  • MSCs express major histocompatibility complex (MHC) class I antigen on their surface, but do not express MHC class II (Le Blanc, et al., Exp. Hematol., Vol. 31, pg. 890 (2003); Potian, et al., J. Immunol., Vol. 171, pg. 3426 (2003)) and do not express B7 or CD40 co-stimulatory molecules (Majumdar, et al., J. Biomed. Sci., Vol. 10, pg. 228 (2003)), suggesting that these cells have a low-immunogenic phenotype (Tse, et al., Transplantation, Vol. 75, pg. 389 (2003)). MSCs also inhibit T-cell proliferative responses in an MHC-independent manner (Bartholomew, et al., Exp. Hematol., Vol. 30, pg. 42 (2002); Devine, et al., Cancer J., Vol. 7, pg. 576 (2001); DiNicola, et al., Blood, Vol. 99, pg. 3838 (2002)). These immunological properties of MSCs can enhance their transplant engraftment and limit the ability of the recipient's immune system to recognize and reject allogeneic cells following transplantation. The production of factors by MSCs, that modulate the immune response and support hematopoiesis together with their ability to differentiate into appropriate cell types under local stimuli make them desirable stem cells for cellular transplantation studies (Majumdar, et al., Hematother. Stem Cell Res., Vol. 9, pg. 841 (2000); Haynesworth, et al., J. Cell. Physiol., Vol. 166, pg. 585 (1996).
  • BRIEF SUMMARY OF THE INVENTION
  • The present technology generally relates to multipotent adult stem cells, such as adult bone marrow-derived stem cells. More particularly, the present technology relates to new and heretofore unappreciated uses for multipotent adult stem cells, such as mesenchymal stem cells, including, but not limited to promoting angiogenesis in various tissues and organs, treating autoimmune diseases, treating allergic responses, treating cancer, treating inflammatory diseases and disorders, promoting wound healing, treating inflammation, and repairing epithelial damage.
  • In accordance with the present technology, multipotent adult stem cells can be used, for example, to treat an autoimmune disease, treat an inflammatory response, treat an allergic disease, treat a pulmonary disease having fibrotic and/or inflammatory components, repair epithelial damage, and promote wound healing in subjects, including a human subject. Autoimmune diseases that can be treated with multipotent adult stem cells include, for example, Type 1 Diabetes, inflammatory bowel disease, Crohn's disease, and uveitis. Pulmonary diseases that can be treated with multipotent adult stem cells include, but are not limited to, Acute Respiratory Distress Syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD), and asthma. Inflammatory responses, including those associated with autoimmune diseases or pulmonary diseases, can be reduced with multipotent adult stem cells. An inflammatory response can be reduced by, for example, reducing the production or expression of pro-inflammatory mediators, increasing the production or expression of anti-inflammatory mediators, or a combination thereof.
  • Multipotent adult stem cells of the present technology have the capacity to differentiate into at least one cell type of each of the mesodermal, ectodermal, and endodermal lineages. For example, the cells can be induced to differentiate into cells of at least osteoblast, chondrocyte, adipocyte, fibroblast, marrow stroma, skeletal muscle, smooth muscle, cardiac muscle, endothelial, epithelial, hematopoietic, glial, neuronal or oligodendrocyte cell types, among others. Multipotent adult stem cells of the present technology are capable of differentiating into at least one cell type of at least one of the endodermal, ectodermal, or mesodermal embryonic lineages.
  • Adult bone marrow is an accessible and renewable source of adult multipotent stem cells that can be greatly expanded in culture. For example, bone marrow-derived mesenchymal stem cells (MSCs) are multipotent cells that have been identified and cultured from various avian and mammalian species.
  • The present technology provides one or more cell preparations comprising adult bone marrow-derived stem cells in one or more doses effective to treat an inflammatory response in a subject. The subject can be a human having, for example, inflammatory bowel disease. The stem cells can be capable of differentiating into at least one cell type of each of the endodermal, ectodermal, and mesodermal embryonic lineages. The effective dose of the cell preparation can contain a sufficient number of stem cells to provide about 1×105 to about 1×107 cells per kilogram of the subject. In certain embodiments, the subject can be a mammal. The mammal can be, for example, a primate, including a human and a non-human primate. Administration of the cell preparation can, for example, elevate interferon-beta levels in the subject.
  • The present technology also provides a method of treating inflammatory bowel disease in a subject, comprising administering to the subject allogeneic multipotent adult stem cells in an amount effective to treat inflammatory bowel disease. In certain embodiments, the subject can be an animal. The animal can be, for example, a primate, including a human and a non-human primate. The multipotent adult stem cells can be administered systemically, such as intravenously, intraarterially, or intraperitoneally. The multipotent adult stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, such as a pharmaceutically acceptable liquid medium. The multipotent adult stem cells can be administered as a suspension of cells. The amount effective to treat inflammatory bowel disease can be about 1×105 to about 1×107 cells per kilogram of the subject.
  • The present technology further provides a method of treating a gastrointestinal autoimmune disease in a subject, comprising administering to the subject allogeneic multipotent adult stem cells in an amount effective to repair or regenerate intestinal tissue. The autoimmune disease can be selected from the group consisting of Crohn's disease, inflammatory bowel disease, and autoimmune gastritis. In certain embodiments, the subject can be an animal. The animal can be, for example, a primate, including a human and a non-human primate. The multipotent adult stem cells can be administered systemically, such as intravenously, intraarterially, or intraperitoneally. The multipotent adult stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, such as a pharmaceutically acceptable liquid medium. The multipotent adult stem cells can be administered as a suspension of cells. The amount effective to repair or regenerate intestinal tissue can be about 1×105 to about 1×107 cells per kilogram of the subject.
  • The multipotent adult stem cells of the present technology can be positive for one or more cell surface markers selected from, for example, integrin al (CD49a); integrin α2 (CD49b); integrin α3 (CD49c); integrin α5 (CD49e); integrin αV (CD51); integrin β1 (CD29); integrin β3 (CD61); integrin β4 (CD104); IL-1R (CD121a); IL-3Rα (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFNγR (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGFβ1R; TGFβIIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD166); hyaluronate (CD44); endoglin (CD105); Thy-1 (CD90); or CD9. The stem cells can also be positive for each of CD49b, CD49e, and CD140a. Further, the stem cells can be negative for one or more cell surface markers such as integrin α4 (CD49d); integrin αL (CD11a); integrin Cβ2 (CD18); CD4; CD14; CD34; CD45; IL-2R (CD25); EGFR-3; Fas ligand; ICAM-3 (CD50); E-Selectin (CD62E); P-Selectin (CD62P); vW Factor; cadherin 5; or Lewis x (CD15). In addition, the multipotent adult stem cells of the present technology can be adult bone marrow-derived stem cells such as, for example, mesenchymal stem cells.
  • The present technology also provides a method of repairing and/or regenerating intestinal tissue in a subject, comprising treating the subject with allogeneic mesenchymal stem cells. In certain embodiments, the subject can be an animal. The animal can be, for example, a primate, including a human and a non-human primate. The stem cells can be from an adult. The mesenchymal stem cells can be administered to the subject as part of, for example, a pharmaceutical formulation having a pharmaceutically acceptable carrier, such as a liquid injectable carrier, a liquid topical carrier, a gel injectable carrier, a gel topical carrier, a solid matrix, and combinations thereof. The mesenchymal stem cell pharmaceutical formulation can be administered via intravenous, intraarterial, or intraperitoneal injection. Mesenchymal stem cells can be administered in an amount effective to repair or regenerate intestinal tissue in a subject. The amount effective to repair or regenerate intestinal tissue can be about 1×105 to about 1×107 cells per kilogram of the subject.
  • Applicants presently have examined the interactions of multipotent adult stem cells with isolated immune cell populations, including dendritic cells (DC1 and DC2), effector T-cells (Th1 and Th2), and NK cells. Based on such interactions, Applicants discovered that multipotent adult stem cells regulate the production of various factors that affect several steps in the inflammatory and/or immune response process. Thus, the multipotent adult stem cells of the present technology can be employed in the treatment of disease conditions and disorders involving the immune system, or diseases, conditions, or disorders involving inflammation, epithelial damage, or allergic responses. Such diseases, conditions, and disorders include, but are not limited to, autoimmune diseases, allergies, arthritis, inflamed wounds, alopecia araeta (baldness), periodontal diseases including gingivitis and periodontitis, and other diseases, conditions or disorders involving an immune response.
  • In addition, it is believed that multipotent adult stem cells, including mesenchymal stem cells, express and secrete vascular endothelial growth factor, or VEGF, which promotes angiogenesis by stimulating the formation of new blood vessels. Mesenchymal stem cells also stimulate peripheral blood mononuclear cells (PBMCs) to produce VEGF.
  • Furthermore, it is believed that multipotent adult stem cells stimulate dendritic cells (DCs) to produce Interferon-Beta (IFN-β), which promotes tumor suppression and immunity against viral infection.
  • BRIEF DESCRIPTION OF SEVERAL VIEWS OF THE DRAWINGS
  • The present technology now will be described with respect to the drawings.
  • FIG. 1. MSCs modulate dendritic cell functions. (A) Flow cytometric analysis of mature monocytic DC1 cells using antibodies against HLA-DR and CD11c and of plasmacytoid DC2 cells using antibodies against HLA-DR and CD123 (IL-3 receptor). (---): isotype control; (-): FITC/PE conjugated antibodies. (B) MSCs inhibit TNF-α secretion (primary y-axis) and increase IL-10 secretion (secondary y-axis) from activated DC1 and DC2 respectively. (C) MSCs cultured with mature DC1 cells inhibit IFN-γ secretion (primary y-axis) by T cells and increase IL-4 levels (secondary y-axis) as compared to MSC or DC alone. The decreased production of pro-inflammatory IFN-γ and increased production of anti-inflammatory IL-4 in the presence of MSCs indicated a shift in the T cell population towards an anti-inflammatory phenotype.
  • FIG. 2. MSCs inhibit pro-inflammatory effector T cell function. (A) Flow cytometric analysis of Treg cell numbers (in %) by staining PBMCs or the non-adherent fraction in MSC+PBMC culture (MSC+PBMC) with FITC-conjugated CD4 (x-axis) and PE conjugated CD25 (y-axis) antibodies. Gates were set based on isotype control antibodies as background. Graphs are representative of 5 independent experiments. (B) Th1 cells generated in presence of MSCs secreted reduced levels of IFN-γ (primary Y-axis) and Th2 cells generated in presence of MSCs secreted increased amounts of IL-4 (secondary y-axis) in cell culture supernatants. (C) MSCs inhibit IFN-γ secretion from purified NK cells cultured for 0, 24, or 48 hours in a 24-well plate. Data shown are mean±SD cytokine secretion in one experiment and are representative of 3 independent experiments.
  • FIG. 3. MSCs lead to increased numbers of Treg cell population and increased GITR expression. (A) A CD44+CD25+Treg cell population from PBMC or MSC+PBMC (MSC to PBMC ratio 1:10) cultures (cultured without any further stimulation for 3 days) was isolated using a 2-step magnetic isolation procedure. These cells were irradiated (to block any further proliferation) and used as stimulators in a mixed lymphocyte reaction (MLR), where responders were allogeneic PBMCs (stimulator to responder ratio 1:100) in the presence of phytohemagglutinin (PHA) (2.5 mg/ml). The cells were cultured for 48 hours; 3H thymidine was added; and incorporated radioactivity was counted after 24 hours. The results showed that the Treg population, generated in the presence of MSCs (lane 3) was similar functionally to the Treg cells generated in the absence of MSCs (lane 2). (B) PBMCs were cultured for 3 days in the absence (top plot) or presence (bottom plot) of MSCs (MSC to PBMC ratio 1:10), following which the non-adherent fraction was harvested and immunostained with FITC-labeled GITR and PE-labeled CD4. Results show a greater than twofold increase in GITR expression in cells cultured in the presence of MSCs.
  • FIG. 4. MSCs produce PGE2 and blocking PGE2 reverses MSC-mediated immuno-modulatory effects. (A) PGE2 secretion (mean±SD) in culture supernatants obtained from MSCs cultured in the presence or absence of PGE2 blockers NS-398 or indomethacin (Indometh.) at various concentrations. Inhibitor concentrations are in μM and data presented are values obtained after 24 hour culture (B) COX-1 and COX-2 expression in MSCs and PBMCs using real-time RT-PCR. MSCs expressed significantly higher levels of COX-2 as compared to PBMCs, and when MSCs were cultured in presence of PBMCs, there was a >3-fold increase in COX-2 expression in MSCs. Representative data from 1 of 3 independent experiments are shown. The MSC+PBMC cultures were setup in a trans-well chamber plate where MSCs were plated onto the bottom chamber and PBMCs onto the top chamber. (C) Presence of PGE2 blockers indomethacin (Ind.) or NS-398 increases TNF-α secretion from activated DCs (open bars) and IFN-γ secretion from Th1 cells (hatched bars) as compared to controls. Data were calculated as % change from cultures generated in absence of MSCs and PGE2 inhibitors (D) Presence of PGE2 blockers indomethacin (Indo) and NS-398 during MSC
    Figure US20100172885A1-20100708-P00001
    PBMC co-culture (1:10) reverses MSC-mediated anti-proliferative effects on PHA-treated PBMCs. Data shown are from one experiment and are representative of 3 independent experiments.
  • FIG. 5. Constituitive MSC cytokine secretion is elevated in the presence of allogeneic PBMCs. Using previously characterized human MSCs, the levels of the cytokines IL-6 and VEGF, lipid mediator PGE2, and matrix metalloproteinase 1 (pro MMP-1) in culture supernatant of MSCs cultured for 24 hours in the presence (hatched bars) or absence (open bars) of PBMCs (MSC to PBMC ratio 1:10) were analyzed. The MSCs produced IL-6, VEGF, and PGE2 constituitively, and the levels of these factors increased upon co-culture with PBMCs, thereby suggesting that MSCs can play a role in modulating immune functions in an inflammatory setting.
  • FIG. 6. MSCs inhibit mitogen-induced T-cell proliferation in a dose-dependent manner. Increasing numbers of allogeneic PBMCs were incubated with constant numbers of MSCs (2,000 cells/well) plated on a 96-well plate in the presence or absence of PHA (2.5 mg/ml) for 72 hours, and 3H thymidine incorporation determined (in counts per minute, or cpm). There was a dose-dependent inhibition of the proliferation of PHA-treated PBMCs in the presence of MSCs. Representative results from 1 of 3 independent experiments are shown. Similar results were reported by LeBlanc, et al., Scand J. Immunol., Vol. 57, pg. 11 (2003).
  • FIG. 7. Schematic diagram of proposed MSC mechanism of action. MSCs mediate their immuno-modulatory effects by affecting cells from both the innate (DC-pathways 2-4; and NK-pathway 6) and adaptive (T-pathways 1 and 5 and B-pathway 7) immune systems. In response to an invading pathogen, immature DCs migrate to the site of potential entry, mature and acquire an ability to prime naïve T cells (by means of antigen specific and co-stimulatory signals) to become protective effector T cells (cell-mediated Th1 or humoral Th2 immunity). During MSC-DC interaction, MSCs, by, for example, means of direct cell-cell contact or via secreted factor, can alter the outcome of immune response by limiting the ability of DCs to mount a cell-mediated response (pathway 2) or by promoting the ability to mount a humoral response (pathway 4). Also, when mature effector T cells are present, MSCs can interact with them to skew the balance of Th1 (pathway 1) responses towards TH2 responses (pathway 5), and probably towards an increased IgE producing B cell activity (pathway 7), desirable outcomes for suppression of GvHD and autoimmune disease symptoms. MSCs in their ability to result in an increased generation of Treg population (pathway 3) can result in a tolerant phenotype and can aid a recipient host by dampening bystander inflammation in their local micro-environment. Dashed line (----) represents proposed mechanism.
  • DETAILED DESCRIPTION OF THE INVENTION
  • In accordance with an aspect of the present technology, there is provided a method of treating a disease selected from the group consisting of autoimmune diseases and graft-versus-host disease in an animal, including, for example, a human. The method comprises at least the step of administering to the animal mesenchymal stem cells in an amount effective to treat the disease in the animal.
  • Although the scope of this aspect of the present technology is not to be limited to any theoretical reasoning, it is believed that at least one mechanism by which the mesenchymal stem cells suppress autoimmune disease and graft-versus-host disease is by causing the release of Interleukin-10 (IL-10) from regulatory T-cells (Treg cells) and/or dendritic cells (DC).
  • Autoimmune diseases which can be treated in accordance with the present technology include, but are not limited to, multiple sclerosis, Type 1 diabetes, rheumatoid arthritis, uveitis, autoimmune thyroid disease, inflammatory bowel disease, scleroderma, Graves' Disease, lupus, Crohn's disease, autoimmune lymphoproliferative disease (ALPS), demyelinating disease, autoimmune encephalomyelitis, autoimmune gastritis (AIG), and autoimmune glomerular diseases. Also, as noted hereinabove, graft-versus-host disease can be treated. It is to be understood, however, that the scope of the present technology is not to be limited to the treatment of the specific diseases mentioned herein.
  • In certain embodiments, the mesenchymal stem cells are administered to a mammal. The mammal can be a primate, including human and non-human primates.
  • In general, the multipotent adult stem cell therapy is based, for example, on the following sequence: harvest of stem cell-containing tissue such as, for example, bone marrow; isolation and/or expansion of stem cells; and administration of the stem cells, with or without biochemical or genetic manipulation, to the animal.
  • The mesenchymal stem cells that are administered can be, for example, a homogeneous composition or a mixed cell population enriched in mesenchymal stem cells. The mesenchymal stem cell compositions can be obtained, for example, by culturing adherent bone marrow or periosteal cells. Adult bone marrow-derived stem cells, such as bone marrow-derived mesenchymal stem cells, can be identified by specific cell surface markers, which are capable of being bound by unique monoclonal antibodies.
  • A method for obtaining a cell population enriched in mesenchymal stem cells is described in, for example, U.S. Pat. No. 5,486,359, the contents of which are hereby incorporated by reference in its entirety. Adult bone marrow contains multipotent stem cells, including mesenchymal stem cells. Alternative sources for multipotent adult stem cells such as mesenchymal stem cells include, but are not limited to, blood, skin, cord blood, muscle, fat, bone, and perichondrium.
  • Multipotent adult stem cells can be obtained from a variety of sources, including bone marrow. For example, multipotent adult stem cells such as human mesenchymal stem cells can be obtained from bone marrow from a number of different sources, including plugs of femoral head cancellous bone pieces, patients with degenerative joint disease during hip or knee replacement surgery, and aspirated marrow from normal donors or oncology patients who have marrow harvested for future bone marrow transplantation. Harvested marrow can be prepared for cell culture by a number of different mechanical isolation processes depending upon the source of the harvested marrow (i.e., the presence of bone chips, peripheral blood, etc.) that are well known in the art. Exemplary culture media and culture conditions are identified in, for example, U.S. Pat. No. 5,486,359 and include media and conditions that allow for expansion, growth, and isolation of mesenchymal stem cells, without differentiation.
  • Multipotent stem cells isolated from human adult bone marrow can be surface antigen positive for integrin al (CD49a); integrin α2 (CD49b); integrin α3 (CD49c); integrin α5 (CD49e); integrin αV (CD51); integrin β1 (CD29); integrin β3 (CD61); integrin β4 (CD104); IL-1R (CD121a); IL-3Rα (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFNγR (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGFβ1R; TGFβIIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD166); hyaluronate (CD44); endoglin (CD105); Thy-1 (CD90); CD9; and combinations thereof. Cell isolated from human adult bone marrow can be surface antigen negative for integrin α4 (CD49d); integrin αL (CD11a); integrin Cβ2 (CD18); CD4; CD14; CD34; CD45; IL-2R (CD25); EGFR-3; Fas ligand, ICAM-3 (CD50); E-Selectin (CD62E); P-Selectin (CD62P); vW Factor; cadherin 5; Lewis x (CD15); and combinations thereof.
  • Cell preparations having greater than about 95%, usually greater than about 98%, of multipotent adult human stem cells can be achieved using techniques for isolation, purification, and culture expansion of stem cells. For example, isolated, cultured adult bone marrow-derived stem cells such as mesenchymal stem cells can comprise a single phenotypic population (about 95% or about 98% homogeneous) by flow cytometric analysis of expressed surface antigens. The desired cells in such composition can be identified, for example, by expression of a cell surface marker (e.g., CD73 or CD105) specifically bound by an antibody produced from hybridoma cell line SH2, ATCC accession number HB 10743, an antibody produced from hybridoma cell line SH3, ATCC accession number HB 10744, or an antibody produced from hybridoma cell line SH4, ATCC accession number HB 10745. Such antibodies selectively bind bone marrow-derived mesenchymal stem cells and, therefore, can be used to identify, quantify, isolate, or purify mesenchymal stem cells from bone marrow samples.
  • The mesenchymal stem cells can be administered by a variety of procedures. The mesenchymal stem cells can be administered systemically, such as by intravenous, intraarterial, or intraperitoneal administration.
  • The mesenchymal stem cells can be from a spectrum of sources including autologous, allogeneic, or xenogeneic.
  • The mesenchymal stem cells are administered in an amount effective to treat an autoimmune disease or graft-versus-host disease in an animal. The mesenchymal stem cells can be administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 2×106 cells/kg to about 4×106 cells/kg. In still other embodiments, the mesenchymal stem cells are administered in an amount of about 3×106 cells/kg. The amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the autoimmune disease to be treated, and the extent and severity thereof.
  • The mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier. For example, the mesenchymal stem cells can be administered as a cell suspension in a pharmaceutically acceptable liquid medium or gel for injection or topical application.
  • In accordance with another aspect of the present technology, there is provided a method of treating an inflammatory response in an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to treat the inflammatory response in the animal. Without wishing to be bound by any particular theory, the mesenchymal stem cells prevent or reverse an inflammatory response by, for example, increasing expression, production, and/or secretion of pro-inflammatory cytokines such as, for example, tumor necrosis factor-alpha (TNF-α) and Interferon-γ (IFN-γ); decreasing expression, production, and/or secretion of anti-inflammatory cytokines such as, for example, IL-10 and IL-4; and/or combinations thereof.
  • Although the scope of this aspect of the present technology is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells promote T-cell maturation to regulatory T-cells (Treg), thereby controlling inflammatory responses. It is also believed that the mesenchymal stem cells inhibit T helper 1 cells (Th1 cells), thereby decreasing the expression of the IFN-γ in certain inflammatory reactions, such as those associated with psoriasis, for example.
  • In certain embodiments, the inflammatory responses which can be treated are those associated with psoriasis.
  • In other embodiments, the mesenchymal stem cells can be administered to an animal such that the mesenchymal stem cells prevent or reduce inflammation in the brain by, for example, contacting or secreting factors that affect, microglia and/or astrocytes in the brain. The mesenchymal stem cells limit neurodegeneration caused by activated glial cells in diseases or disorders such as Alzheimer's disease, Parkinson's disease, stroke, or brain cell injuries.
  • In yet other embodiments, the mesenchymal stem cells can be administered to an animal such that the mesenchymal stem cells reduce skin inflammation as can occur in psoriasis, chronic dermatitis, and contact dermatitis by, for example, contacting or secreting factors that affect, keratinocytes and Langerhans cells in the epidermis of the skin. Although this embodiment is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells can contact the keratinocytes and Langerhans cells in the epidermis, and alter the expression of T-cell receptors and cytokine secretion profiles, leading to decreased expression of TNF-α and increased regulatory T-cell (Treg cell) population.
  • In further embodiments, the mesenchymal stem cells can be used to reduce inflammation in the bone, as occurs in arthritis and arthritis-like conditions, including but not limited to, osteoarthritis and rheumatoid arthritis, and other arthritic diseases such as ankylosing spondylitis, avascular necrosis (osteonecrosis), fibromyalgia, juvenile dermatomyositis, juvenile rheumatoid arthritis, juvenile spondyloarthopathy, lyme disease, marfan syndrome, myositis, osteogenesis imperfecta, osteoporosis, Paget's disease, Raynaud's Phenomenon, scleroderma, Sjorgren's Syndrome, and systemic lupus erythematosus. Although the scope of this embodiment is not intended to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells can inhibit Interleukin-17 secretion by memory T-cells in the synovial fluid.
  • In other embodiments, the mesenchymal stem cells can be used to limit inflammation in the gut and liver during inflammatory bowel disease and chronic hepatitis, respectively. Although the scope of this aspect of the present technology is not intended to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells promote increased secretion of IL-10 and the generation of Treg cells.
  • In other embodiments, the mesenchymal stem cells can be used to inhibit excessive neutrophil and macrophage activation in pathological conditions such as sepsis and trauma, including burn injury, surgery, and transplants. Although the scope of this embodiment is not to be limited to any theoretical reasoning, it is believed the mesenchymal stem cells promote secretion of suppressive cytokines such as IL-10, and inhibit macrophage migration inhibitory factor (MIF).
  • In other embodiments, the mesenchymal stem cells can be used to control inflammation in immune privileged sites such as the eye, including the cornea, lens, pigment epithelium, and retina, brain, spinal cord, pregnant uterus and placenta, ovary, testes, adrenal cortex, liver, and hair follicles. Although the scope of this embodiment is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells promote the secretion of suppressive cytokines such as IL-10 and the generation of Treg cells.
  • In yet other embodiments, the mesenchymal stem cells can be used to treat tissue damage associated with end-stage renal disease (ESRD) infections during dialysis and/or glomerulonephritis. Although the scope of this embodiment is not to be limited to any theoretical reasoning, it is believed that mesenchymal stem cells can promote renal repair. Mesenchymal stem cells also express and secrete vascular endothelial growth factor, or VEGF, which stimulates new blood vessel formation, which should aid in the repair of damaged kidney tissue.
  • In a further embodiment, the mesenchymal stem cells can be used to control viral infections such as influenza, hepatitis C, Herpes Simplex Virus, vaccinia virus infections, and Epstein-Barr virus. Although the scope of this embodiment is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells promote the secretion of Interferon-Beta (IFN-β).
  • In yet other embodiments, the mesenchymal stem cells can be used to control parasitic infections such as Leishmania infections and Helicobacter infections. Although the scope of this embodiment is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells mediate responses by T helper 2 (Th2) cells, and thereby promote increased production of Immunoglobulin E (IgE) by B-cells.
  • It is to be understood, however, that the scope of this aspect of the present technology is not to be limited to the treatment of any particular inflammatory response.
  • The mesenchymal stem cells can be administered to a mammal, including human and non-human primates, as described herein.
  • The mesenchymal stem cells also can be administered systemically, as described herein. Alternatively, in the case of osteoarthritis or rheumatoid arthritis, the mesenchymal stem cells can be administered directly to an arthritic joint.
  • The mesenchymal stem cells are administered in an amount effective to treat an inflammatory response in an animal. The mesenchymal stem cells can be administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 2×106 cells/kg to about 4×106 cells/kg. In still other embodiments, the mesenchymal stem cells are administered in an amount of about 3×106 cells/kg. The exact dosage of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the inflammatory response being treated, and the extent and severity thereof.
  • The mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein.
  • In accordance with another aspect of the present technology, there is provided a method of treating inflammation and/or repairing epithelial damage in an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to treat the inflammation and/or epithelial damage in the animal.
  • Although the scope of this aspect of the present technology is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells cause a decrease in the secretion of the pro-inflammatory cytokines TNF-α and IFN-γ by T-cells, and an increase in the secretion of the anti-inflammatory cytokines IL-10 and Interleukin-4 (IL-4) by T-cells. It is also believed that the mesenchymal stem cells cause a decrease in IFN-γ secretion by natural killer (NK) cells.
  • The inflammation and/or epithelial damage which can be treated in accordance with at least this aspect of the present technology includes, but is not limited to, inflammation and/or epithelial damage caused by a variety of diseases and disorders, including, but not limited to, autoimmune disease, rejection of transplanted organs, burns, cuts, lacerations, and ulcerations, including skin ulcerations and diabetic ulcerations.
  • In certain embodiments, the mesenchymal stem cells are administered to an animal at least in order to repair epithelial damage resulting from autoimmune diseases, including, but not limited to, rheumatoid arthritis, Crohn's Disease, Type 1 diabetes, multiple sclerosis, scleroderma, Graves' Disease, lupus, inflammatory bowel disease, autoimmune gastritis (AIG), and autoimmune glomerular disease. The mesenchymal stem cells also can repair epithelial damage resulting from graft-versus-host disease (GvHD).
  • This aspect of the present technology is applicable particularly to the repair of epithelial damage resulting from graft-versus-host disease, and more particularly, to the repair of epithelial damage resulting from severe graft-versus-host disease, including Grades III and IV GvHD affecting, for example, the skin and/or the gastrointestinal system. Applicants have discovered, in particular, that mesenchymal stem cells, when administered to a patient suffering from severe graft-versus-host disease, and in particular, Grades III and IV GvHD, the administration of the mesenchymal stem cells resulted in repair of skin and/or ulcerated intestinal epithelial tissue in the subject and/or patient.
  • In other embodiments, the mesenchymal stem cells are administered to an animal in order to repair epithelial damage to a transplanted organ or tissue including, but not limited to, kidney, heart, and lung, caused by rejection of the transplanted organ or tissue.
  • In yet other embodiments, the mesenchymal stem cells are administered to an animal to repair epithelial damage caused by burns, cuts, lacerations, and ulcerations, including, but not limited to, skin ulcerations and diabetic ulcerations.
  • The mesenchymal stem cells can be administered to an animal such as a mammal, including human and non-human primates, as described herein.
  • The mesenchymal stem cells also can be administered systemically, as described herein.
  • The mesenchymal stem cells are administered in an amount effective to repair epithelial damage in an animal. The mesenchymal stem cells can be administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 2×106 cells/kg to about 4×106 cells/kg. In still other embodiments, the mesenchymal stem cells are administered in an amount of about 3×106 cells/kg. The exact dosage of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the type of epithelial damage being repaired, and the extent and severity thereof.
  • In accordance with yet another aspect of the present technology, there is provided a method of treating cancer in an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to treat cancer in the animal.
  • Although the scope of this aspect of the present technology is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells interact with dendritic cells, which leads to IFN-β secretion, which in turn acts as a tumor suppressor. Cancers which can be treated include, but are not limited to, hepatocellular carcinoma, cervical cancer, pancreatic cancer, prostate cancer, fibrosarcoma, medullablastoma, and astrocytoma. It is to be understood, however, that the scope of the present technology is not to be limited to any specific type of cancer.
  • As noted herein, it shall be appreciated by those of skill in the art that the term “animal” includes a mammal, such as a human or a non-human primate.
  • The mesenchymal stem cells are administered to the animal in an amount effective to treat cancer in the animal. In general, the mesenchymal stem cells are administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. The exact amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the type of cancer being treated, and the extent and severity thereof.
  • The mesenchymal stem cells are administered in conjunction with an acceptable pharmaceutical carrier, and can be administered systemically, as described herein. Alternatively, the mesenchymal stem cells can be administered directly to the cancer being treated.
  • In accordance with still another aspect of the present technology, there is provided a method of treating an allergic disease or disorder in an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to treat the allergic disease or disorder in the animal.
  • Although the scope of this aspect of the present technology is not to be limited to any theoretical reasoning, it is believed that mesenchymal stem cells, when administered after an acute allergic response, provide for inhibition of mast cell activation and degranulation. Also, it is believed that the mesenchymal stem cells downregulate basophil activation and inhibit cytokines such as TNF-α, chemokines such as Interleukin-8 and monocyte chemoattractant protein, or MCP-1, lipid mediators such as leukotrienes, and inhibit main mediators such as histamine, heparin, chondroitin sulfates, and cathepsin.
  • Allergic diseases or disorders which can be treated include, but are not limited to, asthma, allergic rhinitis, atopic dermatitis, and contact dermatitis. It is to be understood, however, that the scope of the present technology is not to be limited to any specific allergic disease or disorder.
  • The mesenchymal stem cells are administered to the animal in an amount effective to treat the allergic disease or disorder in the animal. The animal can be a mammal. The mammal can be a primate, including human and non-human primates. In general, the mesenchymal stem cells are administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. The exact dosage is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the allergic disease or disorder being treated, and the extent and severity thereof.
  • The mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein. The mesenchymal stem cells can be administered systemically, such as by intravenous or intraarterial administration, for example.
  • In accordance with a further aspect of the present technology, there is provided a method of promoting wound healing in an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to promote wound healing in the animal.
  • Although the scope of the present technology is not to be limited to any theoretical reasoning, it is believed that, as mentioned hereinabove, the mesenchymal stem cells cause Treg cells and dendritic cells to release IL-10, which limits or controls inflammation in a wound, thereby promoting healing of a wound.
  • Furthermore, the mesenchymal stem cells can promote wound healing and fracture healing by inducing secretion factors by other cell types. For example, the mesenchymal stem cells can induce prostaglandin E2 (PGE2)-mediated release of vascular endothelial growth factor (VEGF) by peripheral blood mononuclear cells (PBMCs), as well as PGE2-mediated release of growth hormone, insulin, insulin-like growth factor 1 (IGF-1) insulin-like growth factor binding protein-3 (IGFBP-3), and endothelin-1.
  • Wounds which can be healed include, but are not limited to, those resulting from cuts, lacerations, burns, and skin ulcerations.
  • The mesenchymal stem cells are administered to the animal in an amount effective to promote wound healing in the animal. The animal can be a mammal, and the mammal can be a primate, including human and non-human primates. In general, the mesenchymal stem cells are administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. The exact amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, and the extent and severity of the wound being treated.
  • The mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein. The mesenchymal stem cells can be administered systemically, as described herein. Alternatively, the mesenchymal stem cells can be administered directly to a wound, such as in a fluid on a dressing or reservoir containing the mesenchymal stem cells.
  • In accordance with yet another aspect of the present technology, there is provided a method of treating or preventing fibrosis or fibrotic disorder in an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to treat or prevent fibrosis or a fibrotic disorder in an animal.
  • The mesenchymal stem cells can be administered to the animal in order to treat or prevent any type of fibrosis or fibrotic disorder and in the animal, including, but not limited to, cirrhosis of the liver, fibrosis of the kidneys associated with end-stage renal disease, and lung disorders or diseases having fibrotic and can include in addition, inflammatory components, including, but not limited to, Acute Respiratory Distress Syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD). It is to be understood that the scope of the present technology is not to be limited to any specific type of fibrosis or fibrotic disorder.
  • The mesenchymal stem cells are administered to the animal in an amount effective to treat or prevent fibrosis or a fibrotic disorder in the animal. The animal can be a mammal, and the mammal can be a primate, including human and non-human primates. In general, the mesenchymal stem cells are administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. The exact amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, and the extent and severity of the fibrosis or fibrotic disorder being treated or prevented.
  • The mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier, as described herein. The mesenchymal stem cells can be administered systemically, also as described herein.
  • It is another object of the present technology to promote angiogenesis (i.e., the formation of new blood vessels from a pre-existing microvascular bed) in a tissue or organ of an animal, wherein such tissue or organ is in need of angiogenesis. Thus, in accordance with a further aspect of the present technology, there is provided a method of promoting angiogenesis in an organ or tissue of an animal. The method comprises administering to the animal mesenchymal stem cells in an amount effective to promote angiogenesis in an organ or tissue of the animal.
  • The induction of angiogenesis can be used to treat coronary and peripheral artery insufficiency, and thus can be a noninvasive and curative approach to the treatment of coronary artery disease, ischemic heart disease, and peripheral artery disease. Angiogenesis can play a role in the treatment of diseases and disorders in tissue and organs other than the heart, as well as in the development and/or maintenance of organs other than the heart. Angiogenesis can provide a role in the treatment of internal and external wounds, as well as dermal ulcers. Angiogenesis also plays a role in embryo implantation, and placental growth, as well as the development of the embryonic vasculature. Angiogenesis also is essential for the coupling of cartilage resorption with bone formation, and is essential for correct growth plate morphogenesis.
  • Furthermore, angiogenesis is necessary for the successful engineering and maintenance of highly metabolic organs, such as the liver, where a dense vascular network is necessary to provide sufficient nutrient and gas transport.
  • The mesenchymal stem cells can be administered to the tissue or organ in need of angiogenesis by a variety of procedures. The mesenchymal stem cells can be administered systemically, such as by intravenous, intraarterial, or intraperitoneal administration, or the mesenchymal stem cells can be administered directly to the tissue or organ in need of angiogenesis, such as by direct injection into the tissue or organ in need of angiogenesis.
  • The mesenchymal stem cells can be from a spectrum of sources including autologous, allogeneic, or xenogeneic.
  • Although the scope of the present technology is not to be limited to any theoretical reasoning, it is believed that the mesenchymal stem cells, when administered to an animal, stimulate peripheral blood mononuclear cells (PBMCs) to produce vascular endothelial growth factor, or VEGF, which stimulates the formation of new blood vessels.
  • In certain embodiments, the animal is a mammal. The mammal can be a primate, including human and non-human primates.
  • The mesenchymal stem cells, in accordance with the present technology, can be employed in the treatment, alleviation, or prevention of any disease or disorder which can be alleviated, treated, or prevented through angiogenesis. Thus, for example, the mesenchymal stem cells can be administered to an animal to treat blocked arteries, including those in the extremities, such as arms, legs, hands, and feet, as well as the neck or in various organs. For example, the mesenchymal stem cells can be used to treat blocked arteries which supply the brain, thereby treating or preventing stroke. Also, the mesenchymal stem cells can be used to treat blood vessels in embryonic and post-natal corneas and can be used to provide glomerular structuring. In other embodiments, the mesenchymal stem cells can be employed in the treatment of wounds, both internal and external, as well as the treatment of dermal ulcers found in the feet, hands, legs or arms, including, but not limited to, dermal ulcers caused by diseases such as diabetes and sickle cell anemia.
  • Furthermore, because angiogenesis is involved in embryo implantation and placenta formation, the mesenchymal stem sells can be employed to promote embryo implantation and prevent miscarriage.
  • In addition, the mesenchymal stem cells can be administered to an unborn animal, including humans, to promote the development of the vasculature in the unborn animal.
  • In other embodiments, the mesenchymal stem cells can be administered to an animal, born or unborn, in order to promote cartilage resorption and bone formation, as well as promote correct growth plate morphogenesis.
  • The mesenchymal stem cells are administered in an amount effective in promoting angiogenesis in an animal. The mesenchymal stem cells can be administered in an amount of from about 1×105 cells/kg to about 1×107 cells/kg. In other embodiments, the mesenchymal stem cells are administered in an amount of from about 1×106 cells/kg to about 5×106 cells/kg. The amount of mesenchymal stem cells to be administered is dependent upon a variety of factors, including the age, weight, and sex of the subject and/or patient, the disease or disorder to be treated, alleviated, or prevented, and the extent and severity thereof.
  • The mesenchymal stem cells can be administered in conjunction with an acceptable pharmaceutical carrier. For example, the mesenchymal stem cells can be administered as a cell suspension in a pharmaceutically acceptable liquid medium for injection. Injection can be local, such as by administration directly into the tissue or organ in need of angiogenesis, or systemic, such as intravenously or intraarterially.
  • The mesenchymal stem cells can be genetically engineered with one or more polynucleotides encoding a therapeutic agent. The polynucleotides can be delivered to the mesenchymal stem cells via an appropriate expression vehicle. Expression vehicles which can be employed to genetically engineer the mesenchymal stem cells include, but are not limited to, retroviral vectors, adenoviral vectors, and adeno-associated virus vectors.
  • The selection of an appropriate polynucleotide encoding a therapeutic agent is dependent upon various factors, including the disease or disorder being treated, and the extent and severity thereof. Polynucleotides encoding therapeutic agents, and appropriate expression vehicles are described further in U.S. Pat. No. 6,355,239, the contents of which are hereby incorporated by reference in its entirety.
  • It is to be understood that the mesenchymal stem cells, when employed in the above-mentioned therapies and treatments, can be employed in combination with other therapeutic agents known to those skilled in the art, including, but not limited to, growth factors, cytokines, drugs such as anti-inflammatory drugs, and cells other than mesenchymal stem cells, such as dendritic cells, and can be administered with soluble carriers for cells such as hyaluronic acid, or in combination with solid matrices, such collagen, gelatin, or other biocompatible polymers, as appropriate.
  • It is to be understood that the methods described herein can be carried out in a number of ways and with various modifications and permutations thereof that are well known in the art. It also can be appreciated that any theories set forth as to modes of action or interactions between cell types should not be construed as limiting this technology in any manner, but are presented such that the methods of the present technology can be understood more fully.
  • It is to be understood that the scope of the present technology is not to be limited to the specific embodiments described above. The present technology can be practiced other than as particularly described and still be within the scope of the accompanying claims.
  • Likewise, the following examples are presented in order to more fully illustrate the present technology. They should in no way be construed, however, as limiting the broad scope of the technology disclosed herein.
  • EXAMPLES
  • The present technology now will be described with respect to the following examples; it is to be understood, however, that the scope of the present technology is not to be limited thereby.
  • Example 1
  • Materials and Methods Culture of human MSCs. Human MSCs were cultured as described by Pittenger et al., Science, Vol. 284, pg. 143 (1999). Briefly, marrow samples were collected from the iliac crest of anonymous donors following informed consent by Poietics Technologies, Div of Cambrex Biosciences. MSCs were cultured in complete Dulbecco's Modified Eagle's Medium-Low Glucose (Life Technologies, Carlsbad, Calif.) containing 1% antibiotic-antimyotic solution (Invitrogen, Carlsbad, Calif.) and 10% fetal bovine serum (FBS, JRH BioSciences, Lenexa, Kans.). MSCs grew as an adherent monolayer and were detached with trypsin/EDTA (0.05% trypsin at 37° C. for 3 minutes). All MSCs used were previously characterized for multilineage potential and retained the capacity to differentiate into mesenchymal lineages (chondrocytic, adipogenic, and osteogenic) (Pittenger, et al., Science, Vol. 284, pg. 143 (1999)).
  • Isolation of Dendritic cells. Peripheral blood mononuclear cells (PBMCs) were obtained from Poietics Technologies, Div of Cambrex Biosciences (Walkersville, Md.). Precursors of dendritic cells (DCs) of monocytic lineage (CD1c+) were positively selected from PBMCs using a 2-step magnetic separation method according to Dzionek, et. al., J. Immunol., Vol. 165, pg. 6037 (2000). Briefly, CD1c expressing B cells were magnetically depleted of CD19+ cells using magnetic beads, followed by labeling the B-cell depleted fraction with biotin-labeled CD1c (BDCA1+) and anti-biotin antibodies and separating them from the unlabeled cell fraction utilizing magnetic columns according to the manufacturer's instructions (Miltenyi Biotech, Auburn, Calif.). Precursors of DCs of plasmacytoid lineage were isolated from PBMCs by immuno-magnetic sorting of positively labeled antibody coated cells (BDCA2+) (Miltenyi Biotech, Auburn, Calif.).
  • MSC-DC culture. In most experiments, human MSCs and DCs were cultured in equal numbers for various time periods and cell culture supernatant collected and stored at −80° C. until further evaluation. In selected experiments, MSCs were cultured with mature DC1 or DC2 cells (1:1 MSC:DC ratio) for 3 days, and then the combined cultures (MSCs and DCs) were irradiated to prevent any proliferation. Next, antibody purified, naïve, allogeneic T cells (CD4+,CD45RA+) were added to the irradiated MSCs/DCs and cultured for an additional 6 days. The non-adherent cell fraction (purified T cells) was then collected from the cultures, washed twice and re-stimulated with PHA for another 24 hours, following which cell culture supernatants were harvested and analyzed for secreted IFN-γ and IL-4 by ELISA.
  • Isolation of NK cells. Purified populations of NK cells were obtained by depleting non-NK cells that are magnetically labeled with a cocktail of biotin-conjugated monoclonal antibodies (anti-CD3, -CD14, -CD19, -CD36 and anti-IgE antibodies) as a primary reagent and anti-biotin monoclonal antibodies conjugated to Microbeads as secondary labeling reagent. The magnetically labeled non-NK cells were retained in MACS (Miltenyi Biotech, Auburn, Calif.) columns in a magnetic field, while NK cells passed through and were collected.
  • Isolation of TReg cell population. The TReg cell population was isolated using a 2-step isolation procedure. First non-CD4+T cells were indirectly magnetically labeled with a cocktail of biotin labeled antibodies and anti-biotin microbeads. The labeled cells were then depleted by separation over a MACS column (Miltenyi Biotech, Auburn, Calif.). Next, CD4+CD25+ cells were directly labeled with CD25 microbeads and isolated by positive selection from the pre-enriched CD4+. T cell fraction. The magnetically labeled CD4+CD25+T cells were retained on the column and eluted after removal of the column from the magnetic field.
  • In order to determine whether the increased CD4+CD25+ population generated in the presence of MSCs were suppressive in nature, CD4+CD25+Treg cell populations were isolated from PBMC or MSC+PBMC (MSC to PBMC ratio 1:10) cultures (cultured without any further stimulation for 3 days) using a 2-step magnetic isolation procedure. These cells were irradiated to block any further proliferation and used as stimulators in a mixed lymphocyte reaction (MLR), where responders were allogeneic PBMCs (stimulator to responder ratio 1:100) in the presence of PHA (2.5 μg/ml). The culture was carried out for 48 hours, following which 3H thymidine was added. Incorporated radioactivity was counted after 24 hours.
  • PBMCs were cultured in the absence or presence of MSCs (MSC to PBMC ratio 1:10), following which the non-adherent fraction was harvested and immunostained with FITC-labeled glucocorticoid-induced TNF receptor, or GITR, and PE-labeled CD4.
  • Generation of Th1/Th2 cells. Peripheral blood mononuclear cells (PBMCs) were plated at 2×106 cells/ml for 45 min. at 37° C. in order to remove monocytes. Non-adherent fraction was incubated in the presence of plate-bound anti-CD3 (5 μg/ml) and anti-CD28 (1 μg/ml) antibodies under Th1 (IL-2 (4 ng/ml) IL-12 (5 ng/ml)+anti-IL-4 (1 μg/ml)) or Th2 (IL-2 (4 ng/ml)+IL-4 (4 ng/ml)+anti-IFN-γ (1 μg/ml)) conditions for 3 days in the presence or absence of MSCs. The cells were washed and then re-stimulated with PHA (2.5 μg/ml) for another 24 or 48 hours, following which levels of IFN-γ and IL-4 were measured in culture supernatants by ELISA (R&D Systems, Minneapolis, Minn.).
  • Analysis of levels of VEGF, PGE2, and pro-MMP-1 in culture supernatant of MSCs. Using previously characterized human MSCs, the levels of Interleukin-6 (IL-6), VEGF, lipid mediator PGE2, and matrix metalloproteinase 1 (pro-MMP-1) were analyzed in culture supernatant of MSCs cultured for 24 hours in the presence or absence of PBMCs (MSC to PBMC ratio 1:10).
  • Proliferation of PBMCs. Purified PBMCs were prepared by centrifuging leukopack (Cambrex, Walkersville, Md.) on Ficoll-Hypaque (Lymphoprep, Oslo, Norway). Separated cells were cultured (in triplicates) in the presence or absence of MSCs (plated 3-4 hours prior to PBMC addition to allow them to settle) for 48 hours in presence of the mitogen PHA (Sigma Chemicals, St. Louis, Mo.). In selected experiments, PBMCs were resuspended in medium containing PGE2 inhibitors Indomethacin (Sigma Chemicals, St. Louis, Mo.) or NS-938 (Cayman Chemicals, Ann Arbor, Mich.). (3H)-thymidine was added (20 μI in a 200 μl culture) and the cells harvested after an additional 24 hour culture using an automatic harvester. The effects of MSCs or PGE2 blockers were calculated as the percentage of the control response (100%) in presence of PHA.
  • Quantitative RT-PCR. Total RNA from cell pellets were prepared using a commercially available kit (Qiagen, Valencia, Calif.) and according to the manufacturer's instructions. Contaminating genomic DNA was removed using the DNA-free kit (Ambion, Austin, Tex.). Quantitative RT-PCR was performed on a MJ Research Opticon detection system (South San Francisco, Calif.) using QuantiTect SYBR Green RT-PCR kit (Qiagen, Valencia, Calif.) with primers at concentration of 0.5 μM. Relative changes in expression levels in cells cultured under different conditions were calculated by the difference in Ct values (crossing point) using β-actin as internal control. The sequences for COX-1 and COX-2 specific primers were: COX-1: 5′-CCG GAT GCC AGT CAG GAT GAT G-3′ (forward) (SEQ ID NO:1), 5′-CTA GAC AGC CAG ATG CTG ACA G-3′ (reverse) (SEQ ID NO:2); COX-2: 5′-ATC TAC CCT CCT CAA GTC CC-3′ (forward) (SEQ ID NO:3), 5′-TAC CAG AAG GGC AGG ATA CAG-3′ (reverse) (SEQ ID NO:4).
  • Increasing numbers of allogeneic PBMCs were incubated with constant numbers of MSCs (2,000 cells/well) plated on a 96-well plate in the presence of PHA (2.5 μg/ml) for 72 hours, and 3H thymidine incorporation (counts per minute, cpm) was determined. The PBMCs and MSCs were cultured at ratios of MSC:PBMC of 1:1, 1:3, 1:10, 1:30, and 1:81.
  • Results. In the present studies, the interaction of human MSCs with isolated immune cell populations, including dendritic cells (DC1 and DC2), effector T cells (Th1 and Th2) and NK cells was examined. The interaction of MSCs with each immune cell type had specific consequences, suggesting that MSCs can modulate several steps in the immune response process. The production of secreted factor(s) that modulate and can be responsible for MSC immuno-modulatory effects was evaluated and prostaglandin synthesis was implicated.
  • Myeloid (DC1) and plasmacytoid (DC2) precursor dendritic cells were isolated by immuno-magnetic sorting of BDCA1+ and BDCA2+ cells respectively and matured by incubation with GM-CSF and IL-4 (1×103 IU/ml and 1×103 IU/ml, respectively) for DC1 cells, or IL-3 (10 ng/ml) for DC2 cells. Using flow cytometry, DC1 cells were HLA-DR+ and CD11c+, whereas DC2 cells were HLA-DR+ and CD123+ (FIG. 1A). In the presence of the inflammatory agent bacterial lipopolysaccharide (LPS, 1 ng/ml), DC1 cells produced moderate levels of TNF-α but when MSCs were present (ratios examined 1:1 and 1:10), there was >50% reduction in TNF-α secretion (FIG. 1B). On the other hand, DC2 cells produced IL-10 in the presence of LPS and its levels were increased greater than 2-fold upon MSC:DC2 co-culture (1:1) (FIG. 1B). Therefore, the MSCs modified the cytokine profile of activated DCs in culture towards a more tolerogenic phenotype. Additionally, activated DCs, when cultured with MSCs, were able to reduce IFN-γ and increase IL-4 levels secreted by naïve CD4+T cells (FIG. 1C) suggesting a MSC-mediated shift from pro-inflammatory to anti-inflammatory T cell phenotype.
  • As increased IL-10 secretion plays a role in generation of regulatory cells (Kingsley, et al., J. Immunol., Vol. 168, pg. 1080 (2002)), T-regulatory cells (TReg) were quantified by flow cytometry in co-cultures of PBMCs and MSCs. Upon culture of PBMCs with MSCs for 3-5 days, there was an increase in TReg cell numbers as determined by staining of PBMCs with anti-CD4 and anti-CD25 antibodies (FIG. 2A), further supporting a MSC-induced tolerogenic response. The CD4+CD25+ TReg cell population, generated in presence of MSCs expressed increased levels of gluocorticoid-induced TNF receptor (GITR), a cell surface receptor expressed on TReg cell populations, and was suppressive in nature as it suppressed allogeneic T cell proliferation (FIG. 3A,B). Next, MSCs were investigated as to their direct ability to affect T cell differentiation. Using antibody selected purified T cells (CD4+Th cells), IFN-γ producing Th1 and IL-4 producing T H2 cells were generated in presence or absence of MSCs. When MSCs were present during differentiation, there was reduced IFN-γ secretion by Th1 cells and increased IL-4 secretion by Th2 cells (FIG. 2B). No significant change in IFN-γ or IL-4 levels were seen when MSCs were added to the culture after Th cells had differentiated (at 3 days) into effector Th1 or Th2 types (data not shown). These experiments suggest that MSCs can affect effector T cell differentiation directly and alter the T cell cytokine secretion towards a humoral phenotype.
  • Similarly, when MSCs were cultured with purified NK cells (CD3-, CD14-, CD19-, CD36″) at a ratio 1:1 for different time periods (0-48 hrs), there was decreased IFN-γ secretion in the culture supernatant (FIG. 2C), thereby suggesting that MSCs can modulate NK cell functions also.
  • Previous work has indicated that MSCs modify T-cell functions by soluble factor(s) (LeBlanc, et al., Exp. Hematol., Vol. 31, pg. 890 (2003); Tse, et al., Transplantation, Vol. 75, pg. 389 (2003). It was observed that the MSCs secreted several factors, including IL-6, prostaglandin E2, VEGF and proMMP-lconstitutively, and the levels of each increased upon culture with PBMCs (FIG. 5). In order to investigate MSC-derived factors leading to inhibition of TNF-α and increase of IL-10 production by DCs, the potential role of prostaglandin E2 was investigated, as it has been shown to inhibit TNF-α production by activated DCs (Vassiliou, et al., Cell. Immunol., Vol. 223, pg. 120 (2003)). Conditioned media from MSC culture (24 hour culture of 0.5×106 cells/ml) contained approximately 1000 pg/ml of PGE2 (FIG. 4A). There was no detectable presence of known inducers of PGE2 secretion, such as TNF-α, IFN-γ or IL-10 (data not shown), in the culture supernatant indicating a constitutive secretion of PGE2 by MSCs. The PGE2 secretion by hMSCs was inhibited 60-90% in the presence of known inhibitors of PGE2 production, NS-398 (5 μM) and indomethacin (4 μM) (FIG. 4A). As the release of PGE2 secretion occurs as a result of enzymatic activity of constitutively active cycloxygenase enzyme 1 (COX-1) and inducible cycloxygenase enzyme 2 (COX-2) (Harris, et al., Trends Immunol., Vol. 23, pg. 144 (2002)) the mRNA expression for COX-1 and COX-2 in MSCs and PBMCs using trans-well culture system was analyzed. MSCs expressed significantly higher levels of COX-2 as compared to PBMCs and the expression levels increase >3-fold upon co-culture of MSCs and PBMCs (MSC to PBMC ratio 1:10) for 24 hours (FIG. 4B). Modest changes in COX-1 levels were seen suggesting that the increase in PGE2 secretion upon MSC-PBMC co-culture (FIG. 5) is mediated by COX-2 up-regulation.
  • To investigate whether the immunomodulatory effects of MSC on DCs and T-cells were mediated by PGE2, MSCs were cultured with activated dendritic cells (DC1) or Th1 cells in the presence of PGE2 inhibitors NS-398 or indomethacin. The presence of NS-398 or indomethacin increased TNF-α secretion by DC1s, and IFN-γ secretion from Th1 cells (FIG. 4C), respectively, suggesting that MSC effects on immune cell types can be mediated by secreted PGE2. Recent studies have shown that MSCs inhibit T-cell proliferation induced by various stimuli (DeNicola, et al., Blood, Vol. 99, pg. 3838 (2002); LeBlanc, et al., Scand. J. Immunol., Vol. 57, pg. 11 (2003)). It was observed that MSCs inhibit mitogen-induced T cell proliferation in a dose-dependent manner (FIG. 6) and when PGE2 inhibitors NS-398 (5 μM) or indomethacin (4 μM) were present, there was a >70% increase in (3H) thymidine incorporation by PHA-treated PBMCs in MSC containing cultures as compared to controls without inhibitors (FIG. 4D).
  • In summary, a model of MSC interaction with other immune cell types (FIG. 7) is proposed. When mature T cells are present, MSCs can interact with them directly and inhibit the pro-inflammatory IFN-γ production (pathway 1) and promote regulatory T cell phenotype (pathway 3) and anti-inflammatory T H2 cells (pathway 5). Further, MSCs can alter the outcome of the T cell immune response through DCs by secreting PGE2, inhibiting pro-inflammatory DC1 cells (pathway 2) and promoting anti-inflammatory DC2 cells (pathway 4) or regulatory DCs (pathway 3). A shift towards T H2 immunity in turn, suggests a change in B cell activity towards increased generation of IgE/IgG1 subtype antibodies (pathway 7). MSCs, by their ability to inhibit IFN-γ secretion from NK cells likely modify NK cell function (pathway 6). This model of MSC:Immune cell interactions is consistent with the experimentation performed in several other laboratories (LeBlanc, et al., Exp. Hematol., Vol. 31, pg. 890 (2003); Tse, et al., Transplantation, Vol. 75, pg. 389 (2003); DiNicola, et al., Blood, Vol. 99, pg. 3838 (2002)). Further examination of the proposed mechanisms is underway and animal studies are now necessary to examine the in vivo effects of MSC administration.
  • Example 2
  • Mesenchymal stem cells were given to a 33-year-old female patient suffering from severe Grade IV gastrointestinal graft-versus-host disease (GVHD). The patient was refractory to all other GVHD treatments. Endoscopic views of the patient's colon showed areas of ulceration and inflammation prior to treatment. Histology of the patient's colon showed that the graft-versus-host disease had destroyed the vast majority of the patient's intestinal crypts, prior to treatment.
  • The patient was given an intravenous infusion of allogeneic mesenchymal stem cells in 50 ml of Plasma Lyte A (Baxter) in an amount of 3×106 cells per kilogram of body weight.
  • The patient was evaluated at two weeks post-infusion. At two weeks post-infusion, an endoscopic view of the patient's colon showed that the areas of inflammation and ulceration visible prior to treatment were resolved. In addition, a biopsy of the patient's colon showed significant regeneration of intestinal crypts. Thus, the administration of the mesenchymal stem cells to the patient resulted in a significant reduction in the inflammatory component of gastrointestinal graft-versus-host disease, and resulted in the regeneration of new functional intestinal tissue.
  • The disclosures of all patents, publications, including published patent applications, depository accession numbers, and database accession numbers are hereby incorporated by reference in their entireties to the same extent as if each patent, publication, depository accession number, and database accession number were specifically and individually incorporated by reference.
  • The presently described technology is now described in such full, clear, concise and exact terms as to enable any person skilled in the art to which it pertains, to practice the same. It is to be understood that the foregoing describes preferred embodiments of the technology and that modifications can be made therein without departing from the spirit or scope of the invention as set forth in the appended claims.
  • It is to be understood, however, that the scope of the present technology is not to be limited to the specific embodiments described above. The present technology can be practiced other than as particularly described and still be within the scope of the accompanying claims.

Claims (23)

1. A cell preparation comprising adult bone marrow-derived stem cells in a dose effective to treat an inflammatory response in a subject.
2. The cell preparation of claim 1, wherein the stem cells are capable of differentiating into at least one cell type of each of the endodermal, ectodermal, and mesodermal embryonic lineages.
3. The cell preparation of claim 1, wherein the stem cells are capable of differentiating into at least one cell type of at least one of the endodermal, ectodermal, or mesodermal embryonic lineages.
4. The cell preparation of claim 1, wherein the dose contains a sufficient number of stem cells to provide about 1×105 to about 1×107 cells per kilogram of the subject.
5. The cell preparation of claim 1, wherein the stem cells are positive for one or more cell surface markers selected from the group consisting of integrin al (CD49a); integrin α2 (CD49b); integrin α3 (CD49c); integrin α5 (CD49e); integrin αV (CD51); integrin β1 (CD29); integrin β3 (CD61); integrin β4 (CD104); IL-1R (CD121a); IL-3Rα (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFNγR (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGFβ1R; TGFβIIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD166); hyaluronate (CD44); endoglin (CD105); Thy-1 (CD90); and CD9.
6. The cell preparation of claim 1, wherein administration of the cell preparation elevates interferon-beta levels in the subject.
7. The cell preparation of claim 1, wherein the subject is a subject having inflammatory bowel disease.
8. A method of treating inflammatory bowel disease in a subject, comprising the step of:
administering to the subject allogeneic, multipotent adult bone marrow-derived stem cells in an amount effective to treat the inflammatory bowel disease.
9. The method of claim 8, wherein the multipotent stem cells are administered intravenously or intraarterially.
10. The method of claim 8, wherein the multipotent stem cells are positive for one or more cell surface markers selected from the group consisting of integrin al (CD49a); integrin α2 (CD49b); integrin α3 (CD49c); integrin α5 (CD49e); integrin αV (CD51); integrin β1 (CD29); integrin β3 (CD61); integrin β4 (CD104); IL-1R (CD121a); IL-3Rα (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFNγR (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGFβ1R; TGFβIIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD166); hyaluronate (CD44); endoglin (CD105); Thy-1 (CD90); and CD9.
11. The method of claim 8, wherein the multipotent stem cells are positive for CD49b, CD49e, and CD140a.
12. The method of claim 8, wherein the multipotent stem cells are negative for one or more cell surface markers selected from the group consisting of integrin α4 (CD49d); integrin αL (CD11a); integrin Cβ2 (CD18); CD4; CD14; CD34; CD45; IL-2R (CD25); EGFR-3; Fas ligand; ICAM-3 (CD50); E-Selectin (CD62E); P-Selectin (CD62P); vW Factor; cadherin 5; and Lewis x (CD15).
13. The method of claim 8, wherein the multipotent stem cells are negative for CD34, CD45, CD62E, and CD62P.
14. A method of treating a gastrointestinal autoimmune disease in a subject, comprising the step of:
administering to the subject allogeneic multipotent adult stem cells in an amount effective to repair or regenerate intestinal tissue.
15. The method of claim 14, wherein the autoimmune disease is selected from the group consisting of Crohn's disease, inflammatory bowel disease, and autoimmune gastritis.
16. The method of claim 14, wherein the multipotent stem cells are administered intravenously or intraarterially.
17. The method of claim 14, wherein the multipotent stem cells are positive for one or more cell surface markers selected from the group consisting of integrin al (CD49a); integrin α2 (CD49b); integrin α3 (CD49c); integrin α5 (CD49e); integrin αV (CD51); integrin β1 (CD29); integrin β3 (CD61); integrin β4 (CD104); IL-1R (CD121a); IL-3Rα (CD123); IL-4R (CDw124); IL-6R (CD126); IL-7R (CDw127); IFNγR (CDw119); TNFIR (CD120a); TNFIIR (CD120b); TGFβ1R; TGFβIIR; bFGFR; PDGFR (CD140a); transferrin (CD71); ICAM-1 (CD54); ICAM-2 (CD102); VCAM-1 (CD106); L-Selectin (CD62L); LFA-3 (CD58); ALCAM (CD166); hyaluronate (CD44); endoglin (CD105); Thy-1 (CD90); and CD9.
18. The method of claim 14, wherein the multipotent stem cells are positive for CD49b, CD49e, and CD140a.
19. The method of claim 14, wherein the multipotent stem cells are negative for one or more cell surface markers selected from the group consisting of integrin α4 (CD49d); integrin αL (CD11a); integrin Cβ2 (CD18); CD4; CD14; CD34; CD45; IL-2R (CD25); EGFR-3; Fas ligand; ICAM-3 (CD50); E-Selectin (CD62E); P-Selectin (CD62P); vW Factor; cadherin 5; and Lewis x (CD15).
20. The method of claim 14, wherein the multipotent stem cells are negative for CD34, CD45, CD62E, and CD62P.
21. A method of repairing or regenerating intestinal tissue in a human, comprising the step of:
treating the human with allogeneic adult mesenchymal stem cells.
22. The method of claim 21, wherein the mesenchymal stem cells are administered to the human.
23. The method of claim 22, wherein the administration of the mesenchymal stem cell comprises intravenous, intraarterial, or intraperitoneal injection.
US12/727,971 2004-03-22 2010-03-19 Multipotent Adult Stem Cells And Uses of Multipotent Adult Stem Cells To Treat Inflammation Abandoned US20100172885A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/727,971 US20100172885A1 (en) 2004-03-22 2010-03-19 Multipotent Adult Stem Cells And Uses of Multipotent Adult Stem Cells To Treat Inflammation

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US55511804P 2004-03-22 2004-03-22
US11/080,298 US20050239897A1 (en) 2004-03-22 2005-03-15 Mesenchymal stem cells and uses therefor
US11/541,853 US20080213227A1 (en) 2004-03-22 2006-10-02 Mesenchymal stem cells and uses therefor
US12/727,971 US20100172885A1 (en) 2004-03-22 2010-03-19 Multipotent Adult Stem Cells And Uses of Multipotent Adult Stem Cells To Treat Inflammation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/541,853 Continuation US20080213227A1 (en) 2004-03-22 2006-10-02 Mesenchymal stem cells and uses therefor

Publications (1)

Publication Number Publication Date
US20100172885A1 true US20100172885A1 (en) 2010-07-08

Family

ID=39772836

Family Applications (21)

Application Number Title Priority Date Filing Date
US11/080,298 Abandoned US20050239897A1 (en) 2004-03-22 2005-03-15 Mesenchymal stem cells and uses therefor
US11/541,853 Abandoned US20080213227A1 (en) 2004-03-22 2006-10-02 Mesenchymal stem cells and uses therefor
US12/091,387 Abandoned US20090208463A1 (en) 2004-03-22 2007-09-26 Mesenchymal stem cells and uses therefor
US12/323,129 Abandoned US20090180997A1 (en) 2004-03-22 2008-11-25 Mesenchymal Stem Cells And Uses Therefor
US12/727,971 Abandoned US20100172885A1 (en) 2004-03-22 2010-03-19 Multipotent Adult Stem Cells And Uses of Multipotent Adult Stem Cells To Treat Inflammation
US12/908,119 Abandoned US20110027238A1 (en) 2004-03-22 2010-10-20 Mesenchymal Stem Cells and Uses Therefor
US12/974,125 Abandoned US20110142807A1 (en) 2004-03-22 2010-12-21 Mesenchymal Stem Cells and Uses Therefor
US13/222,701 Abandoned US20110311496A1 (en) 2004-03-22 2011-08-31 Mesenchymal stem cells and uses therefor
US13/222,831 Abandoned US20110318315A1 (en) 2004-03-22 2011-08-31 Mesenchymal stem cells and uses therefor
US13/222,778 Abandoned US20110318314A1 (en) 2004-03-22 2011-08-31 Mesenchymal stem cells and uses therefor
US14/087,830 Active US9943547B2 (en) 2004-03-22 2013-11-22 Mesenchymal stem cells and uses therefor
US14/138,577 Active US10668101B2 (en) 2004-03-22 2013-12-23 Mesenchymal stem cells and uses therefor
US14/305,240 Abandoned US20140328807A1 (en) 2004-03-22 2014-06-16 Mesenchymal Stem Cells and Uses Therefor
US14/334,128 Active US9694035B2 (en) 2004-03-22 2014-07-17 Mesenchymal stem cells and uses therefor
US14/739,924 Abandoned US20150272997A1 (en) 2004-03-22 2015-06-15 Mesenchymal Stem Cells and Uses Therefor
US16/204,936 Active US10729727B2 (en) 2004-03-22 2018-11-29 Mesenchymal stem cells and uses therefor
US16/206,235 Active US10716814B2 (en) 2004-03-22 2018-11-30 Mesenchymal stem cells and uses therefor
US16/214,485 Active 2026-09-15 US11389484B2 (en) 2004-03-22 2018-12-10 Mesenchymal stem cells and uses therefor
US16/811,900 Active US10960025B2 (en) 2004-03-22 2020-03-06 Mesenchymal stem cells and uses therefor
US16/815,784 Active US10828334B1 (en) 2004-03-22 2020-03-11 Mesenchymal stem cells and uses therefor
US17/836,786 Pending US20230076630A1 (en) 2004-03-22 2022-06-09 Mesenchymal stem cells and uses therefor

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US11/080,298 Abandoned US20050239897A1 (en) 2004-03-22 2005-03-15 Mesenchymal stem cells and uses therefor
US11/541,853 Abandoned US20080213227A1 (en) 2004-03-22 2006-10-02 Mesenchymal stem cells and uses therefor
US12/091,387 Abandoned US20090208463A1 (en) 2004-03-22 2007-09-26 Mesenchymal stem cells and uses therefor
US12/323,129 Abandoned US20090180997A1 (en) 2004-03-22 2008-11-25 Mesenchymal Stem Cells And Uses Therefor

Family Applications After (16)

Application Number Title Priority Date Filing Date
US12/908,119 Abandoned US20110027238A1 (en) 2004-03-22 2010-10-20 Mesenchymal Stem Cells and Uses Therefor
US12/974,125 Abandoned US20110142807A1 (en) 2004-03-22 2010-12-21 Mesenchymal Stem Cells and Uses Therefor
US13/222,701 Abandoned US20110311496A1 (en) 2004-03-22 2011-08-31 Mesenchymal stem cells and uses therefor
US13/222,831 Abandoned US20110318315A1 (en) 2004-03-22 2011-08-31 Mesenchymal stem cells and uses therefor
US13/222,778 Abandoned US20110318314A1 (en) 2004-03-22 2011-08-31 Mesenchymal stem cells and uses therefor
US14/087,830 Active US9943547B2 (en) 2004-03-22 2013-11-22 Mesenchymal stem cells and uses therefor
US14/138,577 Active US10668101B2 (en) 2004-03-22 2013-12-23 Mesenchymal stem cells and uses therefor
US14/305,240 Abandoned US20140328807A1 (en) 2004-03-22 2014-06-16 Mesenchymal Stem Cells and Uses Therefor
US14/334,128 Active US9694035B2 (en) 2004-03-22 2014-07-17 Mesenchymal stem cells and uses therefor
US14/739,924 Abandoned US20150272997A1 (en) 2004-03-22 2015-06-15 Mesenchymal Stem Cells and Uses Therefor
US16/204,936 Active US10729727B2 (en) 2004-03-22 2018-11-29 Mesenchymal stem cells and uses therefor
US16/206,235 Active US10716814B2 (en) 2004-03-22 2018-11-30 Mesenchymal stem cells and uses therefor
US16/214,485 Active 2026-09-15 US11389484B2 (en) 2004-03-22 2018-12-10 Mesenchymal stem cells and uses therefor
US16/811,900 Active US10960025B2 (en) 2004-03-22 2020-03-06 Mesenchymal stem cells and uses therefor
US16/815,784 Active US10828334B1 (en) 2004-03-22 2020-03-11 Mesenchymal stem cells and uses therefor
US17/836,786 Pending US20230076630A1 (en) 2004-03-22 2022-06-09 Mesenchymal stem cells and uses therefor

Country Status (8)

Country Link
US (21) US20050239897A1 (en)
EP (10) EP1727892B1 (en)
JP (2) JP2007530543A (en)
KR (1) KR20090065478A (en)
CN (1) CN101384702A (en)
AU (3) AU2005227295A1 (en)
CA (2) CA2564679C (en)
WO (2) WO2005093044A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100150888A1 (en) * 2008-11-14 2010-06-17 Howmedica Osteonics Corp. Cells for joint fluid
US20100183571A1 (en) * 2005-10-13 2010-07-22 Anthrogenesis Corporation Treatment of multiple sclerosis using placental stem cells
US20110020293A1 (en) * 2009-07-21 2011-01-27 Abt Holding Company Use of Stem Cells to Reduce Leukocyte Extravasation
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
WO2014018230A2 (en) * 2012-07-25 2014-01-30 Albert Einstein College Of Medicine Of Yeshiva University Methods to isolate human mesenchymal stem cells
US8709401B2 (en) 2011-02-25 2014-04-29 Howmedica Osteonics Corp. Primed stem cells and uses thereof to treat inflammatory conditions in joints
WO2015066552A1 (en) * 2013-11-04 2015-05-07 Becton, Dickinson And Company Immunomodulatory potential of a multipotent stromal cell (msc) population
WO2014113704A3 (en) * 2013-01-18 2015-11-26 Escape Therapeutics, Inc. Enhanced differentiation of mesenchymal stem cells
US20170189452A1 (en) * 2004-08-25 2017-07-06 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
WO2017127123A1 (en) * 2016-01-21 2017-07-27 Abt Holding Company Stem cells for wound healing
US20170239295A1 (en) * 2011-02-18 2017-08-24 Catholic University Industry Academic Cooperation Foundation Cell therapy composition for preventing or treating immune disease comprising mesenchymal stem cells and immunoregulatory t-cells as active ingredient
US9861660B2 (en) 2013-04-12 2018-01-09 Saverio LaFrancesca Organs for transplantation
US9937208B2 (en) 2010-05-12 2018-04-10 Abt Holding Company Modulation of splenocytes in cell therapy
US20180214489A1 (en) * 2017-01-27 2018-08-02 Neil Riordan Mesenchymal stem cells with enhanced efficacy in treatment of autoimmunity particularly rheumatoid arthritis
US10729726B2 (en) 2004-10-04 2020-08-04 Tigenix, S.A.U. Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
US10907131B2 (en) 2012-02-10 2021-02-02 Orbsen Therapeutics Limited Stromal stem cells
US11268067B2 (en) 2017-07-14 2022-03-08 Orbsen Therapeutics Limited Methods of isolation and use of CD39 stromal stem cells
US11903997B2 (en) 2015-03-20 2024-02-20 Orbsen Therapeutics Limited Modulators of syndecan-2 and uses thereof
US11918687B2 (en) 2016-01-15 2024-03-05 Orbsen Therapeutics Limited SDC-2 exosome compositions and methods of isolation and use

Families Citing this family (127)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7514074B2 (en) * 1997-07-14 2009-04-07 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
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
MXPA03005014A (en) 2000-12-06 2004-09-10 Robert J Hariri Method of collecting placental stem cells.
AUPR298901A0 (en) 2001-02-07 2001-03-08 McComb Foundation, Inc., The Cell suspension preparation technique and device
MXPA03007175A (en) * 2001-02-14 2005-02-14 Anthrogenesis Corp Post-partum mammalian placenta, its use and placental stem cells therefrom.
KR100973615B1 (en) 2001-02-14 2010-08-02 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
US9969980B2 (en) 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
ES2385450T3 (en) * 2004-03-22 2012-07-25 Osiris Therapeutics, Inc. Mesenchymal stem cells and their uses
US20080095749A1 (en) * 2004-03-22 2008-04-24 Sudeepta Aggarwal Mesenchymal stem cells and uses therefor
CA2564679C (en) * 2004-03-22 2015-06-23 Osiris Therapeutics, Inc. Mesenchymal stem cells and uses therefor
KR20080105555A (en) * 2007-05-31 2008-12-04 인하대학교 산학협력단 Therapeutic agent or method for treating graft-versus-host disease using mesenchymal stem cells
SI1926813T2 (en) * 2005-09-23 2019-11-29 Tigenix S A U Cell populations having immunoregulatory activity, method for isolation and uses
WO2007079184A2 (en) 2005-12-29 2007-07-12 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
NZ595786A (en) 2005-12-29 2013-05-31 Anthrogenesis Corp Placental stem cell populations
MX2008008774A (en) 2006-01-13 2008-09-26 Osiris Therapeutics Inc Mesenchymal stem cells expressing tnf-î± receptor.
US20070178073A1 (en) * 2006-02-01 2007-08-02 Samsung Life Public Welfare Foundation Composition Comprising Separated or Proliferated Cells from Umbilical Cord Blood for Treating Developmental and/or Chronic Lung Disease
US20070207130A1 (en) * 2006-03-01 2007-09-06 Calvin Cao Stem cell therapy to treat symptoms of avian flu and other diseases
CA2650638A1 (en) * 2006-04-28 2007-11-08 Tulane University Health Sciences Center Methods for treating diabetes
US9598673B2 (en) * 2006-05-19 2017-03-21 Creative Medical Health Treatment of disc degenerative disease
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
GB0619500D0 (en) * 2006-10-03 2006-11-08 Univ Keele Treatment of fibrosis
WO2008100497A1 (en) 2007-02-12 2008-08-21 Anthrogenesis Corporation Hepatocytes and chondrocytes from adherent placental stem cells; and cd34+, cd45- placental stem cell-enriched cell populations
US8983570B2 (en) 2007-03-27 2015-03-17 Cardiovascular Biotherapeutics, Inc. Therapeutic angiogenesis for treatment of the spine
US11224635B2 (en) * 2007-03-27 2022-01-18 Venturis Thereuptics, Inc. Therapeutic angiogenesis for treatment of the spine and other tissues
US20080254005A1 (en) * 2007-04-06 2008-10-16 Medistem Labortories Stem Cell Therapy for the Treatment of Autism and Other Disorders
US20080260703A1 (en) * 2007-04-23 2008-10-23 Medistem Labortories Treatment of Insulin Resistance and Diabetes
EP2223696B1 (en) 2007-05-24 2013-07-10 Apceth GmbH & Co. KG Mesenchymal CD34- stem cells for use in wound healing gene therapy
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
CA2697265A1 (en) 2007-08-09 2009-02-19 Genzyme Corporation Method of treating autoimmune disease with mesenchymal stem cells
US20110262402A1 (en) * 2007-09-07 2011-10-27 Masahiko Kuroda Therapeutic and prophylactic agents for arthritis
CN101855549A (en) * 2007-09-12 2010-10-06 耶达研究及发展有限公司 Methods of treating tumors in immune-privileged sites
PT2200622E (en) 2007-09-19 2012-11-06 Pluristem Ltd Adherent cells from adipose or placenta tissues and use thereof in therapy
DK2203176T3 (en) 2007-09-28 2015-02-09 Anthrogenesis Corp Tumor suppression with human placental perfusate and human natural killer cells of an intermediate product from placenta
WO2009046377A2 (en) * 2007-10-04 2009-04-09 Medistem Laboratories, Inc. Compositions and methods of stem cell therapy for autism
KR20160003307A (en) 2007-10-17 2016-01-08 티엑셀 Tr1 cells, mesenchymal stem cells and uses thereof
WO2009065093A2 (en) * 2007-11-17 2009-05-22 University Of Medicine And Dentistry Of New Jersey Use of stem cells for wound healing
AU2008340007B2 (en) * 2007-12-21 2013-10-17 Bone Therapeutics S.A. Human bone-forming cells in the treatment of inflammatory rheumatic diseases
US20110044958A1 (en) * 2008-03-14 2011-02-24 The Board Of Trustees Of The University Of Illinois Activated mesenchymal stem cells for the prevention and repair of inflammatory states
US9011840B2 (en) 2008-03-14 2015-04-21 The Board Of Trustees Of The University Of Illinois Activated mesenchymal stem cells for wound healing and impaired tissue regeneration
JP5541845B2 (en) 2008-03-28 2014-07-09 Jcrファーマ株式会社 Atopic dermatitis treatment
WO2009134429A2 (en) * 2008-05-02 2009-11-05 Massachusetts Institute Of Technology Methods and compositions for modulating immunological tolerance
AU2009252722A1 (en) 2008-05-27 2009-12-03 Pluristem Ltd. Methods of treating inflammatory colon diseases
AU2015268704B2 (en) * 2008-08-04 2017-08-10 Consejo Superior De Investigaciones Cientificas Uses of mesenchymal stem cells
GB0814249D0 (en) * 2008-08-04 2008-09-10 Cellerix Sa Uses of mesenchymal stem cells
NZ602248A (en) * 2008-08-14 2014-03-28 Osiris Therapeutics Inc Purified mesenchymal stem cell compositions and methods of purifying mesenchymal stem cell compositions
AU2009283217B2 (en) 2008-08-20 2015-09-17 Celularity Inc. Treatment of stroke using isolated placental cells
CN102186338B (en) 2008-08-20 2016-03-16 人类起源公司 The cell composition improved and prepare the method for described composition
CA2734446C (en) 2008-08-22 2017-06-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
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
NZ602455A (en) 2008-11-19 2014-03-28 Anthrogenesis Corp Amnion derived adherent cells
US9192695B2 (en) 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
US8309343B2 (en) 2008-12-01 2012-11-13 Baxter International Inc. Apparatus and method for processing biological material
US10098333B2 (en) * 2008-12-09 2018-10-16 University Of Southern California Method for treating an SLE-like autoimmune disease in a human subject consisting of administering stem cells from human exfoliated deciduous teeth (SHED) and erythropoietin (EPO) to said human subject
JP2012531916A (en) 2009-07-02 2012-12-13 アンソロジェネシス コーポレーション Method for producing red blood cells without using feeder cells
US8932852B2 (en) 2009-08-14 2015-01-13 Case Western Reserve University Compositions and methods of treating inflammatory bowel disease
WO2011047345A2 (en) * 2009-10-15 2011-04-21 Tai June Yoo Methods of treating diseases of conditions using mesenchymal stem cells
KR101301262B1 (en) * 2009-10-23 2013-08-27 가톨릭대학교 산학협력단 Mesenchymal stem cell introduced with a nucleotide sequence encoding TGFβ and use thereof
CN102048756B (en) * 2009-11-04 2014-02-19 中国医学科学院基础医学研究所 Use of human fat-derived mesenchymal stem cells in treatment of diseases in kidney and ocular fundus
WO2011062584A1 (en) * 2009-11-19 2011-05-26 Regents Of The University Of Minnesota Reducing inflammation using cell therapy
AU2010341703B2 (en) * 2009-12-08 2016-01-21 The Board Of Trustees Of The University Of Illinois Stem cell immune modulation methods of use and apparatus
EP2529007B1 (en) 2010-01-26 2017-07-12 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
AU2011220721B2 (en) * 2010-02-25 2015-02-05 Abt Holding Company Modulation of macrophage activation
TWI578993B (en) 2010-04-07 2017-04-21 安瑟吉納西斯公司 Angiogenesis using placental stem cells
WO2011127113A1 (en) 2010-04-08 2011-10-13 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
WO2011147967A1 (en) * 2010-05-27 2011-12-01 Georg Duda Skeletal muscle regeneration using mesenchymal stem cells
US20120020931A1 (en) * 2010-06-02 2012-01-26 Rutgers, The State University Of New Jersey Therapeutic encapsulated embryonic stem cells and mesenchymal stromal cells
ES2666746T3 (en) 2010-07-13 2018-05-07 Anthrogenesis Corporation Methods to generate natural cytolytic lymphocytes
WO2012009581A2 (en) * 2010-07-14 2012-01-19 University Of Southern California Pharmaceutical compositions comprising gingiva-derived mesenchymal stem cells and methods of treating inflammation, wound healing and contact hypersensitivity
JP2013536860A (en) * 2010-08-31 2013-09-26 クック・ジェネラル・バイオテクノロジー・エルエルシー Systemic, allogeneic stem cell therapy for the treatment of animal diseases
WO2012048093A2 (en) * 2010-10-08 2012-04-12 Osiris Therapeutics, Inc. Enhanced msc preparations
WO2012092485A1 (en) 2010-12-31 2012-07-05 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
US20140154274A1 (en) * 2011-01-12 2014-06-05 Tigenix Nv Methods and compositions for use in intralymphatic cellular therapies
JP2014506453A (en) 2011-01-19 2014-03-17 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 Innate pluripotent somatic cells
US20120288480A1 (en) * 2011-02-22 2012-11-15 Taipei Medical University Cell population comprising orbital fat-derived stem cells (ofscs) and their isolation and applications
EP3443968A1 (en) 2011-06-01 2019-02-20 Celularity, Inc. Treatment of pain using placental stem cells
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
WO2013082667A1 (en) 2011-12-09 2013-06-13 Metabolic Pharmaceuticals Pty Ltd Use of growth hormone fragments
CN102552880A (en) * 2011-12-14 2012-07-11 南方医科大学 Biological preparation for promoting skin wound to be healed
CN103191154B (en) * 2012-01-06 2015-09-09 上海交通大学医学院 Mesenchymal stem cells and the application of extracting method in the psoriatic medicine of preparation thereof
BR112014023272B1 (en) * 2012-03-22 2022-06-07 Avita Medical Ltd Method for preparing cells for cosmetic use in an epithelium-related procedure
WO2013146992A1 (en) 2012-03-29 2013-10-03 日本ケミカルリサーチ株式会社 Method for producing pluripotent stem cells derived from dental pulp
CN103585177A (en) * 2012-08-13 2014-02-19 首都医科大学附属北京口腔医院 Applications of mesenchymal stem cell and genetically modified mesenchymal stem cell
WO2014100857A1 (en) * 2012-12-24 2014-07-03 Cell Ideas Pty Ltd Vaccines for the treatment of cancer and compositions for enhancing vaccine efficacy
CN105142651A (en) 2013-02-05 2015-12-09 人类起源公司 Natural killer cells from placenta
CN105051186B (en) 2013-02-12 2018-11-02 复制细胞生命科学公司 Treatment and the composition and method for repairing tendon
US20140271568A1 (en) 2013-03-12 2014-09-18 Hawking Biological Technology Co., Ltd Method and kit for providing an increased expression of telomerase, brain-derived neurotrophic factor, stromal cell-derived factor-1, cxc chemokine receptor 4, and/or immune regulatory factor of stem cell
WO2014143383A1 (en) 2013-03-13 2014-09-18 Agilent Technologies, Inc. Transposome tethered to a gene delivery vehicle
AU2013205148B2 (en) 2013-03-14 2014-10-30 AVITA Medical Americas, LLC Systems and methods for tissue processing and preparation of cell suspension therefrom
WO2014150784A1 (en) 2013-03-15 2014-09-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
PL2972368T3 (en) * 2013-03-15 2020-03-31 Tigenix, S.A.U. Lymphocyte biomarkers for determining the clinical response to cell therapy
CN104138391B (en) * 2013-05-08 2018-07-31 中国科学院上海生命科学研究院 Application of the mescenchymal stem cell in preventing or treating immunity degradation caused by stress reaction
US20160129531A1 (en) * 2014-01-13 2016-05-12 Camarc Llc Electrode of a welding torch
TW201538730A (en) 2014-02-12 2015-10-16 Replicel Life Sciences Inc Compositions and methods for treating bone, joints and cartilage
MX2017000142A (en) * 2014-06-30 2017-07-28 Tigenix S A U Mesenchymal stromal cells for treating sepsis.
JP6452107B2 (en) 2014-09-05 2019-01-16 国立大学法人 東京大学 Pluripotent stem cells for the treatment of diabetic skin ulcers
TWI625391B (en) 2014-09-17 2018-06-01 國璽幹細胞應用技術股份有限公司 Uses of ligustilide
US20160095885A1 (en) 2014-10-01 2016-04-07 WibiWorks Therapeutics, Inc. Induction Medium & Methods for Stem Cell Culture & Therapy
CN104357383A (en) * 2014-10-11 2015-02-18 张炳强 Preparation method of human adipose-derived MSCs (mesenchymal stem cells) and application of human adipose-derived mesenchymal stem cell in preparation of medicine for treating diseases
RU2644650C2 (en) 2014-12-01 2018-02-13 Общество с ограниченной ответственностью "Т-Хелпер Клеточные Технологии" Stem cell material and method for its reception
US20180099009A1 (en) * 2015-03-20 2018-04-12 Rediens, Inc. Bony Tissue Delivery Systems and Methods
CA2983950A1 (en) * 2015-04-24 2016-10-27 Tigenix S.A.U. Biomarkers for determining the clinical response to cell therapy
JP6992983B2 (en) 2015-09-15 2022-01-13 カンステム バイオテック カンパニー リミテッド A composition for the prevention or treatment of inflammatory diseases containing stem cells overexpressing SOD3 as an active ingredient.
GB201604304D0 (en) 2016-03-14 2016-04-27 Tigenix S A U Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in crohn's disease
RU2756561C2 (en) * 2016-03-16 2021-10-01 Сината Терапьютикс Лимитед Colony formation medium and its application
RU2708329C2 (en) 2016-05-31 2019-12-05 Общество с ограниченной ответственностью "Т-Хелпер Клеточные Технологии" Stem cell material, compositions and methods of use
WO2017221879A1 (en) * 2016-06-20 2017-12-28 富士フイルム株式会社 Trophic factor release agent and inflammatory disease treatment agent
KR101816246B1 (en) * 2016-09-07 2018-01-08 에스씨엠생명과학 주식회사 Pharmaceutical composition for prevention or treatment of immune diseases and inflammatory diseases comprising immunogenic stimulated mesenchymal stem cells
JP7105195B2 (en) 2016-11-15 2022-07-22 株式会社カネカ Cell population containing mesenchymal stem cells derived from fetal appendages, method for producing the same, and pharmaceutical composition
KR20200104300A (en) 2017-12-22 2020-09-03 키에시 파르마슈티시 엣스. 피. 에이. Mesenchymal stromal cells and method for obtaining mesenchymal stromal cells from umbilical cord
CN111527198A (en) 2017-12-28 2020-08-11 株式会社钟化 Cell population comprising adherent stem cells, method for producing same, and pharmaceutical composition
JP7217533B2 (en) * 2018-01-24 2023-02-03 学校法人順天堂大学 Compositions for amplifying the effect of treatment with mesenchymal stem cells
EP3787646A4 (en) * 2018-05-04 2021-12-15 Spinalcyte, LLC Intradiscal t-regulatory cell administration for treatment of disc degenerative disease
CN108646035B (en) * 2018-05-18 2021-06-11 北京亦科诺生物科技有限公司 Novel use of insulin-like growth factor 1
CA3100471A1 (en) * 2018-06-05 2019-12-12 Medipost Co., Ltd. Pharmaceutical composition comprising mesenchymal stem cells as effective ingredient for prevention or treatment of inflammatory disease
WO2019240962A1 (en) * 2018-06-14 2019-12-19 Abraham J And Phyllis Katz Cord Blood Foundation Isolation of mesenchymal stromal cells from umbilical cord blood
EP3862016B1 (en) * 2018-10-04 2024-04-24 National Institute for Materials Science Angiogenesis promoter for use in treating a peripheral artery disease
EP3918341A4 (en) * 2019-01-31 2023-09-13 Primegen Biotech LLC Treatment of atopic dermatitis using mesenchymal stem cells and immune modulation
CA3132414A1 (en) * 2019-03-12 2020-09-17 Global Stem Cell Technology Immunomodulating mesenchymal stem cells
US20220160777A1 (en) * 2019-03-19 2022-05-26 Scm Lifescience Co., Ltd. Method for treating atopic dermatitis using monoclonal stem cells
RU2704322C1 (en) * 2019-06-11 2019-10-28 Федеральное государственное автономное образовательное учреждение высшего образования "Белгородский государственный национальный исследовательский университет" (НИУ "БелГУ") Cream with secretory of multipotent mesenchymal stromal cells for correction of psoriasiform inflammation in experiment
US20220233600A1 (en) 2019-06-14 2022-07-28 Kaneka Corporation Cell population comprising mesenchymal cells, pharmaceutical composition comprising the same, and method for producing the same
US20230293589A1 (en) * 2020-03-05 2023-09-21 Mesoblast International Sárl Method for treating inflammatory lung diseases using mesenchymal lineage precursor or stem cells
CN111557952A (en) * 2020-05-28 2020-08-21 澳门大学 Application of mesenchymal stem cells in preparation of preparation for promoting fat transplantation
CN111979186B (en) * 2020-08-21 2022-04-08 遵义医科大学附属医院 Method for rapidly and efficiently amplifying human mesenchymal stem cells in vitro and application
CN112076217A (en) * 2020-10-26 2020-12-15 重庆医科大学附属第一医院 Preparation method of anti-inflammatory oxidized lipid composition derived from mesenchymal stem cells
CN114736852A (en) * 2022-06-13 2022-07-12 南京艾尔普再生医学科技有限公司 In-vitro experimental model for myocardial cell transplantation immune rejection

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5700691A (en) * 1992-03-23 1997-12-23 Baxter Healthcare Inc. Method for the preparation of in vitro-derived human neutrophil precursor cells
US5843425A (en) * 1992-02-19 1998-12-01 The General Hospital Corporation Transplantation and graft-versus-host-disease
US6010696A (en) * 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US6077987A (en) * 1997-09-04 2000-06-20 North Shore-Long Island Jewish Research Institute Genetic engineering of cells to enhance healing and tissue regeneration
US6328960B1 (en) * 1998-03-18 2001-12-11 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US6355239B1 (en) * 1998-03-13 2002-03-12 Osiris Therapeutics, Inc. Uses for non-autologous mesenchymal stem cells
US20020037278A1 (en) * 2000-07-26 2002-03-28 Takafumi Ueno Therapeutic angiogenesis by bone marrow-derived cell transplantation in myocardial ischemic tissue and skeletal muscle ischemic tissue
US20020045260A1 (en) * 2000-10-17 2002-04-18 Shih-Chieh Hung Method of isolating mesenchymal stem cells
US20020044923A1 (en) * 1998-04-03 2002-04-18 Joseph D. Mosca Mesenchymal stem cells as immunosuppressants
US20020085996A1 (en) * 1998-03-18 2002-07-04 Mcintosh Kevin R. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US20020110544A1 (en) * 1998-05-13 2002-08-15 Victor M. Goldberg Osteoarthritis cartilage regeneration
US6497875B1 (en) * 1996-04-26 2002-12-24 Case Western Reserve University Multilayer skin or dermal equivalent having a layer containing mesenchymal stem cells
US20030049843A1 (en) * 2001-07-07 2003-03-13 Havenga Menzo Jans Emco Gene delivery vectors with cell type specificity for mesenchymal stem cells
US20030118567A1 (en) * 1999-03-26 2003-06-26 Stewart Duncan John Cell-based therapy for the pulmonary system
WO2004022579A2 (en) * 2002-09-06 2004-03-18 University Of South Florida Cellular delivery of natriuretic peptides
US20040166097A1 (en) * 1995-03-28 2004-08-26 Thomas Jefferson University Isolated stromal cells and methods of using the same
US20050093044A1 (en) * 2003-10-29 2005-05-05 Kangguo Cheng DRAM cell with buried collar and self-aligned buried strap
US6936281B2 (en) * 2001-03-21 2005-08-30 University Of South Florida Human mesenchymal progenitor cell
US20050239897A1 (en) * 2004-03-22 2005-10-27 Pittenger Mark F Mesenchymal stem cells and uses therefor
US20060112365A1 (en) * 2004-11-19 2006-05-25 Fujitsu Limited Design support apparatus, design support program and design support method for supporting design of semiconductor integrated circuit
US20070084354A1 (en) * 2005-10-14 2007-04-19 Hsiao Ming Folding grill

Family Cites Families (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH0651641B2 (en) * 1983-08-29 1994-07-06 株式会社ミドリ十字 Gamma interferon composition
JPS6048933U (en) 1983-09-12 1985-04-06 株式会社大隈鐵工所 Splash-proof cover on top of grinder bed
WO1987005518A1 (en) 1986-03-17 1987-09-24 Schering Corporation Treatment of cancers with gamma interferon
US5382514A (en) 1992-03-31 1995-01-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services In vivo angiogenesis assay
US6653134B2 (en) 1995-03-28 2003-11-25 Cp Hahnemann University Isolated stromal cells for use in the treatment of diseases of the central nervous system
ES2266715T3 (en) 1995-03-28 2007-03-01 Thomas Jefferson University IMPLANTS THAT INCLUDE IMMUNOLOGICALLY ISOLATED THERMAL CELLS AND ITS USE.
US6165785A (en) 1996-05-24 2000-12-26 University Of Cincinnati Bone marrow cultures for developing suppressor and stimulator cells for research and therapeutic applications
CA2271758A1 (en) * 1996-11-15 1998-05-22 Osiris Therapeutics, Inc. Msc-megakaryocyte precursor composition and method of isolating mscs associated with isolated megakaryocytes by isolating megakaryocytes
US20030103951A1 (en) 1997-07-14 2003-06-05 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
WO1999051247A1 (en) * 1998-04-03 1999-10-14 Osiris Therapeutics, Inc. Use of human mesenchymal stem cells to induce t-cell apoptosis
AU5545499A (en) * 1998-07-31 2000-02-21 Genzyme Corporation Improvement of cardiac function by mesenchymal stem cell transplantation
AU2998300A (en) * 1999-02-17 2000-09-04 United States Surgical Corporation Genetically altered mesenchymal stem cells and methods of use thereof
US20050153442A1 (en) * 1999-03-10 2005-07-14 Adam Katz Adipose-derived stem cells and lattices
EP1165830B1 (en) * 1999-03-10 2017-07-12 University of Pittsburgh of the Commonwealth System of Higher Education Adipose-derived stem cells and lattices
US8017112B2 (en) 1999-05-14 2011-09-13 Henry Ford Health System Transplantation of bone marrow stromal cells for treatment of neurodegenerative diseases
US6685936B2 (en) 1999-10-12 2004-02-03 Osiris Therapeutics, Inc. Suppressor cells induced by culture with mesenchymal stem cells for treatment of immune responses in transplantation
JP2003520254A (en) * 2000-01-19 2003-07-02 エス. ジル,パーカッシュ Methods and compositions relating to antisense VEGF oligonucleotides
JP2004505886A (en) 2000-02-18 2004-02-26 スィーヴィー セラピューティクス インコーポレイテッド Partial fatty acid oxidation inhibitors in the treatment of congestive heart failure
WO2001062901A2 (en) 2000-02-26 2001-08-30 Artecel Sciences, Inc. Pleuripotent stem cells generated from adipose tissue-derived stromal cells and uses thereof
PT1276486E (en) * 2000-04-25 2011-02-07 Osiris Therapeutics Inc Joint repair using mesenchymal stem cells
US20040022787A1 (en) 2000-07-03 2004-02-05 Robert Cohen Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
CA2438047A1 (en) * 2001-02-14 2002-08-22 Hildegard M. Kramer Biocompatible fleece for hemostasis and tissue engineering
CN100516204C (en) 2001-04-24 2009-07-22 味之素株式会社 Stem cells and method of separating the same
US6549704B2 (en) 2001-06-26 2003-04-15 Corning Incorporated Fabrication of microlensed fiber using doped silicon dioxide
EP1279738A1 (en) * 2001-07-06 2003-01-29 Crucell Holland B.V. Gene delivery vectors with cell type specificity for mesenchymal stem cells
WO2003010305A1 (en) * 2001-07-27 2003-02-06 Arhus Amt Immortilized stem cells
WO2003018077A1 (en) * 2001-08-22 2003-03-06 National Institute Of Advanced Industrial Science And Technology Method of bone regeneration
US20030104997A1 (en) 2001-09-05 2003-06-05 Black Ira B. Multi-lineage directed induction of bone marrow stromal cell differentiation
CA2465950A1 (en) 2001-11-09 2003-05-15 Artecel Sciences, Inc. Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
US6939863B2 (en) 2002-01-04 2005-09-06 Wei-Jan Chen Prevention of atherosclerosis and restenosis
MXPA04006771A (en) 2002-01-14 2005-04-25 Ford Henry Health System Materials from bone marrow stromal cells for use in forming blood vessels and producing angiogenic and trophic factors.
JP2005514926A (en) 2002-01-14 2005-05-26 ザ・ボード・オブ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・イリノイ Novel mammalian multipotent stem cells and compositions, methods for their preparation and administration
US20030161815A1 (en) * 2002-02-12 2003-08-28 Intercytex Limited Cell delivery system
AU2003225791A1 (en) * 2002-03-15 2003-09-29 Department Of Veterans Affairs, Rehabilitation R And D Service Methods and compositions for directing cells to target organs
AUPS112802A0 (en) * 2002-03-15 2002-04-18 Monash University Methods of inducing differentiation of stem cells into a specific cell lineage
JP2005520511A (en) * 2002-03-18 2005-07-14 ガミダ セル リミテッド Methods for inducing differentiation in ex vivo expanded stem cells
JP4554940B2 (en) * 2002-04-03 2010-09-29 直秀 山下 Medicament containing mesenchymal cells derived from human placenta and method for producing VEGF using the cells
ITTO20020311A1 (en) 2002-04-10 2003-10-10 Medestea Int Spa PROCEDURE FOR THE PREPARATION OF STEM CELLS FROM MUSCLE FABRIC AND HUMAN FAT FABRIC AND STEM CELLS OBTAINABLE BY T
US7498171B2 (en) * 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
CN1187058C (en) * 2002-04-16 2005-02-02 中国医学科学院血液学研究所泰达生命科学技术研究中心 Stem cell medicine for repairing damage of central nerve and its preparing process
US7628988B2 (en) 2002-06-27 2009-12-08 The General Hospital Corporation Methods and compositions for treating type 1 diabetes
US20040009155A1 (en) * 2002-07-12 2004-01-15 Maria Palasis Method for sustaining direct cell delivery
US20050249731A1 (en) * 2002-07-16 2005-11-10 Hadi Aslan Methods of implating mesenchymal stem cells for tissue repair and formation
EP1545219A4 (en) 2002-07-23 2009-09-30 Boston Scient Ltd Cell therapy for regeneration
CA2494040A1 (en) * 2002-07-29 2004-02-05 Es Cell International Pte Ltd. Multi-step method for the differentiation of insulin positive, glucose
JP3897343B2 (en) * 2002-08-01 2007-03-22 株式会社林原生物化学研究所 Protein that induces production of interferon-γ in immunocompetent cells
WO2004016779A1 (en) * 2002-08-17 2004-02-26 Hae-Young Suh A method for transdifferentiating mesenchymal stem cells into neuronal cells
KR20040022134A (en) * 2002-09-06 2004-03-11 송준석 Method for anticancer treatment using human mesenhymal stem cell and adenovirus producing p53
WO2004052177A2 (en) 2002-12-05 2004-06-24 Case Western Reserve University Cell-based therapies for ischemia
US20060263336A1 (en) * 2003-03-24 2006-11-23 Caplan Arnold I Cell targeting methods and compositions
CN1536075A (en) * 2003-04-09 2004-10-13 中国人民解放军军事医学科学院野战输 Method for inducing bone marrow mesenchymal stem cells to differentiate into insulin-like cells
ES2597837T3 (en) * 2003-06-27 2017-01-23 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of manufacturing and using them
CA2535029C (en) * 2003-08-07 2013-07-16 Healor Ltd. Pharmaceutical compositions and methods for accelerating wound healing
WO2005050904A1 (en) * 2003-11-20 2005-06-02 Advantest Corporation Clock recovery circuit and communication device
US20080095749A1 (en) 2004-03-22 2008-04-24 Sudeepta Aggarwal Mesenchymal stem cells and uses therefor
US7405186B2 (en) 2005-03-25 2008-07-29 Chemsil Silicones, Inc. Lubricant compositions, condom products and methods of making same
WO2006112365A1 (en) 2005-04-14 2006-10-26 Japan Health Sciences Foundation Treatment of pulmonary emphysema with a mesenchymal stem cell
US20070253931A1 (en) 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
MX2008008774A (en) 2006-01-13 2008-09-26 Osiris Therapeutics Inc Mesenchymal stem cells expressing tnf-î± receptor.
WO2007124594A1 (en) 2006-04-27 2007-11-08 Cell Therapy Technologies, Inc. Et Al. Stem cells for treating lung diseases
RU2416179C2 (en) 2006-10-06 2011-04-10 Конинклейке Филипс Электроникс Н.В. Apparatus for supplying luminous elements with energy and method of powering luminous elements
CA2674028C (en) 2008-07-25 2012-09-04 Her Majesty The Queen In Right Of Canada As Represented By The Minister Of National Defence Recombinant b. pseudomallei adhesin protein and methods and uses thereof
NZ602248A (en) 2008-08-14 2014-03-28 Osiris Therapeutics Inc Purified mesenchymal stem cell compositions and methods of purifying mesenchymal stem cell compositions
WO2012048093A2 (en) 2010-10-08 2012-04-12 Osiris Therapeutics, Inc. Enhanced msc preparations

Patent Citations (23)

* 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
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5843425A (en) * 1992-02-19 1998-12-01 The General Hospital Corporation Transplantation and graft-versus-host-disease
US5700691A (en) * 1992-03-23 1997-12-23 Baxter Healthcare Inc. Method for the preparation of in vitro-derived human neutrophil precursor cells
US20040166097A1 (en) * 1995-03-28 2004-08-26 Thomas Jefferson University Isolated stromal cells and methods of using the same
US6497875B1 (en) * 1996-04-26 2002-12-24 Case Western Reserve University Multilayer skin or dermal equivalent having a layer containing mesenchymal stem cells
US6077987A (en) * 1997-09-04 2000-06-20 North Shore-Long Island Jewish Research Institute Genetic engineering of cells to enhance healing and tissue regeneration
US20020064519A1 (en) * 1998-03-13 2002-05-30 Bruder Scott P. Uses for non-autologous mesenchymal stem cells
US6355239B1 (en) * 1998-03-13 2002-03-12 Osiris Therapeutics, Inc. Uses for non-autologous mesenchymal stem cells
US6328960B1 (en) * 1998-03-18 2001-12-11 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US20020085996A1 (en) * 1998-03-18 2002-07-04 Mcintosh Kevin R. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US20020044923A1 (en) * 1998-04-03 2002-04-18 Joseph D. Mosca Mesenchymal stem cells as immunosuppressants
US20020110544A1 (en) * 1998-05-13 2002-08-15 Victor M. Goldberg Osteoarthritis cartilage regeneration
US20030118567A1 (en) * 1999-03-26 2003-06-26 Stewart Duncan John Cell-based therapy for the pulmonary system
US20020037278A1 (en) * 2000-07-26 2002-03-28 Takafumi Ueno Therapeutic angiogenesis by bone marrow-derived cell transplantation in myocardial ischemic tissue and skeletal muscle ischemic tissue
US20020045260A1 (en) * 2000-10-17 2002-04-18 Shih-Chieh Hung Method of isolating mesenchymal stem cells
US6936281B2 (en) * 2001-03-21 2005-08-30 University Of South Florida Human mesenchymal progenitor cell
US20030049843A1 (en) * 2001-07-07 2003-03-13 Havenga Menzo Jans Emco Gene delivery vectors with cell type specificity for mesenchymal stem cells
WO2004022579A2 (en) * 2002-09-06 2004-03-18 University Of South Florida Cellular delivery of natriuretic peptides
US20050093044A1 (en) * 2003-10-29 2005-05-05 Kangguo Cheng DRAM cell with buried collar and self-aligned buried strap
US20050239897A1 (en) * 2004-03-22 2005-10-27 Pittenger Mark F Mesenchymal stem cells and uses therefor
US20060112365A1 (en) * 2004-11-19 2006-05-25 Fujitsu Limited Design support apparatus, design support program and design support method for supporting design of semiconductor integrated circuit
US20070084354A1 (en) * 2005-10-14 2007-04-19 Hsiao Ming Folding grill

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Gotherstrom et al., 2003, Bone Marrow transplantation 32: 265-272. *
Mangi et al 2003 Aug, Nature Medicine 9:1195-1201. *
Minguel., 2001, Exp. Biol. Med. 226:507-520. *

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8753883B2 (en) 2002-02-13 2014-06-17 Anthrogenesis Corporation Treatment of psoriasis using placental stem cells
US10780132B2 (en) * 2004-08-25 2020-09-22 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US20170189452A1 (en) * 2004-08-25 2017-07-06 Tigenix, S.A.U. Use of adipose tissue-derived stromal stem cells in treating fistula
US10729726B2 (en) 2004-10-04 2020-08-04 Tigenix, S.A.U. Identification and isolation of multipotent cells from non-osteochondral mesenchymal tissue
US8216566B2 (en) 2005-10-13 2012-07-10 Anthrogenesis Corporation Treatment of multiple sclerosis using placental stem cells
US9539288B2 (en) 2005-10-13 2017-01-10 Anthrogenesis Corporation Immunomodulation using placental stem cells
US20100183571A1 (en) * 2005-10-13 2010-07-22 Anthrogenesis Corporation Treatment of multiple sclerosis using placental stem cells
US8895256B2 (en) 2005-10-13 2014-11-25 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8916146B2 (en) 2007-02-12 2014-12-23 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8343480B2 (en) 2008-11-14 2013-01-01 Howmedica Osteonics Corp. Administration of stem or progenitor cells to a joint to enhance recovery from joint surgery
US20100150888A1 (en) * 2008-11-14 2010-06-17 Howmedica Osteonics Corp. Cells for joint fluid
US20110020293A1 (en) * 2009-07-21 2011-01-27 Abt Holding Company Use of Stem Cells to Reduce Leukocyte Extravasation
US9937208B2 (en) 2010-05-12 2018-04-10 Abt Holding Company Modulation of splenocytes in cell therapy
US10758570B2 (en) 2010-05-12 2020-09-01 Abt Holding Company Modulation of splenocytes in cell therapy
US20170239295A1 (en) * 2011-02-18 2017-08-24 Catholic University Industry Academic Cooperation Foundation Cell therapy composition for preventing or treating immune disease comprising mesenchymal stem cells and immunoregulatory t-cells as active ingredient
US10792309B2 (en) * 2011-02-18 2020-10-06 Catholic University Industry Academic Cooperation Foundation Cell therapy composition for preventing or treating immune disease comprising mesenchymal stem cells and immunoregulatory t-cells as active ingredient
US8709401B2 (en) 2011-02-25 2014-04-29 Howmedica Osteonics Corp. Primed stem cells and uses thereof to treat inflammatory conditions in joints
US10907131B2 (en) 2012-02-10 2021-02-02 Orbsen Therapeutics Limited Stromal stem cells
US10920197B2 (en) * 2012-02-10 2021-02-16 Orbsen Therapeutics Limited Stromal stem cells
US11952590B2 (en) 2012-02-10 2024-04-09 Orbsen Therapeutics Limited Stromal stem cells
US11952589B2 (en) 2012-02-10 2024-04-09 Orbsen Therapeutics Limited Stromal stem cells
US11926848B2 (en) 2012-02-10 2024-03-12 Orbsen Therapeutics Limited Stromal stem cells
US11884936B2 (en) 2012-02-10 2024-01-30 Orbsen Therapeutics Limited Stromal stem cells
US11434471B2 (en) 2012-02-10 2022-09-06 Orbsen Therapeutics Limited Stromal stem cells
US11230700B2 (en) 2012-02-10 2022-01-25 Orbsen Therapeutics Limited Stromal stem cells
US11142747B2 (en) 2012-02-10 2021-10-12 Orbsen Therapeutics Limited Stromal stem cells
WO2014018230A3 (en) * 2012-07-25 2014-04-17 Albert Einstein College Of Medicine Of Yeshiva University Methods to isolate human mesenchymal stem cells
WO2014018230A2 (en) * 2012-07-25 2014-01-30 Albert Einstein College Of Medicine Of Yeshiva University Methods to isolate human mesenchymal stem cells
US20180327718A1 (en) * 2013-01-18 2018-11-15 Escape Therapeutics, Inc. Enhanced differentiation of mesenchymal stem cells
US20160130556A1 (en) * 2013-01-18 2016-05-12 Escape Therapeutics, Inc. Enhanced differentiation of mesenchymal stem cells
WO2014113704A3 (en) * 2013-01-18 2015-11-26 Escape Therapeutics, Inc. Enhanced differentiation of mesenchymal stem cells
US10053670B2 (en) * 2013-01-18 2018-08-21 Escape Therapeutics, Inc. Enhanced differentiation of mesenchymal stem cells
US9861660B2 (en) 2013-04-12 2018-01-09 Saverio LaFrancesca Organs for transplantation
US11071752B2 (en) 2013-04-12 2021-07-27 Abt Holding Company Organs for transplantation
WO2015066552A1 (en) * 2013-11-04 2015-05-07 Becton, Dickinson And Company Immunomodulatory potential of a multipotent stromal cell (msc) population
KR102301945B1 (en) 2013-11-04 2021-09-14 벡톤 디킨슨 앤드 컴퍼니 Immunomodulatory potential of a multipotent stromal cell (msc) population
KR20160079774A (en) * 2013-11-04 2016-07-06 벡톤 디킨슨 앤드 컴퍼니 Immunomodulatory potential of a multipotent stromal cell (msc) population
US9632096B2 (en) 2013-11-04 2017-04-25 Becton, Dickinson And Company Methods of assessing the immunomodulatory potential of a multipotent stromal cell (MSC) population, and systems and kits for practicing the same
US11903997B2 (en) 2015-03-20 2024-02-20 Orbsen Therapeutics Limited Modulators of syndecan-2 and uses thereof
US11918687B2 (en) 2016-01-15 2024-03-05 Orbsen Therapeutics Limited SDC-2 exosome compositions and methods of isolation and use
WO2017127123A1 (en) * 2016-01-21 2017-07-27 Abt Holding Company Stem cells for wound healing
US11918609B2 (en) 2016-01-21 2024-03-05 Abt Holding Company Stem cells for wound healing
US10967006B2 (en) 2016-01-21 2021-04-06 Abt Holding Company Stem cells for wound healing
US20180214489A1 (en) * 2017-01-27 2018-08-02 Neil Riordan Mesenchymal stem cells with enhanced efficacy in treatment of autoimmunity particularly rheumatoid arthritis
US10987381B2 (en) * 2017-01-27 2021-04-27 Neil Riordan Mesenchymal stem cells with enhanced efficacy in treatment of autoimmunity particularly rheumatoid arthritis
US11268067B2 (en) 2017-07-14 2022-03-08 Orbsen Therapeutics Limited Methods of isolation and use of CD39 stromal stem cells

Also Published As

Publication number Publication date
EP2298861A2 (en) 2011-03-23
CN101384702A (en) 2009-03-11
US10716814B2 (en) 2020-07-21
CA2564679C (en) 2015-06-23
US10668101B2 (en) 2020-06-02
US20110318315A1 (en) 2011-12-29
US20110142807A1 (en) 2011-06-16
US20110311496A1 (en) 2011-12-22
WO2005093044A1 (en) 2005-10-06
AU2009200019A1 (en) 2009-02-05
EP2298864A2 (en) 2011-03-23
US20190175657A1 (en) 2019-06-13
EP2243826A3 (en) 2013-11-06
US20150272997A1 (en) 2015-10-01
KR20090065478A (en) 2009-06-22
CA2659825A1 (en) 2008-04-12
EP2298864B1 (en) 2017-10-11
AU2005227295A1 (en) 2005-10-06
EP2824175A2 (en) 2015-01-14
US20050239897A1 (en) 2005-10-27
EP1974016A2 (en) 2008-10-01
EP2824175A3 (en) 2015-05-06
US20140328807A1 (en) 2014-11-06
US20140161776A1 (en) 2014-06-12
EP2298862B1 (en) 2017-08-30
EP2824174A1 (en) 2015-01-14
US20090208463A1 (en) 2009-08-20
EP1727892A4 (en) 2007-08-22
EP2298862A2 (en) 2011-03-23
EP2298863A3 (en) 2011-04-13
WO2008042174A2 (en) 2008-04-10
US20150017132A1 (en) 2015-01-15
EP1727892B1 (en) 2012-05-02
US20140154276A1 (en) 2014-06-05
EP2298862A3 (en) 2011-08-24
US11389484B2 (en) 2022-07-19
CA2564679A1 (en) 2005-10-06
EP2824175B2 (en) 2020-10-28
EP2298861A3 (en) 2011-11-30
US10729727B2 (en) 2020-08-04
EP2298863B1 (en) 2015-07-22
US20080213227A1 (en) 2008-09-04
US20090180997A1 (en) 2009-07-16
AU2009200019B2 (en) 2012-08-02
US9694035B2 (en) 2017-07-04
EP2298861B1 (en) 2017-09-13
WO2008042174A3 (en) 2008-06-19
US20110027238A1 (en) 2011-02-03
JP2010505764A (en) 2010-02-25
EP3461884A1 (en) 2019-04-03
EP2824174B1 (en) 2018-11-28
EP2298864A3 (en) 2011-11-23
US10828334B1 (en) 2020-11-10
US20190201447A1 (en) 2019-07-04
EP2824175B1 (en) 2017-11-29
US20200246387A1 (en) 2020-08-06
EP1974016A4 (en) 2009-04-01
US20200360442A1 (en) 2020-11-19
EP1727892A1 (en) 2006-12-06
EP2243826A2 (en) 2010-10-27
US10960025B2 (en) 2021-03-30
JP2007530543A (en) 2007-11-01
US9943547B2 (en) 2018-04-17
AU2010200916A1 (en) 2010-04-01
US20110318314A1 (en) 2011-12-29
EP2298863A2 (en) 2011-03-23
US20190240259A1 (en) 2019-08-08
US20230076630A1 (en) 2023-03-09

Similar Documents

Publication Publication Date Title
US10828334B1 (en) Mesenchymal stem cells and uses therefor
CA2868733A1 (en) Mesenchymal stem cells for treating inflammation
AU2023204145A1 (en) Mesenchymal stem cells and uses therefor

Legal Events

Date Code Title Description
AS Assignment

Owner name: OSIRIS THERAPEUTICS, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:AGGARWAL, SUDEEPTA;PITTENGER, MARK F.;SIGNING DATES FROM 20070813 TO 20070920;REEL/FRAME:024137/0243

AS Assignment

Owner name: MESOBLAST INTERNATIONAL S?RL, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:OSIRIS THERAPEUTICS, INC.;REEL/FRAME:031533/0828

Effective date: 20131010

Owner name: MESOBLAST INTERNATIONAL SARL, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:OSIRIS THERAPEUTICS, INC.;REEL/FRAME:031533/0828

Effective date: 20131010

STCB Information on status: application discontinuation

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