US20090169522A1 - Mesenchymal stem cells expressing tnf-a receptor - Google Patents

Mesenchymal stem cells expressing tnf-a receptor Download PDF

Info

Publication number
US20090169522A1
US20090169522A1 US12/091,391 US9139107A US2009169522A1 US 20090169522 A1 US20090169522 A1 US 20090169522A1 US 9139107 A US9139107 A US 9139107A US 2009169522 A1 US2009169522 A1 US 2009169522A1
Authority
US
United States
Prior art keywords
mesenchymal stem
stem cells
cells
tnfri
composition
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
US12/091,391
Inventor
Alla Danilkovitch
Diane Carter
Alicia Tyrell
Simon Bubnic
Michelle Marcelino
Rodney Monroy
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
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US12/091,391 priority Critical patent/US20090169522A1/en
Assigned to OSIRIS THERAPEUTICS, INCORPORATED reassignment OSIRIS THERAPEUTICS, INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MONROY, RODNEY, MARCELINO, MICHELLE, CARTER, DIANE, DANILKOVITCH, ALLA, TYRELL, ALICIA, BUBNIC, SIMON
Publication of US20090169522A1 publication Critical patent/US20090169522A1/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

    • 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
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/25Tumour necrosing factors [TNF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/525Tumor necrosis factor [TNF]

Definitions

  • This invention relates to mesenchymal stem cells. More particularly, this invention relates to mesenchymal stem cells which express tumor necrosis factor-alpha (TNF- ⁇ ) receptors, and in particular, the tumor necrosis factor-alpha (TNF- ⁇ acute over ( ⁇ ) ⁇ ) receptor Type I (TNFRI), in an amount of at least 13 pg/10 6 cells.
  • TNF- ⁇ tumor necrosis factor-alpha
  • TNFRI tumor necrosis factor-alpha
  • Such mesenchymal stem cells inhibit lymphocyte proliferation.
  • MSCs Mesenchymal stem cells
  • MSCs have been shown to provide effective feeder layers for expansion of hematopoietic stem cells (Eaves, et al., Ann. N.Y. Acad. Sci ., Vol. 938, pg.
  • MSCs may be useful in the repair 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.
  • MSCs can inhibit T-cell proliferation either in mixed lymphocyte cultures or by other stimuli such as antigens and mitogens (Di Nicola, et al., Blood , Vol. 99, pgs. 3638-3843 (2002); Tse, et al., Transplantation , Vol. 75, pgs. 389-397 (2003); Aggarwal, et al., Blood , Vol. 105, pgs. 1815-1822 (2005)).
  • MSCs decrease the secretion of pro-inflammatory cytokines, tumor necrosis factor- ⁇ (TNF- ⁇ ), and Interferon- ⁇ (IFN- ⁇ ), and simultaneously increase production of anti-inflammatory cytokines Interleukin-10 (IL-10) and Interleukin-4 (IL-4) by immune cells.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • IFN- ⁇ Interferon- ⁇
  • MSCs can be beneficial for treatment of immunological responses which occur in graft-versus-host disease (GVHD), solid organ transplantation, and autoimmune diseases such as multiple sclerosis and rheumatoid arthritis.
  • GVHD graft-versus-host disease
  • solid organ transplantation graft-versus-host disease
  • autoimmune diseases such as multiple sclerosis and rheumatoid arthritis.
  • TNF- ⁇ receptors are expressed on the surface of mesenchymal stem cells. Accumulated data indicate that TNF- ⁇ is an important regulator of mesenchymal stem cell function. Incubation of TNF- ⁇ with human mesenchymal stem cells in culture upregulates prostaglandin E2 (PGE 2 ) and keratinocyte growth factor (KGF) secretion, induces indoleamine 2,3 deoxygenase (IDO) enzyme activity and stimulates cell migration. TNF- ⁇ has been shown to be present at wound and inflammatory sites, especially in organs targeted by graft-versus-host disease. (Koide, et al., Transplantation , Vol. 64, pgs.
  • TNF- ⁇ receptors There are two types of TNF- ⁇ receptors, or TNFRs: Type I (TNFRI), also known as p55, and Type II (TNFRII), also known as p75.
  • TNFRI Type I
  • TNFRII Type II
  • Both types of TNF- ⁇ receptors are present on MSCs; however, TNFRI is the predominant type.
  • TNFRI is the predominant type.
  • FIG. 1 is a graph of the correlation between TNFRI expression and the ability of MSCs to inhibit PBMC proliferation in vitro;
  • FIG. 2 is a graph showing TNFRI expression by human mesenchymal stem cells stored at ⁇ 80° C., ⁇ 70° C., ⁇ 60° C., and ⁇ 50° C.;
  • FIG. 3 is a graph showing TNFRI expression and the ability to inhibit PBMC proliferation in vitro, of human mesenchymal stem cells stored at ⁇ 80° C. and ⁇ 50° C.;
  • FIG. 4 is a graph showing TNFRI expression by human mesenchymal stem cells stored at ⁇ 135° C. or below, and then thawed and kept at room temperature for 6, 8, 24, or 32 hours.
  • a composition comprising mesenchymal stem cells.
  • the mesenchymal stem cells express the TNF- ⁇ acute over ( ⁇ ) ⁇ receptor Type I (TNFRI) in an amount effective to inhibit the proliferation of lymphocytes.
  • TNFRI TNF- ⁇ acute over ( ⁇ ) ⁇ receptor Type I
  • the mesenchymal stem cells express TNFRI in an amount of at least 13 pg/10 6 cells.
  • the mesenchymal stem cells express TNFRI in an amount of at least 15 pg/10 6 cells.
  • the mesenchymal stem cells express TNFRI in an amount of at least 18 pg/10 6 cells.
  • mesenchymal stem cells which express the TNF- ⁇ acute over ( ⁇ ) ⁇ receptor Type I in an amount from at least 13 pg/10 6 cells inhibit the proliferation of lymphocytes.
  • mesenchymal stem cells are particularly useful in inhibiting immune responses, and more particularly such mesenchymal stem cells are useful in the treatment of graft-versus-host disease; solid organ transplant rejection such as, for example, heart transplant rejection, liver transplant rejection, pancreas transplant rejection, intestine transplant rejection, and kidney transplant rejection; and autoimmune diseases such as, for example, rheumatoid arthritis, multiple sclerosis, Type I diabetes, Crohn's disease, Guillain-Barré syndrome, lupus erythematosus, myasthenia gravis, optic neuritis, psoriasis, Graves' disease, Hashimoto's disease, Ord's thyroiditis, aplastic anemia, Reiter's syndrome, autoimmune hepatitis, primary biliary cirrhosis, antiphospholipid antibody syndrome, opsoclonus myoclonus syndrome, temporal arteritis, acute disseminated encephalomyelitis, Goodpasture's syndrome
  • the mesenchymal stem cells are obtained from a mammal.
  • the mammal may be a primate, including human and non-human primates.
  • the mesenchymal stem cells may be a homogeneous composition or may be a mixed cell population enriched in MSCs.
  • Homogeneous mesenchymal stem cell compositions may be obtained by culturing adherent marrow or periosteal cells, and the mesenchymal stem cells may be identified by specific cell surface markers which are identified with unique monoclonal antibodies.
  • a method for obtaining a cell population enriched in mesenchymal stem cells is described, for example, in U.S. Pat. No. 5,486,359.
  • Alternative sources for mesenchymal stem cells include, but are not limited to, blood, skin, cord blood, muscle, fat, bone, and perichondrium.
  • the amount of cellular TNF- ⁇ receptor, such as TNF- ⁇ receptor Type I, that is expressed in a culture of mesenchymal stem cells may be determined by methods known to those skilled in the art. Such methods include, but are not limited to, quantitative assays such as quantitative ELISA assays, for example. It is to be understood, however, that the scope of the present invention is not to be limited to any particular method for determining the amount of TNF- ⁇ receptor.
  • the amount of TNF- ⁇ receptor expressed by a culture of mesenchymal stem cells is determined by an ELISA assay.
  • a cell lysate from a culture of mesenchymal stem cells is added to a well of an ELISA plate.
  • the well may be coated with an antibody, either a monoclonal or a polyclonal antibody(ies), against the TNF- ⁇ receptor.
  • the well then is washed, and then contacted with an antibody, either a monoclonal or a polyclonal antibody(ies), against the TNF- ⁇ receptor.
  • the antibody is conjugated to an appropriate enzyme, such as horseradish peroxidase, for example.
  • the well then may be incubated, and then is washed after the incubation period.
  • the wells then are contacted with an appropriate substrate, such as one or more chromogens.
  • Chromogens which may be employed include, but are not limited to, hydrogen peroxide and tetramethylbenzidine. After the substrate(s) is (are) added, the well is incubated for an appropriate period of time.
  • a “stop” solution is added to the well in order to stop the reaction of the enzyme with the substrate(s).
  • the optical density (OD) of the sample then is measured.
  • the optical density of the sample is correlated to the optical densities of samples containing known amounts of TNF- ⁇ receptor in order to determine the amount of TNF- ⁇ receptor expressed by the culture of mesenchymal stem cells being tested.
  • the present invention provides for the selection of a population of mesenchymal stem cells which express TNF- ⁇ receptor Type I in an amount of at least 13 pg/10 6 cells.
  • Such selected mesenchymal stem cells then may be admixed with an appropriate pharmaceutical carrier for treatment of the diseases and disorders mentioned hereinabove.
  • the mesenchymal stem cells may be administered as a cell suspension including a pharmaceutically acceptable liquid medium for injection.
  • the mesenchymal stem cells of the present invention are administered to an animal in an amount effective to treat one or more of the above-mentioned diseases or disorders in the animal.
  • the animal may be a mammal, and the mammal may be a primate, including human and non-human primates.
  • the mesenchymal stem cells may be administered systemically, such as, for example, by intravenous, intraarterial, or intraperitoneal administration.
  • the exact dosage of mesenchymal stem cells to be administered is dependent upon a variety of factors, including, but not limited to, the age, weight, and sex of the patient, the disease(s) or disorder(s) being treated, and the extent and severity thereof.
  • hMSCs were transfected transiently by antisense TNFRI type oligonucleotides with the purpose to decrease TNFRI expression (Shen et al., J. Biol. Chem ., Vol. 272, pgs. 3550-3553 (1997)).
  • three different concentrations of oligonucleotides were used for transfection experiments. Non-transfected MSCs and MSCs transfected with a sense oligonucleotide were used as controls.
  • TNFRI expression on hMSCs was analyzed in cell lysates by ELISA, and effect of reduction in TNFRI expression on hMSC capacity to inhibit hPBMC proliferation in vitro was investigated.
  • Human bone marrow-derived MSCs at Passage 5 from 7 different donors were used for analysis.
  • Cells were obtained from bone marrow aspirates, and isolated using hespan. The cells then were cultured through Passage 5, and frozen in a standard cryopreservation solution containing 5% human serum albumin (HSA) and 10% dimethylsulfoxide in Plasmalyte A. (Baxter)
  • HSA human serum albumin
  • Plasmalyte A Plasmalyte A.
  • TNFRI sense or antisense oligonucleotides were transfected with TNFRI sense or antisense oligonucleotides at concentrations of 1.25, 2.5 and 5 pg/mL according to the transfection reagent manufacturer's protocol (Invitrogen, the Cellfectin transfection reagent product insert). At 24 hours post-transfection, the cells were collected from the plates. One group of cells was lysed, and expression of TNFRI in cell lysates was analyzed by ELISA according to the sTNFRI ELISA protocol (R&D Systems, product insert). TNFRI expression was expressed in pg of receptor per 1 ⁇ 10 6 cells.
  • This assay provides for the measurement of both soluble as well as cell-associated TNFRI (Qjwang, et al., Biochemistry , Vol. 36, pg. 6033 (1997).)
  • the assay employs the quantitative sandwich enzyme immunoassay technique.
  • the assay employs a microplate that includes wells that have been pre-coated with a monoclonal antibody specific for TNFRI. TNFRI present in calibrator samples, quality control samples, or samples of MSC cell lysates is captured by the immobilized TNFRI antibody.
  • enzyme-linked polyclonal antibodies specific for TNFRI is added to the wells.
  • a substrate solution was added to the wells, and color develops in proportion to the amount of bound TNFRI. The color development then is stopped, and the intensity of the color is measured using an ELISA reader.
  • assay diluent HD1-7 50 ⁇ l of assay diluent HD1-7, a buffered protein base with preservative, were added to the wells of an ELISA plate.
  • the wells were coated with a monoclonal antibody specific for TNFRI.
  • 200 ⁇ l of either calibrator samples containing 500 pg/ml, 250 pg/ml, 125 pg/ml, 62.5 pg/ml, 31.25 pg/ml, 15.625 pg/ml, or 7.813 pg/ml of soluble human TNFRI
  • quality control samples containing 45 pg/ml, 100 pg/ml, or 250 pg/ml of human TNFRI
  • cell lysates then were added to the wells.
  • the liquid then was decanted from each well by inverting the plate over a sink, and then the plate was washed three times. The plate is washed each time with 400 ⁇ l of a wash buffer added to each well. Residual liquid was removed by inverting the plate and blotting.
  • TNFRI expression data were expressed in picograms of receptor per 1 ⁇ 10 6 cells.
  • the raw data in pg/ml reflect TNFRI in picograms per 1 ⁇ 10 6 cells (2.5 ⁇ 10 5 cells were lysed in 250 ⁇ l of the lysis reagent, thus corresponding to 1 ⁇ 10 6 cells/ml).
  • TNFRI expression in pg per 1 ⁇ 10 6 cells for samples of mesenchymal stem cells from the donors was determined.
  • the mesenchymal stem cells from each donor were non-transfected, or transfected with a TNFRI sense or antisense oligonucleotide at a concentration of 1.25, 2.5, or 5 ⁇ g/ml.
  • the ELISA values and the amount of TNFRI expressed by each of the mesenchymal stem cell samples from each of the donors are given in Table 3 below.
  • the mean TNFRI expression in picograms per 1 ⁇ 10 6 cells, was determined for non-transfected (control) mesenchymal stem cells, as well as mesenchymal stem cells transfected with 1.25, 2.5, or 5 ⁇ l/ml of antisense or sense oligonucleotides.
  • the mean TNFRI expression values are given in Table 4 below.
  • TNFRI expression by hMSCs transfected with anti-sense and control (sense) oligonucleotides summary for 7 tested hMSC donors TNFRI expression in pg per 1 ⁇ 10 6 cells hMSC donor #: Mean for 486 13 24 007 14 15 23 7 donors SD Control (non- 41* 34 54 51 53 67 40 48.57 11.09 transfected cells) TNFRI anti-sense 7 10 11 11 9 13 14 10.71 2.36 oligo-transfected cells 5 ⁇ g/mL TNFRI anti-sense 21 20 32 32 26 35 28 27.71 5.74 oligo-transfected cells 2.5 ⁇ g/mL TNFRI anti-sense 28 27 42 41 40 53 32 37.57 9.22 oligo-transfected cells 1.25 ⁇ g/mL Control (sense) 20 20 35 28 33 37 27 28.57 6.85 oligo-transfected cells 5 ⁇ g/mL Control (sense) 38 30 49 36 43 47 32 39.29
  • PBMCs Human PBMCs from two different donors were used for this assay.
  • PBMCs were isolated from leukopheresed blood using Ficoll-Paque gradient centrifugation according to the manufacturer's protocol (Amersham Biosciences, Ficoll-Paque Plus product insert). Cells were stored frozen at ⁇ 80° C. in a medium including 90% FBS and 10% DMSO prior to analysis.
  • hPBMCs were thawed, counted and plated into 96-well tissue culture plates at 1 ⁇ 10 5 cells/well together with hMSCs (1 ⁇ 10 4 cells/well).
  • a combination of anti-CD3 (1 ⁇ g/mL) and anti-CD28 (1 pg/mL) antibodies was used to stimulate lymphocyte proliferation that represents an in vitro model for immune cell activation characteristics of GVHD and rejection of allogeneic organs.
  • the plates then were incubated in a humidified atmosphere containing 5% CO 2 .
  • the proliferation of PBMCs alone and in the presence of MSCs was measured at day 5 from culture initiation by the addition of [Methyl- 3 H]-thymidine at 1 ⁇ Ci/well for the final 18-20 hrs of culture.
  • the cells were transferred onto a glass filter using a 96-well plate harvester, and radioactivity incorporated into DNA was measured by a liquid scintillation beta-counter.
  • the uptake of [Methyl- 3 H]-thymidine into DNA in counts per minute (cpm) represents hPBMC proliferation.
  • Final results were expressed as % inhibition of PBMC proliferation in the presence of MSCs calculated as:
  • hMSCs with decreased expression of TNFR type I lose their ability to suppress hPBMC proliferation in vitro.
  • the data support the premise that the expression of TNFRI is an essential link to the suppression of PBMC proliferation by MSCs.
  • TNFRI can be used as a potency marker for MSC immunomodulative activity.
  • a potency threshold of 13.07 pg of TNFRI (mean ⁇ SD) per 1 ⁇ 10 6 cells correlates with less than 50% inhibition of hPBMC proliferation (Table 6, FIG. 1 ).
  • non-potent MSCs are cells expressing less than 13 pg TNFRI per 1 ⁇ 10 6 cells.
  • TNFRI is a Temperature-Sensitive Marker of hMSC Functionality
  • Ex vivo handling of mammalian cells is restricted by a number of factors including temperature. For example, low temperatures such as ⁇ 80 ⁇ 5° C., or lower, even as low as ⁇ 135° C. or below (liquid nitrogen) are required for cell storage whereas ex vivo cell expansion requires a temperature of 37 ⁇ 0.5° C. Cell exposure to temperatures outside of the optimal ranges may lead to a decrease in cell functionality or cell death. Mammalian cells are able to withstand short-term minor temperature fluctuations; however, each type of cells has its own temperature tolerance range for cell culture maintenance, shipping, and storage.
  • TNFRI expression correlates with hMSC immunosuppressive activity.
  • the level of TNFRI expression by hMSCs of less than 13 pg/10 6 cells has been determined as a threshold, below which hMSCs begin to lose their ability to suppress an immune response (See FIG. 1 ).
  • TNFRI expression is a marker of hMSC immunosuppression, an activity that is believed essential for MSCs to be efficacious for treatment of immunological reactions taking place in GVHD, organ rejection, autoimmune diseases, and other diseases.
  • effects of temperature fluctuations during storage of frozen hMSCs as well as the effect of time of exposure of cells to room temperature on expression of TNFRI on hMSCs was investigated.
  • the thawed and diluted hMSCs were kept at room temperature (22° C.-24° C.), and samples were taken and tested for the amount of TNFRI at 0 (immediately post-thaw-baseline), 6, 8, 10, 24, and 32 hours post-thawing.
  • the results showed that exposure of hMSCs to room temperature decreased the TNFRI expression level on the hMSCs ( FIG. 4 , bars represent mean ⁇ SD of the TNFRI expression level for 2 hMSC lots.
  • the solid line represents the TNFRI expression level of 13 pg/10 6 cells, which is the hMSC potency threshold).
  • the significant decrease in TNFRI expression was observed at 24 hours and 32 hours, and it correlated with a significant decrease in cell viability (below 20%, data not shown).
  • TNFRI expression by hMSCs is sensitive to temperature, and TNFRI can be used as a marker of functionality of hMSC that were exposed to non-optimal temperatures during storage, shipping or cell processing.

Abstract

Mesenchymal stem cells which express TNF-α receptor Type I in an amount of at least 13 pg/106 cells. Such mesenchymal stem cells inhibit the proliferation of lymphocytes and may be employed, in particular, in the treatment of graft-versus-host disease.

Description

  • This application claims priority based on application Ser. No. 60/759,157, filed Jan. 13, 2006, the contents of which are incorporated by reference in their entirety.
  • This invention relates to mesenchymal stem cells. More particularly, this invention relates to mesenchymal stem cells which express tumor necrosis factor-alpha (TNF-α) receptors, and in particular, the tumor necrosis factor-alpha (TNF-{acute over (α)}) receptor Type I (TNFRI), in an amount of at least 13 pg/106 cells. Such mesenchymal stem cells inhibit lymphocyte proliferation.
  • Mesenchymal stem cells (MSCs) are multipotent stem cells 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 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 may be useful in the repair 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, pgs. 601-604 (2001)). 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 (Horowitz, 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, pgs. 307-316 (2000)).
  • In addition, in vitro studies from different laboratories have shown that MSCs can inhibit T-cell proliferation either in mixed lymphocyte cultures or by other stimuli such as antigens and mitogens (Di Nicola, et al., Blood, Vol. 99, pgs. 3638-3843 (2002); Tse, et al., Transplantation, Vol. 75, pgs. 389-397 (2003); Aggarwal, et al., Blood, Vol. 105, pgs. 1815-1822 (2005)). Recent in vitro data demonstrate further that MSCs decrease the secretion of pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α), and Interferon-γ (IFN-γ), and simultaneously increase production of anti-inflammatory cytokines Interleukin-10 (IL-10) and Interleukin-4 (IL-4) by immune cells. (Aggarwal, 2005). These results indicate that due to immunomodulatory and anti-inflammatory activities, MSCs can be beneficial for treatment of immunological responses which occur in graft-versus-host disease (GVHD), solid organ transplantation, and autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. A clinical case report demonstrating the therapeutic effect of MSCs for acute GVHD supports strongly this hypothesis. (Le Blanc, et al., The Lancet, Vol. 363, pgs. 1439-1441 (2004).)
  • The TNF-α receptors are expressed on the surface of mesenchymal stem cells. Accumulated data indicate that TNF-α is an important regulator of mesenchymal stem cell function. Incubation of TNF-α with human mesenchymal stem cells in culture upregulates prostaglandin E2 (PGE2) and keratinocyte growth factor (KGF) secretion, induces indoleamine 2,3 deoxygenase (IDO) enzyme activity and stimulates cell migration. TNF-α has been shown to be present at wound and inflammatory sites, especially in organs targeted by graft-versus-host disease. (Koide, et al., Transplantation, Vol. 64, pgs. 518-524 (1997); Kuroiwa, et al., J. Clin. Invest., Vol. 107, pgs. 1365-1373 (2001); Deans, et al., Exp. Hematol., Vol. 28, pgs. 875-884 (2002); Ellison, et al., J. Clin. Immunol., Vol. 24, pgs. 197-211 (2004)). Thus, such data indicate that expression of TNF-α receptors by mesenchymal stem cells may be critical for immunosuppressive, immunomodulatory, anti-inflammatory, tissue-repairing, or wound-healing activities, as well as migration to sites of inflammation.
  • There are two types of TNF-α receptors, or TNFRs: Type I (TNFRI), also known as p55, and Type II (TNFRII), also known as p75. (Tartaglia, et al., Proc. Nat. Acad. Sci, Vol. 88, pgs. 9292-9296 (1991).) Both types of TNF-α receptors are present on MSCs; however, TNFRI is the predominant type. (Vancheri, et al., Am. J. Respir. Cell Mol. Biol., Vol. 22, pgs. 628-634 (2000); Debets, et al., Cytokine, Vol. 8, pgs. 80-88 (1996).)
  • The invention now will be described with respect to the drawings wherein:
  • FIG. 1 is a graph of the correlation between TNFRI expression and the ability of MSCs to inhibit PBMC proliferation in vitro;
  • FIG. 2 is a graph showing TNFRI expression by human mesenchymal stem cells stored at −80° C., −70° C., −60° C., and −50° C.;
  • FIG. 3 is a graph showing TNFRI expression and the ability to inhibit PBMC proliferation in vitro, of human mesenchymal stem cells stored at −80° C. and −50° C.; and
  • FIG. 4 is a graph showing TNFRI expression by human mesenchymal stem cells stored at −135° C. or below, and then thawed and kept at room temperature for 6, 8, 24, or 32 hours.
  • In accordance with an aspect of the present invention, there is provided a composition comprising mesenchymal stem cells. The mesenchymal stem cells express the TNF-{acute over (α)} receptor Type I (TNFRI) in an amount effective to inhibit the proliferation of lymphocytes. In one embodiment, the mesenchymal stem cells express TNFRI in an amount of at least 13 pg/106 cells. In another embodiment, the mesenchymal stem cells express TNFRI in an amount of at least 15 pg/106 cells. In yet another embodiment, the mesenchymal stem cells express TNFRI in an amount of at least 18 pg/106 cells.
  • Although the scope of the present invention is not to be limited to any theoretical reasoning, Applicants have found that mesenchymal stem cells which express the TNF-{acute over (α)} receptor Type I in an amount from at least 13 pg/106 cells inhibit the proliferation of lymphocytes. Such mesenchymal stem cells are particularly useful in inhibiting immune responses, and more particularly such mesenchymal stem cells are useful in the treatment of graft-versus-host disease; solid organ transplant rejection such as, for example, heart transplant rejection, liver transplant rejection, pancreas transplant rejection, intestine transplant rejection, and kidney transplant rejection; and autoimmune diseases such as, for example, rheumatoid arthritis, multiple sclerosis, Type I diabetes, Crohn's disease, Guillain-Barré syndrome, lupus erythematosus, myasthenia gravis, optic neuritis, psoriasis, Graves' disease, Hashimoto's disease, Ord's thyroiditis, aplastic anemia, Reiter's syndrome, autoimmune hepatitis, primary biliary cirrhosis, antiphospholipid antibody syndrome, opsoclonus myoclonus syndrome, temporal arteritis, acute disseminated encephalomyelitis, Goodpasture's syndrome, Wegener's granulomatosis, coeliac disease, pemphigus, polyarthritis, warm autoimmune hemolytic anemia, and scleroderma.
  • In one embodiment, the mesenchymal stem cells are obtained from a mammal. The mammal may be a primate, including human and non-human primates.
  • The mesenchymal stem cells may be a homogeneous composition or may be a mixed cell population enriched in MSCs. Homogeneous mesenchymal stem cell compositions may be obtained by culturing adherent marrow or periosteal cells, and the mesenchymal stem cells may be identified by specific cell surface markers which are identified with unique monoclonal antibodies. A method for obtaining a cell population enriched in mesenchymal stem cells is described, for example, in U.S. Pat. No. 5,486,359. Alternative sources for mesenchymal stem cells include, but are not limited to, blood, skin, cord blood, muscle, fat, bone, and perichondrium.
  • The amount of cellular TNF-α receptor, such as TNF-α receptor Type I, that is expressed in a culture of mesenchymal stem cells may be determined by methods known to those skilled in the art. Such methods include, but are not limited to, quantitative assays such as quantitative ELISA assays, for example. It is to be understood, however, that the scope of the present invention is not to be limited to any particular method for determining the amount of TNF-α receptor.
  • In one embodiment, the amount of TNF-α receptor expressed by a culture of mesenchymal stem cells is determined by an ELISA assay. In such an assay, a cell lysate from a culture of mesenchymal stem cells is added to a well of an ELISA plate. The well may be coated with an antibody, either a monoclonal or a polyclonal antibody(ies), against the TNF-α receptor. The well then is washed, and then contacted with an antibody, either a monoclonal or a polyclonal antibody(ies), against the TNF-α receptor. The antibody is conjugated to an appropriate enzyme, such as horseradish peroxidase, for example. The well then may be incubated, and then is washed after the incubation period. The wells then are contacted with an appropriate substrate, such as one or more chromogens. Chromogens which may be employed include, but are not limited to, hydrogen peroxide and tetramethylbenzidine. After the substrate(s) is (are) added, the well is incubated for an appropriate period of time.
  • Upon completion of the incubation, a “stop” solution is added to the well in order to stop the reaction of the enzyme with the substrate(s). The optical density (OD) of the sample then is measured. The optical density of the sample is correlated to the optical densities of samples containing known amounts of TNF-α receptor in order to determine the amount of TNF-α receptor expressed by the culture of mesenchymal stem cells being tested.
  • Thus, the present invention provides for the selection of a population of mesenchymal stem cells which express TNF-α receptor Type I in an amount of at least 13 pg/106 cells. Such selected mesenchymal stem cells then may be admixed with an appropriate pharmaceutical carrier for treatment of the diseases and disorders mentioned hereinabove. For example, the mesenchymal stem cells may be administered as a cell suspension including a pharmaceutically acceptable liquid medium for injection.
  • The mesenchymal stem cells of the present invention are administered to an animal in an amount effective to treat one or more of the above-mentioned diseases or disorders in the animal. The animal may be a mammal, and the mammal may be a primate, including human and non-human primates. The mesenchymal stem cells may be administered systemically, such as, for example, by intravenous, intraarterial, or intraperitoneal administration. The exact dosage of mesenchymal stem cells to be administered is dependent upon a variety of factors, including, but not limited to, the age, weight, and sex of the patient, the disease(s) or disorder(s) being treated, and the extent and severity thereof.
  • The invention now will be described with respect to the following examples; however, the scope of the present invention is not intended to be limited thereby.
  • EXAMPLE 1
  • In order to investigate the role of TNFRI on the immunosuppressive hMSC activity, hMSCs were transfected transiently by antisense TNFRI type oligonucleotides with the purpose to decrease TNFRI expression (Shen et al., J. Biol. Chem., Vol. 272, pgs. 3550-3553 (1997)). In order to reach different degrees of TNFRI expression inhibition, three different concentrations of oligonucleotides were used for transfection experiments. Non-transfected MSCs and MSCs transfected with a sense oligonucleotide were used as controls. TNFRI expression on hMSCs was analyzed in cell lysates by ELISA, and effect of reduction in TNFRI expression on hMSC capacity to inhibit hPBMC proliferation in vitro was investigated.
  • Human bone marrow-derived MSCs at Passage 5 from 7 different donors were used for analysis. Cells were obtained from bone marrow aspirates, and isolated using hespan. The cells then were cultured through Passage 5, and frozen in a standard cryopreservation solution containing 5% human serum albumin (HSA) and 10% dimethylsulfoxide in Plasmalyte A. (Baxter) The cells were stored at −80° C. prior to analysis. On the day of the experiment, the hMSCs were thawed, counted, and plated into 6-well tissue culture plates at 2.5×105 cells/well. After overnight incubation, cells were transfected with TNFRI sense or antisense oligonucleotides at concentrations of 1.25, 2.5 and 5 pg/mL according to the transfection reagent manufacturer's protocol (Invitrogen, the Cellfectin transfection reagent product insert). At 24 hours post-transfection, the cells were collected from the plates. One group of cells was lysed, and expression of TNFRI in cell lysates was analyzed by ELISA according to the sTNFRI ELISA protocol (R&D Systems, product insert). TNFRI expression was expressed in pg of receptor per 1×106 cells.
  • For the ELISA assay, 2.5×105 MSCs per well were lysed directly in wells using 250 μl/well of Cell Lytic-mammalian cell lysis/extraction reagent (Sigma, Catalog No. C-2978) containing a complete protein inhibitor cocktail (Roche). The cell lysates then were centrifuged for 10 minutes at 12,000-14,000 rpm in an Eppendorf centrifuge to remove insoluble material from the lysis buffer solution. The cell lysates then were collected in a new tube for use in the ELISA assay.
  • An alternative method of cell lysis, i.e., lysis of cell pellets in tubes, also was carried out for frozen cells and for cells collected from tissue culture plates or flasks. Both methods, direct cell lysis in culture plates and lysis of cell pellets in tubes, gave comparable results.
  • A commercially available ELISA kit, Quantikine®, Human sTNFRI (Catalog No. DRT 100, R&D Systems) was used for the detection of TNFRI in cell lysates. This assay provides for the measurement of both soluble as well as cell-associated TNFRI (Qjwang, et al., Biochemistry, Vol. 36, pg. 6033 (1997).) The assay employs the quantitative sandwich enzyme immunoassay technique. The assay employs a microplate that includes wells that have been pre-coated with a monoclonal antibody specific for TNFRI. TNFRI present in calibrator samples, quality control samples, or samples of MSC cell lysates is captured by the immobilized TNFRI antibody. After washing away any unbound substances, enzyme-linked polyclonal antibodies specific for TNFRI is added to the wells. Following a wash step to remove any unbound enzyme-linked antibody, a substrate solution was added to the wells, and color develops in proportion to the amount of bound TNFRI. The color development then is stopped, and the intensity of the color is measured using an ELISA reader.
  • The details of the ELISA are given hereinbelow.
  • 50 μl of assay diluent HD1-7, a buffered protein base with preservative, were added to the wells of an ELISA plate. The wells were coated with a monoclonal antibody specific for TNFRI. 200 μl of either calibrator samples (containing 500 pg/ml, 250 pg/ml, 125 pg/ml, 62.5 pg/ml, 31.25 pg/ml, 15.625 pg/ml, or 7.813 pg/ml of soluble human TNFRI), quality control samples (containing 45 pg/ml, 100 pg/ml, or 250 pg/ml of human TNFRI), or cell lysates then were added to the wells. Prior to the addition of the calibration and quality control sample to the wells, such samples were treated with the Cell Lytic-mammalian cell lysis extraction agent (Sigma) and complete protein inhibitor cocktail (Roche) as hereinabove described. The plate then was covered with an adhesive strip, and incubated for 2 hours ±10 minutes at room temperature.
  • The liquid then was decanted from each well by inverting the plate over a sink, and then the plate was washed three times. The plate is washed each time with 400 μl of a wash buffer added to each well. Residual liquid was removed by inverting the plate and blotting.
  • 200 μl of soluble TNFRI polyclonal antibodies conjugated to horseradish peroxidase then were added to each well. The plate then was incubated for 2 hours ±10 minutes at room temperature. The liquid then was decanted from each well, and each well was washed three times with 400 μl of wash buffer as hereinabove described.
  • 200 μl of a substrate solution of stabilized hydrogen peroxide and stabilized tetramethylbenzidine chromogen then were added to each well. The plate then was incubated for 20 minutes ±10 minutes at room temperature in the dark. 50 μl of a solution of 2N sulfuric acid then were added to each well. The optical density (OD) of each sample then was measured within 30 minutes with a 450 nm test and a 570 nm reference filter. The optical density values then were correlated to the amounts of TNFRI in the cell lysate samples.
  • Quantitation was achieved by comparing the signal from samples of MSC cell lysates to TNFRI standards assayed at the same time. Each ELISA run provided a calibration curve and included duplicate quality control samples plated in front and after test samples. Quality control samples were used for ELISA run validity assessment. TNFRI expression data were expressed in picograms of receptor per 1×106 cells. The raw data (in pg/ml) reflect TNFRI in picograms per 1×106 cells (2.5×105 cells were lysed in 250 μl of the lysis reagent, thus corresponding to 1×106 cells/ml).
  • The ELISA values for the calibration samples are given in Table 1 below.
  • TABLE 1
    Calculations for ELISA run calibration standards
    Theoretical Back
    Concentration Calculated Calculated Mean
    of OD Concentration Concentration
    Calibrator Calibratiors OD* Mean Standard for Standards for Standards
    Sample (pg/mL) Values Value Deviation (pg/mL) (pg/mL) % DFT* % CV*
    St01 500 2.431 2.437 0.008 498.003 499.923 −0.015 0.3
    2.443 501.842
    St02 250 1.487 1.476 0.016 252.746 250.306 0.123 1.1
    1.464 247.867
    St03 125 0.804 0.815 0.015 122.64 124.447 −0.442 1.8
    0.825 126.255
    St04 62.5 0.453 0.442 0.016 64.774 63.024 0.839 3.5
    0.431 61.274
    St05 31.25 0.25 0.239 0.016 32.749 30.939 −0.996 6.8
    0.227 29.128
    St06 15.625 0.143 0.145 0.002 15.765 16.007 2.446 1.5
    0.146 16.249
    St07 7.813 0.092 0.093 0.001 7.368 7.537 −3.528 1.5
    0.094 7.706
    *Note:
    OD—optical density;
    % DFT—% Difference from Theoretical;
    CV %—% Coefficient of Variance
  • The ELISA values for the quality control samples are given in Table 2 below.
  • TABLE 2
    Calculations for ELISA run Quality Control (QC) samples
    Back
    Theoretical Calculated Calculated Mean
    Concentrations OD Concentration Concentration
    QC for QCs OD* Mean Standard for QCs for QCs
    Samples: (pg/mL) Values Value Deviation (pg/mL) (pg/mL) % DFT* % CV*
    Front
    QCs
    QC01 45 0.366 0.372 0.008 50.991 51.938 15.417 2.3
    0.378 52.884
    QC02 100 0.753 0.733 0.028 113.944 110.572 10.572 3.9
    0.713 107.2
    QC03 250 1.503 1.509 0.008 256.165 257.454 2.982 0.6
    1.515 258.742
    Back
    QCs
    QC01 45 0.315 0.332 0.024 42.964 45.638 1.418 7.2
    0.349 48.312
    QC02 100 0.712 0.698 0.021 107.033 104.609 4.609 2.9
    0.683 102.185
    QC03 250 1.547 1.558 0.015 265.671 267.967 7.187 1
    1.568 270.263
    *Note:
    OD—optical density;
    % DFT—% Difference from Theoretical;
    CV %—% Coefficient of Variance
  • Based on the ELISA values for the calibration and quality control samples shown in Tables 1 and 2 hereinabove, TNFRI expression in pg per 1×106 cells for samples of mesenchymal stem cells from the donors was determined. As described hereinabove, the mesenchymal stem cells from each donor were non-transfected, or transfected with a TNFRI sense or antisense oligonucleotide at a concentration of 1.25, 2.5, or 5 μg/ml. The ELISA values and the amount of TNFRI expressed by each of the mesenchymal stem cell samples from each of the donors are given in Table 3 below.
  • TABLE 3
    Calculations for ELISA run test samples
    hMSC OD Calculated Mean TNFRI in
    Donor OD* Mean Concentration Concentration pg per
    # Sample description: Values Value SD* (pg/mL) (pg/mL) 1 × 106 cells % CV*
    24 Control 0.385 0.384 0.001 53.989 53.831 53.831 0.4
    (non-transfected cells) 0.383 53.674
    Control oligo- 0.278 0.266 0.018 37.15 35.186 35.186 6.7
    transfected cells 5 0.253 33.221
    μg/mL
    Control oligo- 0.348 0.352 0.006 48.155 48.785 48.785 1.6
    transfected cells 2.5 0.356 49.415
    μg/mL
    Control oligo- 0.386 0.378 0.012 54.147 52.806 52.806 3.2
    transfected cells 1.25 0.369 51.464
    μg/mL
    TNFRI anti-sense 0.117 0.113 0.006 11.533 10.79 10.79 5.7
    oligo-transfected cells 5 0.108 10.047
    μg/mL
    TNFRI anti-sense 0.254 0.245 0.013 33.378 31.962 31.962 5.2
    oligo-transfected cells 0.236 30.546
    2.5 μg/mL
    TNFRI anti-sense 0.321 0.311 0.015 43.907 42.257 42.257 4.8
    oligo-transfected cells 0.3 40.607
    1.25 μg/mL
    007 Control 0.368 0.367 0.002 51.306 51.07 51.07 0.6
    (non-transfected cells) 0.365 50.833
    Control oligo- 0.226 0.219 0.01 28.97 27.866 27.866 4.5
    transfected cells 5 0.212 26.761
    μg/mL
    Control oligo- 0.293 0.272 0.03 39.507 36.128 36.128 11.2
    transfected cells 2.5 0.25 32.749
    μg/mL
    Control oligo- 0.308 0.286 0.032 41.864 38.329 38.329 11.1
    transfected cells 1.25 0.263 34.793
    μg/mL
    TNFRI anti-sense 0.123 0.114 0.013 12.517 10.949 10.949 11.8
    oligo-transfected cells 5 0.104 9.382
    μg/mL
    TNFRI anti-sense 0.269 0.243 0.037 35.736 31.565 31.565 15.5
    oligo-transfected cells 0.216 27.393
    2.5 μg/mL
    TNFRI anti-sense 0.313 0.303 0.014 42.65 41.078 41.078 4.7
    oligo-transfected cells 0.293 39.507
    1.25 μg/mL
    014 Control 0.377 0.38 0.004 52.726 53.2 53.2 1.1
    (non-transfected cells) 0.383 53.674
    Control oligo- 0.251 0.249 0.003 32.907 32.592 32.592 1.1
    transfected cells 5 0.247 32.277
    μg/mL
    Control oligo- 0.338 0.315 0.033 46.581 42.887 42.887 10.6
    transfected cells 2.5 0.291 39.193
    μg/mL
    Control oligo- 0.356 0.347 0.013 49.415 47.919 47.919 3.9
    transfected cells 1.25 0.337 46.424
    μg/mL
    TNFRI anti-sense 0.11 0.104 0.008 10.378 9.379 9.379 8.2
    oligo-transfected cells 5 0.098 8.379
    μg/mL
    TNFRI anti-sense 0.211 0.206 0.008 26.603 25.733 25.733 3.8
    oligo-transfected cells 0.2 24.864
    2.5 μg/mL
    TNFRI anti-sense 0.3 0.294 0.008 40.607 39.664 39.664 2.9
    oligo-transfected cells 0.288 38.722
    1.25 μg/mL
    015 Control 0.475 0.469 0.009 68.284 67.246 67.246 2
    (non-transfected cells) 0.462 66.209
    Control oligo- 0.278 0.279 0.001 37.15 37.308 37.308 0.5
    transfected cells 5 0.28 37.465
    μg/mL
    Control oligo- 0.34 0.343 0.004 46.896 47.289 47.289 1
    transfected cells 2.5 0.345 47.683
    μg/mL
    Control oligo- 0.419 0.413 0.009 59.37 58.34 58.34 2.2
    transfected cells 1.25 0.406 57.31
    μg/mL
    TNFRI anti-sense 0.13 0.125 0.007 13.658 12.842 12.842 5.7
    oligo-transfected cells 5 0.12 12.025
    μg/mL
    TNFRI anti-sense 0.253 0.262 0.012 33.221 34.557 34.557 4.6
    oligo-transfected cells 0.27 35.893
    2.5 μg/mL
    TNFRI anti-sense 0.377 0.381 0.005 52.726 53.279 53.279 1.3
    oligo-transfected cells 0.384 53.831
    1.25 μg/mL
    23 Control 0.260 0.255 0.008 40.591 39.632 39.632 3.1
    (non-transfected cells) 0.249 38.672
    Control oligo- 0.191 0.184 0.010 28.560 27.339 27.339 5.4
    transfected cells 5 0.177 26.117
    μg/mL
    Control oligo- 0.216 0.209 0.009 32.919 31.786 31.786 4.4
    transfected cells 2.5 0.203 30.653
    μg/mL
    Control oligo- 0.222 0.222 0.000 33.965 33.965 33.965 0.0
    transfected cells 1.25 0.222 33.965
    μg/mL
    TNFRI anti-sense 0.107 0.106 0.001 13.798 13.620 13.620 1.3
    oligo-transfected cells 5 0.105 13.441
    μg/mL
    TNFRI anti-sense 0.206 0.187 0.027 31.176 27.860 27.860 14.4
    oligo-transfected cells 0.168 24.544
    2.5 μg/mL
    TNFRI anti-sense 0.213 0.212 0.001 32.396 32.222 32.222 0.7
    oligo-transfected cells 0.211 32.048
    1.25 μg/mL
    486 Control 0.249 0.249 0.001 41.244 41.148 41.148 0.3
    (non-transfected cells) 0.248 41.053
    Control oligo- 0.149 0.136 0.018 22.401 19.981 19.981 13.5
    transfected cells 5 0.123 17.560
    μg/mL
    Control oligo- 0.246 0.231 0.022 40.672 37.732 37.732 9.5
    transfected cells 2.5 0.215 34.792
    μg/mL
    Control oligo- 0.263 0.253 0.015 43.915 41.913 41.913 5.9
    transfected cells 1.25 0.242 39.911
    μg/mL
    TNFRI anti-sense 0.071 0.068 0.004 7.917 7.361 7.361 6.2
    oligo-transfected cells 5 0.065 6.805
    μg/mL
    TNFRI anti-sense 0.142 0.142 0.000 21.096 21.096 21.096 0.0
    oligo-transfected cells 0.142 21.096
    2.5 μg/mL
    TNFRI anti-sense 0.193 0.179 0.021 30.644 27.924 27.924 11.5
    oligo-transfected cells 0.164 25.204
    1.25 μg/mL
    13 Control 0.211 0.209 0.003 34.037 33.659 33.659 1.4
    (non-transfected cells) 0.207 33.282
    Control oligo- 0.134 0.134 0.01 19.606 19.513 19.513 0.5
    transfected cells 5 0.133 19.420
    μg/mL
    Control oligo- 0.195 0.188 0.011 31.020 29.611 29.611 5.7
    transfected cells 2.5 0.180 28.201
    μg/mL
    Control oligo- 0.207 0.192 0.022 33.282 30.366 38.329 11.4
    transfected cells 1.25 0.176 27.451
    μg/mL
    TNFRI anti-sense 0.087 0.080 0.010 10.882 9.585 9.585 12.4
    oligo-transfected cells 5 0.073 8.288
    μg/mL
    TNFRI anti-sense 0.156 0.135 0.030 23.708 19.706 19.706 22.6
    oligo-transfected cells 0.113 15.703
    2.5 μg/mL
    TNFRI anti-sense 0.208 0.174 0.048 33.470 27.097 27.097 27.6
    oligo-transfected cells 0.140 20.723
    1.25 μg/mL
    *Note:
    OD—optical density;
    SD—Standard Deviation;
    CV %—% Coefficient of Variance
  • From the above data shown in Table 3, the mean TNFRI expression, in picograms per 1×106 cells, was determined for non-transfected (control) mesenchymal stem cells, as well as mesenchymal stem cells transfected with 1.25, 2.5, or 5 μl/ml of antisense or sense oligonucleotides. The mean TNFRI expression values are given in Table 4 below.
  • TABLE 4
    TNFRI expression by hMSCs transfected with anti-sense and control
    (sense) oligonucleotides: summary for 7 tested hMSC donors
    TNFRI expression in pg per 1 × 106 cells
    hMSC donor #: Mean for
    486 13 24 007 14 15 23 7 donors SD
    Control (non- 41* 34 54 51 53 67 40 48.57 11.09
    transfected cells)
    TNFRI anti-sense 7 10 11 11 9 13 14 10.71 2.36
    oligo-transfected
    cells 5 μg/mL
    TNFRI anti-sense 21 20 32 32 26 35 28 27.71 5.74
    oligo-transfected
    cells 2.5 μg/mL
    TNFRI anti-sense 28 27 42 41 40 53 32 37.57 9.22
    oligo-transfected
    cells 1.25 μg/mL
    Control (sense) 20 20 35 28 33 37 27 28.57 6.85
    oligo-transfected
    cells 5 μg/mL
    Control (sense) 38 30 49 36 43 47 32 39.29 7.30
    oligo-transfected
    cells 2.5 μg/mL
    Control (sense) 42 30 53 38 48 58 34 43.29 10.21
    oligo-transfected
    cells 1.25 μg/mL
    *Note:
    These values represent mean TNFRI numbers (from table 3, column 8: “TNFRI in pg per 1 × 106 cells”) rounded to whole numbers
  • A second group of transfected cells was used for investigation of the effect of hMSCs on hPBMC proliferation in vitro. Human PBMCs from two different donors were used for this assay. PBMCs were isolated from leukopheresed blood using Ficoll-Paque gradient centrifugation according to the manufacturer's protocol (Amersham Biosciences, Ficoll-Paque Plus product insert). Cells were stored frozen at −80° C. in a medium including 90% FBS and 10% DMSO prior to analysis. On the day of the experiment hPBMCs were thawed, counted and plated into 96-well tissue culture plates at 1×105 cells/well together with hMSCs (1×104 cells/well). A combination of anti-CD3 (1 μg/mL) and anti-CD28 (1 pg/mL) antibodies was used to stimulate lymphocyte proliferation that represents an in vitro model for immune cell activation characteristics of GVHD and rejection of allogeneic organs. (Trickett, et al., J. Immunol. Methods, Vol. 275, pgs. 251-255 (2003); Koulova, et al., J. Exp. Med., Vol. 173, No. 3, pgs. 759-762 (1991); Foster, et al., Transplantation, Vol. 76, No. 6; Czitrom, Clin. Ortho. Relat. Res., Vol. 326, pgs. 11-24 (1996)). The plates then were incubated in a humidified atmosphere containing 5% CO2. The proliferation of PBMCs alone and in the presence of MSCs was measured at day 5 from culture initiation by the addition of [Methyl-3H]-thymidine at 1 μCi/well for the final 18-20 hrs of culture. After labeling, the cells were transferred onto a glass filter using a 96-well plate harvester, and radioactivity incorporated into DNA was measured by a liquid scintillation beta-counter. The uptake of [Methyl-3H]-thymidine into DNA in counts per minute (cpm) represents hPBMC proliferation. Final results were expressed as % inhibition of PBMC proliferation in the presence of MSCs calculated as:

  • 100%−[Proliferation (PBMC+MSC, cpm)×100/Proliferation (PBMC, cpm)]
  • The results for the mesenchymal stem cells from each of the donors are given in Table 5 below.
  • TABLE 5
    Inhibition of CD3/CD28-induced hPBMC proliferation by hMSCs transfected with anti-sense
    and control (sense) oligonucleotides: summary for 7 tested hMSC donors
    % inhibition of hPBMC proliferation by hMSCs
    hMSC donor #: Mean %
    486 13 24 007 14 15 23 for 7
    hPBMC donor #: 2 3 2 3 3 3 3 2 2 3 donors SD
    Control (non- 65 73 82 94 70 66 82 62 68 91 75.30 11.26
    transfected cells)
    TNFRI anti-sense 40 45 46 68 32 10 39 19 38 52 38.90 16.29
    oligo-transfected
    cells 5 μg/mL
    TNFRI anti-sense 83 90 59 86 ND 73 ND 63 47 58 69.88 15.48
    oligo-transfected
    cells 2.5 μg/mL
    TNFRI anti-sense 62 74 86 ND 72 64 57 ND 72 80 70.88 9.58
    oligo-transfected
    cells 1.25 μg/mL
    Control (sense) 38 87 60 77 58 77 62 44 52 53 60.80 15.50
    oligo-transfected
    cells 5 μg/mL
    Control (sense) 60 91 67 ND ND 62 66 57 70 95 71.00 14.22
    oligo-transfected
    cells 2.5 μg/mL
    Control (sense) 87 ND 68 71 66 68 36 ND 49 85 70.57 12.77
    oligo-transfected
    cells 1.25 μg/mL
    Note:
    ND—no data
  • The above data with respect to inhibition of CD3/CD28 induced PBMC proliferation were correlated to the mean TNFRI expression data shown in Table 4 hereinabove. The correlated data with respect to mean TNFRI expression and inhibition of CD3/CD28 induced PBMC proliferation are given in Table 6 below.
  • TABLE 6
    TNFRI expression and effect on hPBMC proliferation in
    vitro by hMSCs transfected with TNFRI oligonucleotides
    % Inhibition of TNFRI
    Oligonucleotide hPBMC expression in
    concentration proliferation pg/1 × 106 MSCs
    Human MSCs condition (μg/mL) (Mean ± SD) (Mean ± SD)
    Untransfected (Control Not applicable 75.30 ± 11.26  48.57 ± 11.09
    MSCs)
    Antisense oligonucleotide 1.25 70.88 ± 9.58  37.57 ± 9.22
    2.5 69.88 ± 15.48 27.71 ± 5.74
    5 38.90 ± 16.29 10.71 ± 2.36
    Sense oligonucleotide 1.25 70.57 ± 12.77  43.29 ± 10.21
    (control oligonucleotide) 2.5 71.00 ± 14.22 39.29 ± 7.30
    5 60.80 ± 15.50 28.57 ± 6.85
  • The results from these experiments show that hMSCs with decreased expression of TNFR type I (TNFRI) lose their ability to suppress hPBMC proliferation in vitro. The data support the premise that the expression of TNFRI is an essential link to the suppression of PBMC proliferation by MSCs. Thus, TNFRI can be used as a potency marker for MSC immunomodulative activity. Based on the obtained data, a potency threshold of 13.07 pg of TNFRI (mean±SD) per 1×106 cells correlates with less than 50% inhibition of hPBMC proliferation (Table 6, FIG. 1). Thus, non-potent MSCs are cells expressing less than 13 pg TNFRI per 1×106 cells.
  • EXAMPLE 2 TNFRI is a Temperature-Sensitive Marker of hMSC Functionality
  • Ex vivo handling of mammalian cells is restricted by a number of factors including temperature. For example, low temperatures such as −80±5° C., or lower, even as low as −135° C. or below (liquid nitrogen) are required for cell storage whereas ex vivo cell expansion requires a temperature of 37±0.5° C. Cell exposure to temperatures outside of the optimal ranges may lead to a decrease in cell functionality or cell death. Mammalian cells are able to withstand short-term minor temperature fluctuations; however, each type of cells has its own temperature tolerance range for cell culture maintenance, shipping, and storage.
  • The expression level of TNFRI on hMSCs correlates with hMSC immunosuppressive activity. The level of TNFRI expression by hMSCs of less than 13 pg/106 cells has been determined as a threshold, below which hMSCs begin to lose their ability to suppress an immune response (See FIG. 1). Thus, TNFRI expression is a marker of hMSC immunosuppression, an activity that is believed essential for MSCs to be efficacious for treatment of immunological reactions taking place in GVHD, organ rejection, autoimmune diseases, and other diseases. Here, effects of temperature fluctuations during storage of frozen hMSCs as well as the effect of time of exposure of cells to room temperature on expression of TNFRI on hMSCs was investigated.
  • Effect of Store Temperature Fluctuations on TNFRI Expression and hMSC immunosuppressive potential.
  • The objective of these experiments was to investigate the ability of hMSCs to retain their functional characteristics after an exposure to temperatures above −80° C., which are not optimal temperatures for storage of frozen cells. Human MSCs were frozen at passage 5 and placed for storage in a freezer at −80±5° C. After several weeks, bags of frozen cells were removed from the −80±5° C. freezer and placed at either −70±5° C., −60±5° C., or −50±5° C. for 72±2 hours. After 72±2 hours, the bags were returned to storage at −80±5° C. for at least 24 hours before thaw and analysis. A set of bags moved from one −80±5° C. freezer to another, following the same schedule as the other bags, served as a control. On the day of the experiment the bags containing the cells were thawed, cells were counted, and cell lysates for the TNFRI ELISA were prepared as described in Example 1. The TNFRI ELISA was performed as described in Example 1. Results are summarized in FIG. 2 (bars show mean TNFRI values ±SD for 3 hMSC bags). The data showed that exposure of hMSCs to temperatures of −60±5° C. or −50±5° C. decreases the TNFRI expression level: the level of TNFRI detected by ELISA was below the determined hMSC potency threshold of 13 pg/106 cells (represented by the solid line on the graph).
  • Parallel with TNFRI measurement, two bags with hMSCs stored at −80±5° C. (optimal storage temperature served as a control) and at −50±5° C. (corresponding to a ±30° C. greater than the −80±5° C. optimal storage temperature) were used for investigation of hMSC immunosuppressive activity. The ability of the MSCs to suppress anti-CD3/CD28-induced proliferation of hPBMCs in vitro was evaluated as described in Example 1. The results showed that hMSCs stored at −50±5° C. lost their ability to suppress hPBMC proliferation, whereas cells stored at −80±5° C. inhibited hPBMC proliferation by 92% (FIG. 3, dark bars represent mean±SD % inhibition of hPBMC proliferation. Numbers inside the dark bars show numerical values). The immunosuppressive activity of MSCs is dependent on the level of TNFRI expression: cells expressing more than 13 pg/106 cells of TNFRI, which was determined as an MSC immunosuppressive potential threshold, are biologically active, and cells with the TNFRI level below 13 pg/106 cells are not (FIG. 3, light bars represent mean±SD of the TNFRI expression level. Numbers inside the light bars show numerical values). Thus, non-optimal storage temperatures decrease TNFRI expression on hMSCs, and which correlates with decrease in hMSC functionality.
  • Effect of Cell Exposure Time to Room Temperature on TNFRI Expression on hMSC.
  • The results of this experiment serve as additional evidence that TNFRI expression on hMSCs is decreasing under cell exposure to non-optimal temperatures. In this experiment the effect of cell suspension storage at room temperature on TNFRI expression was studied. Two hMSC lots were used in the experiment. Bags containing hMSCs were stored at ≦−135° C. prior to the experiment. On the day of the experiment the cells were thawed and diluted with Plasmalyte A physiological solution (Baxter) in a manner that mimics the current cell processing for intravenous hMSC administration at clinical sites. The thawed and diluted hMSCs were kept at room temperature (22° C.-24° C.), and samples were taken and tested for the amount of TNFRI at 0 (immediately post-thaw-baseline), 6, 8, 10, 24, and 32 hours post-thawing. The results showed that exposure of hMSCs to room temperature decreased the TNFRI expression level on the hMSCs (FIG. 4, bars represent mean±SD of the TNFRI expression level for 2 hMSC lots. The solid line represents the TNFRI expression level of 13 pg/106 cells, which is the hMSC potency threshold). The significant decrease in TNFRI expression was observed at 24 hours and 32 hours, and it correlated with a significant decrease in cell viability (below 20%, data not shown).
  • Thus, the experiments described above show that TNFRI expression by hMSCs is sensitive to temperature, and TNFRI can be used as a marker of functionality of hMSC that were exposed to non-optimal temperatures during storage, shipping or cell processing.
  • The disclosures of all patents, publications, including published patent applications, depository accession numbers, and database accession numbers are hereby incorporated by reference to the same extent as if each patent, publication, depository accession number, and database accession number were specifically and individually incorporated by reference.
  • It is to be understood, however, that the scope of the present invention is not to be limited to the specific embodiments described above. The invention may be practiced other than as particularly described and still be within the scope of the accompanying claims.

Claims (25)

1-9. (canceled)
10. A composition comprising allogeneic mesenchymal stem cells wherein said mesenchymal stem cells express TNF-α receptor Type I in an amount of at least 27 pg/106 cells.
11. The composition of claim 10, wherein said mesenchymal stem cells are human mesenchymal stem cells.
12. The composition of claim 10, wherein said mesenchymal stem cells are bone marrow-derived.
13. The composition of claim 10, wherein said mesenchymal stem cells are exposed to at least one freeze-thaw cycle.
14. The composition of claim 10, wherein said mesenchymal stem cells are expanded in culture.
15. The composition of claim 14, wherein said mesenchymal stem cells are expanded for three to eight passages.
16. The composition of claim 15, wherein said mesenchymal stem cells are expanded for four to six passages.
17. The composition of claim 16, wherein said mesenchymal stem cells are expanded for five passages.
18. The composition of claim 10, further comprising an acceptable pharmaceutical carrier.
19. The composition of claim 10, further comprising dimethyl sulfoxide.
20. A method of treating an immunological response in a patient comprising:
administering to said patient allogeneic mesenchymal stem cells, wherein said mesenchymal stem cells express TNF-α receptor Type I in an amount of at least 13 pg/106 cells.
21. The method of claim 20, wherein said mesenchymal stem cells express TNF-α receptor Type I in an amount of at least 18 pg/106 cells.
22. The method of claim 21, wherein said mesenchymal stem cells express TNF-α receptor Type I in an amount of at least 27 pg/106 cells.
23. The method of claim 20, wherein said mesenchymal stem cells are human mesenchymal stem cells.
24. The method of claim 20, wherein said immunological response is associated with an autoimmune disease.
25. The method of claim 24, wherein said autoimmune disease is from the group consisting essentially of rheumatoid arthritis, multiple sclerosis, Type I diabetes, Guillain-Barré syndrome, lupus erythematosus, myasthenia gravis, optic neuritis, psoriasis, Graves' disease, Hashimoto's disease, Ord's thyroiditis, aplastic anemia, Reiter's syndrome, autoimmune hepatitis, primary biliary cirrhosis, antiphospholipid antibody syndrome, opsoclonus myoclonus syndrome, temporal arteritis, acute disseminated encephalomyelitis, Good pasture's syndrome, Wegener's granulomatosis, coeliac disease, pemphigus, polyarthritis, warm autoimmune hemolytic anemia, and scleroderma.
26. The method of claim 20, wherein said immunological response is associated with Crohn's disease.
27. The method of claim 20, wherein said immunological response is associated with graft versus host disease.
28. A method of determining immunosuppressive potency of a population of mesenchymal stem cells comprising:
obtaining a sample from the population of mesenchymal stem cells;
quantifying TNF-α receptor Type I expression in the sample to obtain a measured value; and
comparing the measured value to a threshold value.
29. The method of claim 28, wherein said threshold value is at least 13 pg/106 cells.
30. The method of claim 29, wherein said threshold value is at least 18 pg/106 cells.
31. The method of claim 30, wherein said threshold value is at least 27 pg/106 cells.
32. The method of claim 28, wherein TNF-α receptor Type I expression is quantified using an enzyme-linked immunosorbent assay.
33. The method of claim 28, further comprising lysing cells in the sample prior to quantifying TNF-α receptor Type I expression.
US12/091,391 2006-01-13 2007-01-05 Mesenchymal stem cells expressing tnf-a receptor Abandoned US20090169522A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/091,391 US20090169522A1 (en) 2006-01-13 2007-01-05 Mesenchymal stem cells expressing tnf-a receptor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US75915706P 2006-01-13 2006-01-13
US12/091,391 US20090169522A1 (en) 2006-01-13 2007-01-05 Mesenchymal stem cells expressing tnf-a receptor
PCT/US2007/000274 WO2007087139A2 (en) 2006-01-13 2007-01-05 MESENCHYMAL STEM CELLS EXPRESSING TNF-α RECEPTOR

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/000274 A-371-Of-International WO2007087139A2 (en) 2006-01-13 2007-01-05 MESENCHYMAL STEM CELLS EXPRESSING TNF-α RECEPTOR

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/402,498 Continuation US8486695B2 (en) 2006-01-13 2012-02-22 Mesenchymal stem cells expressing TNF-alpha receptors

Publications (1)

Publication Number Publication Date
US20090169522A1 true US20090169522A1 (en) 2009-07-02

Family

ID=38309740

Family Applications (9)

Application Number Title Priority Date Filing Date
US12/091,391 Abandoned US20090169522A1 (en) 2006-01-13 2007-01-05 Mesenchymal stem cells expressing tnf-a receptor
US11/650,374 Abandoned US20070258963A1 (en) 2006-01-13 2007-01-05 Mesenchymal stem cells expressing TNF-alpha receptors
US13/085,720 Abandoned US20110189768A1 (en) 2006-01-13 2011-04-13 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US13/402,498 Active US8486695B2 (en) 2006-01-13 2012-02-22 Mesenchymal stem cells expressing TNF-alpha receptors
US13/906,592 Abandoned US20130259841A1 (en) 2006-01-13 2013-05-31 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US14/228,327 Abandoned US20140248244A1 (en) 2006-01-13 2014-03-28 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US14/484,444 Abandoned US20150004693A1 (en) 2006-01-13 2014-09-12 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US15/693,754 Abandoned US20180087032A1 (en) 2006-01-13 2017-09-01 Mesenchymal stem cells expressing tnf-alpha receptors
US17/087,124 Active 2027-09-14 US11821004B2 (en) 2006-01-13 2020-11-02 Mesenchymal stem cells expressing TNF-α receptors

Family Applications After (8)

Application Number Title Priority Date Filing Date
US11/650,374 Abandoned US20070258963A1 (en) 2006-01-13 2007-01-05 Mesenchymal stem cells expressing TNF-alpha receptors
US13/085,720 Abandoned US20110189768A1 (en) 2006-01-13 2011-04-13 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US13/402,498 Active US8486695B2 (en) 2006-01-13 2012-02-22 Mesenchymal stem cells expressing TNF-alpha receptors
US13/906,592 Abandoned US20130259841A1 (en) 2006-01-13 2013-05-31 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US14/228,327 Abandoned US20140248244A1 (en) 2006-01-13 2014-03-28 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US14/484,444 Abandoned US20150004693A1 (en) 2006-01-13 2014-09-12 Mesenchymal Stem Cells Expressing TNF-alpha Receptors
US15/693,754 Abandoned US20180087032A1 (en) 2006-01-13 2017-09-01 Mesenchymal stem cells expressing tnf-alpha receptors
US17/087,124 Active 2027-09-14 US11821004B2 (en) 2006-01-13 2020-11-02 Mesenchymal stem cells expressing TNF-α receptors

Country Status (11)

Country Link
US (9) US20090169522A1 (en)
EP (3) EP2465922B1 (en)
CN (1) CN101370930A (en)
AU (1) AU2007208504B2 (en)
BR (1) BRPI0706529A2 (en)
CA (2) CA2635915C (en)
ES (1) ES2415855T3 (en)
HK (1) HK1151553A1 (en)
MX (2) MX348735B (en)
WO (1) WO2007087139A2 (en)
ZA (1) ZA200805609B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012026712A3 (en) * 2010-08-23 2012-05-24 주식회사 강스템홀딩스 Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing stem cells treated with nod2 agonist or cultured product thereof
WO2012048093A3 (en) * 2010-10-08 2012-08-02 Osiris Therapeutics, Inc. Enhanced msc preparations
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US8591883B2 (en) 2005-12-29 2013-11-26 Anthrogenesis Corporation Placental stem cell populations
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005227295A1 (en) 2004-03-22 2005-10-06 Osiris Therapeutics, Inc. Mesenchymal stem cells and uses therefor
MX348735B (en) 2006-01-13 2017-06-27 Mesoblast Int Sarl MESENCHYMAL STEM CELLS EXPRESSING TNF-a RECEPTOR.
US9192695B2 (en) 2008-11-20 2015-11-24 Allosource Allografts combined with tissue derived stem cells for bone healing
US8956862B2 (en) * 2009-11-27 2015-02-17 Stempeutics Research Pvt. Ltd. Methods of preparing mesenchymal stem cells, compositions and kit thereof
US9725689B2 (en) 2010-10-08 2017-08-08 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
CN104508124B (en) 2011-07-06 2017-11-17 细胞治疗有限公司 Mesodermal lineage progenitor cells
WO2014150784A1 (en) 2013-03-15 2014-09-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
EP3122866B1 (en) 2014-03-25 2019-11-20 Terumo BCT, Inc. Passive replacement of media
EP3198006B1 (en) 2014-09-26 2021-03-24 Terumo BCT, Inc. Scheduled feed
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor
EP4137562A1 (en) 2017-05-04 2023-02-22 Mesoblast International Sàrl Mesenchymal lineage precursor or stem cells with enhanced immunosuppression
US11701391B2 (en) 2017-10-24 2023-07-18 Dalia ELANI Methods of treating an ischemic disease
CN110938668A (en) * 2019-11-21 2020-03-31 北京贝来生物科技有限公司 Method for detecting biological efficacy of mesenchymal stem cells
US20230398154A1 (en) * 2020-08-10 2023-12-14 Mesoblast International Sárl A composition comprising mesenchymal precursor or stem cells and their use
WO2024009226A1 (en) * 2022-07-05 2024-01-11 Mesoblast International Sarl Cryopreserved intermediate and potency assay for same

Citations (6)

* 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
US6007995A (en) * 1998-06-26 1999-12-28 Isis Pharmaceuticals Inc. Antisense inhibition of TNFR1 expression
US6071889A (en) * 1991-07-08 2000-06-06 Neurospheres Holdings Ltd. In vivo genetic modification of growth factor-responsive neural precursor cells
US6797514B2 (en) * 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US20040241141A1 (en) * 2001-04-17 2004-12-02 Genetix Pharmaceuticals, Inc. Method of treating arthritis using lentiviral vectors in gene therapy
US20070258963A1 (en) * 2006-01-13 2007-11-08 Osiris Therapeutics, Inc. Mesenchymal stem cells expressing TNF-alpha receptors

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6156304A (en) * 1990-12-20 2000-12-05 University Of Pittsburgh Of The Commonwealth System Of Higher Education Gene transfer for studying and treating a connective tissue of a mammalian host
JPH10501815A (en) 1994-06-07 1998-02-17 リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミネソタ Methods for inhibiting antigen-specific T cell responses
US6974571B2 (en) 1995-03-28 2005-12-13 Thomas Jefferson University Isolated stromal cells and methods of using the same
US5955257A (en) * 1997-10-21 1999-09-21 Regents Of The University Of Minnesota Infusible grade short-term cell storage medium for mononuclear cells
US6251295B1 (en) 1998-01-08 2001-06-26 Nexell Therapeutics Inc. Method for recirculation washing of blood cells
US6368636B1 (en) 1998-03-18 2002-04-09 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
PT1066052E (en) * 1998-03-18 2006-06-30 Osiris Therapeutics Inc MESENCHIMATE STEM CELLS FOR PREVENTION AND TREATMENT
AU2999199A (en) 1998-04-03 1999-10-25 Osiris Therapeutics, Inc. Mesenchymal stem cells as immunosuppressants
WO1999061588A1 (en) 1998-05-22 1999-12-02 Osiris Therapeutics, Inc. Production of megakaryocytes by co-culturing human mesenchymal stem cells with cd34+ cells
AUPQ147799A0 (en) 1999-07-07 1999-07-29 Medvet Science Pty. Ltd. Mesenchymal precursor cell
US8147824B2 (en) * 1999-08-05 2012-04-03 Athersys, Inc. Immunomodulatory properties of multipotent adult progenitor cells and uses thereof
WO2001021767A2 (en) * 1999-09-24 2001-03-29 Morphogen Pharmaceuticals, Inc. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US7144729B2 (en) * 2000-09-01 2006-12-05 Dfb Pharmaceuticals, Inc. Methods and compositions for tissue regeneration
US20050048035A1 (en) 2001-12-07 2005-03-03 Fraser John K. Methods of using regenerative cells in the treatment of stroke and related diseases and disorders
US8404229B2 (en) 2001-12-07 2013-03-26 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to treat acute tubular necrosis
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
CH700956B1 (en) * 2003-05-08 2010-11-15 Cellartis Ab Transferring human blastocyst-derived stem (hBS) cells into a feeder-free culture system, useful in treating cardio-related diseases, comprises transferring the hBS cells from feeder to feeder-free culture by mechanical treatment
US20080095749A1 (en) 2004-03-22 2008-04-24 Sudeepta Aggarwal Mesenchymal stem cells and uses therefor
AU2005227295A1 (en) 2004-03-22 2005-10-06 Osiris Therapeutics, Inc. Mesenchymal stem cells and uses therefor
US20060063141A1 (en) * 2004-09-17 2006-03-23 Mcgann Locksley E Method of cryopreserving cells
EP1795588A1 (en) * 2005-12-07 2007-06-13 Cellerix, S.L. Use of adipose tissue derived mesenchymal stem cells for the treatment of graft versus host disease
US20070253931A1 (en) 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
KR100908481B1 (en) 2006-04-24 2009-07-21 코아스템(주) Mesenchymal stem cell culture medium and culture method of mesenchymal stem cells using the same
ES2608974T3 (en) 2008-08-14 2017-04-17 Mesoblast International Sàrl Compositions of purified mesenchymal stem cells
EP2624845A4 (en) 2010-10-08 2015-09-09 Mesoblast Internat S Rl Enhanced msc preparations
CN107206027A (en) 2014-12-23 2017-09-26 迈索布拉斯特国际有限公司 Carry out the prevention of DHF
AU2018372631A1 (en) 2017-11-22 2020-05-21 Mesoblast International Sarl Cellular compositions and methods of treatment I
WO2020141473A1 (en) 2019-01-02 2020-07-09 Mesoblast International Sárl Method for treating lower back pain
JP2022516187A (en) 2019-01-03 2022-02-24 メゾブラスト・インターナショナル・エスアーエールエル How to improve your eyesight
US20220160776A1 (en) 2019-01-28 2022-05-26 Mesoblast International Sárl Method for treating or preventing gastrointestinal bleeding
AU2020277691A1 (en) 2019-05-23 2021-12-23 Mesoblast International Sarl Functional recovery from cerebral infarction
CA3149478A1 (en) 2019-08-05 2021-02-11 Mesoblast International Sarl Cellular compositions comprising viral vectors and methods of treatment
JP2023513847A (en) 2020-02-19 2023-04-03 メゾブラスト・インターナショナル・エスアーエールエル Methods of treating chronic graft-versus-host disease
WO2021180851A1 (en) 2020-03-11 2021-09-16 Mesoblast International Sàrl Method for treating inflammatory bowel disease ii

Patent Citations (6)

* 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
US6071889A (en) * 1991-07-08 2000-06-06 Neurospheres Holdings Ltd. In vivo genetic modification of growth factor-responsive neural precursor cells
US6007995A (en) * 1998-06-26 1999-12-28 Isis Pharmaceuticals Inc. Antisense inhibition of TNFR1 expression
US6797514B2 (en) * 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US20040241141A1 (en) * 2001-04-17 2004-12-02 Genetix Pharmaceuticals, Inc. Method of treating arthritis using lentiviral vectors in gene therapy
US20070258963A1 (en) * 2006-01-13 2007-11-08 Osiris Therapeutics, Inc. Mesenchymal stem cells expressing TNF-alpha receptors

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9078898B2 (en) 2005-12-29 2015-07-14 Anthrogenesis Corporation Placental stem cell populations
US8591883B2 (en) 2005-12-29 2013-11-26 Anthrogenesis Corporation Placental stem cell populations
US10383897B2 (en) 2005-12-29 2019-08-20 Celularity, Inc. Placental stem cell populations
US8691217B2 (en) 2005-12-29 2014-04-08 Anthrogenesis Corporation Placental stem cell populations
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US9198938B2 (en) 2008-11-19 2015-12-01 Antrhogenesis Corporation Amnion derived adherent cells
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
WO2012026712A3 (en) * 2010-08-23 2012-05-24 주식회사 강스템홀딩스 Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing stem cells treated with nod2 agonist or cultured product thereof
CN103118691A (en) * 2010-08-23 2013-05-22 康干细胞控股有限公司 Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing stem cells treated with NOD2 agonist or cultured product thereof
US9408873B2 (en) 2010-08-23 2016-08-09 Kang Stem Biotech Co., Ltd. Pharmaceutical composition comprising stem cells treated with NOD2 agonist or culture thereof for prevention and treatment of immune disorders and inflammatory diseases
CN105727250A (en) * 2010-08-23 2016-07-06 康干细胞生物科技有限公司 Pharmaceutical composition comprising stem cells treated with NOD2 agonist or culture thereof for prevention and treatment of immune disorders and inflammatory diseases
US9828586B2 (en) 2010-10-08 2017-11-28 Mesoblast International Sárl Enhanced MSC preparations
US9963678B2 (en) 2010-10-08 2018-05-08 Mesoblast International Sàrl Enhanced MSC preparations
WO2012048093A3 (en) * 2010-10-08 2012-08-02 Osiris Therapeutics, Inc. Enhanced msc preparations
US10550369B2 (en) 2010-10-08 2020-02-04 Mesoblast International Sarl Enhanced MSC preparation
US11708560B2 (en) 2010-10-08 2023-07-25 Mesoblast International Sarl Enhanced MSC preparations
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US11090339B2 (en) 2011-06-01 2021-08-17 Celularity Inc. Treatment of pain using placental stem cells

Also Published As

Publication number Publication date
CN101370930A (en) 2009-02-18
US20130259841A1 (en) 2013-10-03
MX348735B (en) 2017-06-27
EP1971679A2 (en) 2008-09-24
ES2415855T3 (en) 2013-07-29
US20110189768A1 (en) 2011-08-04
MX2008008774A (en) 2008-09-26
ZA200805609B (en) 2009-04-29
US20150004693A1 (en) 2015-01-01
US20120214178A1 (en) 2012-08-23
EP2465922A2 (en) 2012-06-20
BRPI0706529A2 (en) 2011-03-29
US20210171913A1 (en) 2021-06-10
WO2007087139A2 (en) 2007-08-02
US20140248244A1 (en) 2014-09-04
EP2465922A3 (en) 2012-12-05
EP1971679B1 (en) 2013-04-10
US20180087032A1 (en) 2018-03-29
HK1151553A1 (en) 2012-02-03
EP2465922B1 (en) 2018-08-01
CA2635915C (en) 2015-09-08
CA2635915A1 (en) 2007-08-02
EP1971679A4 (en) 2009-12-09
US8486695B2 (en) 2013-07-16
WO2007087139A3 (en) 2007-10-11
US11821004B2 (en) 2023-11-21
AU2007208504B2 (en) 2011-04-21
US20070258963A1 (en) 2007-11-08
CA2893204C (en) 2016-11-29
EP3287520A1 (en) 2018-02-28
AU2007208504A1 (en) 2007-08-02
CA2893204A1 (en) 2007-08-02

Similar Documents

Publication Publication Date Title
US11821004B2 (en) Mesenchymal stem cells expressing TNF-α receptors
EP2298864B1 (en) Mesenchymal stem cells and uses therefor
KR101891260B1 (en) Treatment of t-cell mediated immune disorders
US20220218758A1 (en) Quadri-positive stromal cell (qpsc) population for superior cell protection and immunomodulation
CN103502437A (en) High telomerase activity bone marrow mesenchymal stem cells, methods of producing the same and pharmaceuticals and treatment methods based thereon
KR102002090B1 (en) Methods of treating or preventing neurological diseases
JP2012510279A (en) Method for producing adipose-derived stem cells and use of the cells in the treatment of diseases
AU2011202952B2 (en) Mesenchymal stem cells expressing TNF-alpha receptor
AU2015264932A1 (en) Mesenchymal stem cells expressing TNF-alpha receptor
Larson Characterization of Stromal Cells Derived from Bone Marrow, Dermis, and Myocardium

Legal Events

Date Code Title Description
AS Assignment

Owner name: OSIRIS THERAPEUTICS, INCORPORATED, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DANILKOVITCH, ALLA;CARTER, DIANE;TYRELL, ALICIA;AND OTHERS;REEL/FRAME:021748/0824;SIGNING DATES FROM 20080815 TO 20080905

STCB Information on status: application discontinuation

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

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