CA2563518C - Multi-lineage progenitor cells - Google Patents

Multi-lineage progenitor cells Download PDF

Info

Publication number
CA2563518C
CA2563518C CA2563518A CA2563518A CA2563518C CA 2563518 C CA2563518 C CA 2563518C CA 2563518 A CA2563518 A CA 2563518A CA 2563518 A CA2563518 A CA 2563518A CA 2563518 C CA2563518 C CA 2563518C
Authority
CA
Canada
Prior art keywords
mlpc
cells
clonal line
phenotype
purified population
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2563518A
Other languages
French (fr)
Other versions
CA2563518A1 (en
Inventor
Daniel P. Collins
Stacey L. Sprague
Barbara M. Tigges
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.)
Bioe LLC
Original Assignee
Bioe LLC
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 Bioe LLC filed Critical Bioe LLC
Publication of CA2563518A1 publication Critical patent/CA2563518A1/en
Application granted granted Critical
Publication of CA2563518C publication Critical patent/CA2563518C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC

Abstract

Fetal blood multi-lineage progenitor cells that are capable of a wide spectrum of transdifferentiation are described.

Description

Multi-lineage Progenitor Cells CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority from U.S. Provisional Application Serial No. 60/564,687, filed on April 23, 2004.
TECHNICAL FIELD
The invention relates to multi-lineage progenitor cells (MLPC) from human blood and more particularly, to MLPC with the potential to differentiate into multiple tissue lineages and use of such cells for regenerative therapies.
BACKGROUND
Progenitor cells capable of hematopoietic reconstitution after myeloablative therapy have been identified in a number of sources including the bone marrow, umbilical cord and placental blood, and in the peripheral blood of subjects treated with stem cell-mobilizing doses of granulocyte-colony stimulation factor. These cells, often referred to as hematopoietic stem cells (HSC), are identified by the presence of cell surface glycoproteins such as and CD133. HSC represent a very small percentage of the total population of cells given as part of a 'bone marrow transplant' and are considered to be the life-saving therapeutic portion of this treatment responsible for the restoration of the blood-forming capacity of patients given myeloablative doses of chemotherapy or radiation therapy. Stem cell therapies via bone marrow transplantation have become a standard treatment for a number of intractable leukemias and genetic blood disorders.
Recent studies have suggested the presence of a more primitive cell population in the bone marrow capable of self-renewal as well as differentiation into a number of different tissue types other than blood cells. These multi-potential cells were discovered as a minor component in the CD34-plastic-adherent cell population of adult bone marrow, and are variously referred to as mesenchyrnal stem cells (MSC) (Pittenger, et al., Science 284:143-147 (1999)) or multi-potent adult progenitor cells (MAPC) cells (Furcht, L.T., et al., U.S. patent publication 20040107453 Al). MSC cells do not have a single specific identifying marker, but have been shown to be positive for a number of markers, including CD29, CD90, CD105, and CD73, and negative for other markers, including CD14, CD3, and CD34. Various groups have reported to differentiate MSC cells into myocytes, neurons, pancreatic beta-cells, liver cells, bone cells, and connective tissue. Another group (Wernet et al., U.S. patent publication 20020164794 Al) has described an unrestricted somatic stem cell (USSC) with multi-potential capacity that is derived from a CD457CD34-population within cord blood.
SUMMARY
The invention is based on the identification of a rare undifferentiated cell population from human fetal blood that is capable of self-renewal and has the potential to differentiate into cells representing each of the three embryonic germ layers. These fetal blood-derived cells are referred to as multi-lineage progenitor cells (MLPC). As described herein, fetal blood MLPC are distinguished from bone marrow-derived MSC, HSC, and USSC on the basis of their immunophenotypic characteristics, morphology, and distinct growth pattern.
The invention provides methods for developing monotypic clonal cell lines from individual cells.. The invention also provides methods for cryopreserving MLPC (e.g., for cord blood banking) and methods of using MLPC in regenerative therapies.
In one aspect, the invention features a purified population of human fetal blood (e.g., cord blood) MLPC, wherein the MLPC are positive for CD9 and CD45. The MLPC can display a leukocyte morphology. The MLPC can be further positive for SSEA-4 or CD34, as well as CD133, CD41, CD44, CD105, CD29, CD73, CD90, stem cell factor, SSEA-3, and CD13. The MLPC can be negative for CD15, CD38, glycophorin-A, CD2, CD3, CD8, CD19, CD20, CD22, CD5, CD7, CD10, CD14, CD4, HLA-DR, CD16, CD33, and CD61. The MLPC can attain a fibroblast-like morphology over time in culture. The MLPC
also can adhere to a plastic surface when cultured. The MLPC are capable of differentiating into cells from all three embryonic germ layers, including, for example, cells having an osteocytic phenotype, cells having an adipocytic phenotype, cells having a neural stem cell phenotype, cells having a myocytic phenotype, cells having an endothelial phenotype, cells having a hepatocytic phenotype, and cells having a pancreatic phenotype. The MLPC can include an exogenous nucleic acid (e.g., an exogenous nucleic acid encoding a polypeptide).
In another aspect, the invention features a purified population of human fetal blood (e.g., cord blood) MLPC, wherein the MLPC are positive for CD9, negative for CD45, negative for CD34, and negative for SSEA-4. The MLPC
can display a fibroblast morphology. The MLPC can be further positive for CD13, CD29, CD44, CD73, CD90, and CD105, and can be further negative for CD2, CD3, CD4, CD5, CD7, CD8, CD10, CD14, CD15, CD16, CD19, CD20, CD22, CD33, CD36, CD38, CD41, CD61, CD62E, CD133, glycophorin-A, stem cell factor, SSEA-3, and HLA-DR. The MLPC can adhere to a plastic surface when cultured. The MLPC are capable of differentiating into cells from all three embryonic germ layers, including, for example, cells having an osteocytic phenotype, cells having an adipocytic phenotype, cells having a neural stem cell phenotype, cells having a myocytic phenotype, cells having an endothelial phenotype, cells having a hepatocytic phenotype, and cells having a pancreatic phenotype. The MLPC can include an exogenous nucleic acid (e.g., an exogenous nucleic acid encoding a polypeptide).
The invention also features a clonal line of human fetal blood (e.g., cord blood) MLPC, wherein the MLPC are positive for CD9, negative for CD45, negative for CD34, and negative for SSEA-4. The MLPC can display a fibroblast morphology. The MLPC can be further positive for CD13, CD29, CD44, CD73, CD90, and CD105, and can be further negative for CD2, CD3, CD4, CD5, CD7, CD8, CD10, CD14, CD15, CD16, CD19, CD20, CD22, CD33, CD36, CD38, CD41, CD61, CD62E, CD133, glycophorin-A, stem cell factor, SSEA-3, and HLA-DR. The MLPC can adhere to a plastic surface when cultured. The MLPC are capable of differentiating into cells from all three embryonic germ layers, including, for example, cells having an osteocytic phenotype, cells having an adipocytic phenotype, cells having a neural stem cell phenotype, cells having a myocytic phenotype, cells having an endothelial phenotype, cells having a hepatocytic phenotype, and cells having a pancreatic phenotype. The MLPC can include an exogenous nucleic acid (e.g., an exogenous nucleic acid encoding a polypeptide). In some embodiments, the MLPC have undergone at least 5 doublings (e.g., at least 8, at least 10, at least 15, or at least 25 doublings) in culture.
In another aspect, the invention features a composition that includes a purified population of MLPC or a clonal line of MLPC and a culture medium.
The composition further can include a cryopreservative. In one embodiment, the cryopreservative is dimethylsulfoxide (DMSO) (e.g., 1 to 10% DMSO). In another embodiment, the cryopreservative is fetal bovine serum, human serum, or human serum albumin in combination with one or more of the following:
DMSO, trehalose, and dextran. For example, the cryopreservative can be DMSO and trehalose, or fetal bovine serum and DMSO.
In yet an another aspect, the invention features an article of manufacture that includes a purified population of MLPC or a clonal line of MLPC. The purified population of MLPC or the clonal line can be housed within a container (e.g., a vial or a bag). The container further can include a cryopreservative.
The invention also features a method for purifying a population of MLPC
from human fetal blood. The method includes contacting a human fetal blood sample (e.g., cord blood) with a composition that includes dextran, anti-glycophorin A antibody, anti-CD15 antibody, and anti-CD9 antibody; allowing the sample to partition into an agglutinate and a supernatant phase;
recovering cells from the supernatant phase; and purifying MLPC from the recovered cells by adherence to a solid substrate (e.g., a plastic substrate), wherein the MLPC
are positive for CD9 and positive for CD45. The MLPC can be further positive for CD34, CD133, CD41, CD44, CD105, CD29, CD73, CD90, stem cell factor, SSEA-3, SSEA-4, and CD13. The MLPC can be further negative for CD15, CD38, glycophorin-A, CD2, CD3, CD8, CD19, CD20, CD22, CD5, CD7, CD10, CD14, CD4, HLA-DR, CD16, CD33, and CD61. The method further can include testing the MLPC for CD9 or testing for CD9, CD29, CD45, CD73, and CD90.
The method further can include culturing the MLPC such that the MLPC
obtain a fibroblast morphology, wherein the MLPC, after obtaining the fibroblast morphology, are positive for CD9, negative for CD45, negative for CD34, and negative for SSEA-4. The MLPC, after obtaining the fibroblast morphology, can be further positive for CD13, CD29, CD44, CD73, CD90, and CD105. The MLPC, after obtaining the fibroblast morphology, can be further negative for CD2, CD3, CD4, CD5, CD7, CD8, CD10, CD14, CD15, CD16, CD19, CD20, CD22, CD33, CD36, CD38, CD41, CD61, CD62E, CD133, glycophorin-A, stem cell factor, SSEA-3, and HLA-DR. The method further can include testing the MLPC for CD9 or testing for CD9, CD29, CD45, CD73, and CD90.
In another aspect, the invention features a method for cryopreserving MLPC. The method includes contacting a purified population of MLPC or clonal line of MLPC with a cryopreservative; and freezing the purified population of MLPC or the clonal line. In one embodiment, the cryopreservative is DMSO (e.g., 1 to 10% DMSO). In another embodiment, the cryopreservative is fetal bovine serum, human serum, or human serum albumin in combination with one or more of the following: DMSO, trehalose, and dextran. For example, the cryopreservative can be DMSO and trehalose, or fetal bovine serum and DMSO. The purified population of MLPC or the clonal line can be suspended in the cryopreservative at a concentration between 1 x 105 and 5 x cells/mL. The purified population or clonal line can be frozen at a controlled rate (e.g., the freezing rate is controlled electronically) or by placement in an ethanol bath in the vapor phase of a liquid nitrogen cryogenic storage tank.
In yet another aspect, the invention features a method of producing a population of differentiated cells. The method includes culturing a purified population of MLPC or a clonal line of MLPC with an agent effective to induce differentiation of the MLPC. The agent can include insulin, glutamine, dexamethasone, indomethacin, and 3-isobuty1-1-methyl-xanthine;
dexamethasone, glutamine, ascorbate, and 13-glycerophosphate; epithelial growth factor, insulin, fetuin, dexamethasone, and fibroblast growth factor-basic;
fibroblast growth factor-basic, epidermal growth factor, NSF-1, and retinoic acid; heparin, bovine brain extract, epithelial growth factor, and hydrocortisone;
or ascorbic acid, hydrocortisone, transferrin, insulin, epidermal growth factor, hepatocyte growth factor, fibroblast growth factor-basic, fibroblast growth factor-4, and stem cell factor.
In various aspects, the present invention relates to a purified population of human fetal blood multi-lineage progenitor cells (MLPC), wherein said MLPC are positive for CD9, CD13, CD29, CD44, CD73, CD90 and CD105, and negative for CD10, CD34, CD41, CD45, Stro-1, Stage Specific Embryonic Antigen-3 (SSEA-3) and SSEA-4.
In various aspects, the present invention relates to a clonal line of human fetal blood multi-lineage progenitor cells (MLPC), wherein said MLPC are positive for CD9, CD13, CD29, CD44, CD73, CD90 and CD105, and negative for CD10, CD34, CD41, CD45, Stro-1, Stage Specific Embryonic Antigen-3 (SSEA-3) and SSEA-4.
In various aspects, the present invention relates to a composition comprising the purified population of MLPC as defined herein, or the clonal line as defined herein and a culture medium.
In various aspects, the present invention relates to an article of manufacture comprising the purified population of MLPC as defined herein, or the clonal line as defined herein, wherein said purified population of MLPC or said clonal line is housed within a container.
In various aspects, the present invention relates to a method for purifying a population of multi-lineage progenitor cells (MLPC) from human fetal blood, said method comprising: contacting a human fetal blood sample with a composition, said composition comprising:
dextran; anti-glycophorin A antibody; anti-CD15 antibody; and anti-CD9 antibody; allowing said sample to partition into an agglutinate and a supernatant phase; recovering cells from said supernatant phase;
purifying MLPC from the recovered cells by adherence to a solid substrate, wherein said MLPC are positive for CD9 and positive for CD45; and culturing said MLPC such that said MLPC obtain a fibroblast morphology, wherein said MLPC after obtaining said fibroblast morphology are positive for CD9, CD13, CD29, CD44, CD73, CD90 and CD105, and negative for CD10, CD34, CD41, CD45, Stro-1, Stage Specific Embryonic Antigen-3 (SSEA-3) and SSEA-4.
In various aspects, the present invention relates to a method for cryopreserving MLPC, said method comprising: contacting said purified population of MLPC as defined herein, or said as defined herein with a cryopreservative; and freezing said purified population of MLPC or said clonal line.
In various aspects, the present invention relates to a method of producing a population of differentiated cells, said method comprising culturing the purified population of MLPC as defined herein, or the clonal line as defined herein with an agent effective to induce differentiation of said MLPC.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
DESCRIPTION OF DRAWINGS
FIG 1 is a schematic of a cell separation procedure for purifying MLPC from fetal blood.
FIG 2A-2D are photomicrographs depicting the morphology of developing MLPC.

shows an early culture of MLPC isolated from umbilical cord blood demonstrating the cells in the leukocyte morphology phase. FIG 2B shows a culture of MLPC beginning to change their morphology from leukocyte to fibroblast morphology. FIG 2C shows a later culture of MLPC in logarithmic growth phase. FIG 2D shows a fully confluent culture of MLPC.
DETAILED DESCRIPTION
In general, the invention provides purified populations of MLPC from human fetal blood (e.g., umbilical cord blood ("cord blood"), placental blood, or the blood from a fetus) and clonal MLPC lines derived from individual MLPC. Fetal blood provides a source of cells that is more immature than adult bone marrow and has a higher percentage of cells bearing immature cell surface markers. Consequently, there may be advantages in the expansion and differentiation capacity of the progenitor cells from fetal blood. As described herein, MLPC
have immunophenotypic characteristics distinct from bone marrow derived MSC's, bone marrow-derived HSC, and umbilical cord blood-6a derived HSC and USSC. The cells described herein have the capacity to self renew and differentiate into diverse tissue types similar to the bone marrow-derived MSC and MAPC cells. MLPC can be used to develop cellular therapies and establish cryopreserved cell banks for future regenerative medicine procedures. MLPC also can be modified such that the cells can produce one or more polypeptides or other therapeutic compounds of interest.
Cell Separation Compositions MLPC can be isolated from fetal blood (e.g., cord blood) using the negative selection process and cell separation compositions disclosed in U.S.
Patent Publication No. 2003-0027233-Al. Such cell compositions can include dextran and one or more antibodies against (i.e., that have binding affinity for) a cell surface antigen.
Dextran is a polysaccharide consisting of glucose units linked predominantly in alpha (1 to 6) mode. Dextran can cause stacking of erythrocytes (i.e., rouleau formation) and thereby facilitate the removal of erythroid cells from solution. Antibodies against cell surface antigens can facilitate the removal of blood cells from solution via homotypic agglutination (i.e., agglutination of cells of the same cell type) and/or heterotypic agglutination (i.e., agglutination of cells of different cell types).
For example, a cell separation composition can include dextran and antibodies against glycophorin A, CD15, and CD9. Cell separation compositions also can contain antibodies against other blood cell surface antigens including, for example, CD2, CD3, CD4, CD8, CD72, CD16, CD41a, HLA Class I, HLA-DR, CD29, CD1 I a, CD1 lb, CD11c, CD19, CD20, CD23, CD39, CD40, CD43, CD44, CDw49d, CD53, CD54, CD62L, CD63, CD66, CD67, CD81, CD82, CD99, CD100, Leu-13, TPA-1, surface Ig, and combinations thereof. Thus, cell separation compositions can be formulated to selectively agglutinate particular types of blood cells.
Typically, the concentration of anti-glycophorin A antibodies in a cell separation composition ranges from 0.1 to 15 mg/L (e.g., 0.1 to 10 mg/L, 1 to mg/L, or 1 mg/L). Anti-glycophorin A antibodies can facilitate the removal of red cells from solution by at least two mechanisms. First, anti-glycophorin A
antibodies can cause homotypic agglutination of erythrocytes since glycophorin A is the major surface glycoprotein on erythrocytes. In addition, anti-glycophorin A antibodies also can stabilize dextran-mediated rouleau formation.
Exemplary monoclonal anti-glycophoiin A antibodies include, without limitation, 107FMN (Murine IgG1 isotype), YTH89.1 (Rat IgG2b isotype), 2.2.2.E7 (Murine IgM isotype; BioE, St. Paul, MN), and E4 (Murine IgM
isotype). See e.g., M. Vanderlaan et al., Molecular Immunology 20:1353 (1983);

Telen M. J. and Bolk, T. A., Transfusion 27: 309 (1987); and Outram S. et al., Leukocyte Research. 12:651 (1988).
The concentration of anti-CD15 antibodies in a cell separation composition can range from 0.1 to 15 mg/L (e.g., 0.1 to 10, 1 to 5, or 1 mg/L).
Anti-CD15 antibodies can cause homotypic agglutination of granulocytes by crosslinking CD15 molecules that are present on the surface of granulocytes.
Anti CD15 antibodies also can cause homotypic and heterotypic agglutination of granulocytes with monocytes, NK-cells and B-cells by stimulating expression of adhesion molecules (e.g., L-selectin and beta-2 integrin) on the surface of granulocytes that interact with adhesion molecules on monocytes, NK-cells and B-cells. Heterotypic agglutination of these cell types can facilitate the removal of these cells from solution along with red cell components. Exemplary monoclonal anti-CD15 antibodies include, without limitation, AHN1.1 (Murine IgM isotype), FMC-10 (Murine IgM isotype), BU-28 (Murine IgM isotype), MEM-157 (Murine IgM isotype), MEM-158 (Murine IgM isotype), 324.3.B9 (Murine IgM isotype; BioE, St. Paul, MN), and MEM-167 (Murine IgM
isotype). See e.g., Leukocyte typing IV (1989); Leukocyte typing 11 (1984);
Leukocyte typing VI (1995); Solter D. et al., Proc. Natl. Acad. Sci. USA
75:5565 (1978); Kannagi R. et al., J. Biol. Chem. 257:14865 (1982); Magnani, J.
L. et al., Arch. Biochem. Biophys 233:501 (1984); Eggens I. et al., J. Biol.
Chem. 264:9476 (1989).
The concentration of anti-CD9 antibodies in a cell separation composition can range from 0.1 to 15, 0.1 to 10, 1 to 5, or 1 mg/L. Anti-CD9 antibodies can cause homotypic agglutination of platelets. Anti-CD9 antibodies also can cause heterotypic agglutination of granulocytes and monocytes via platelets that have adhered to the surface of granulocytes and monocytes. CD9 antibodies can promote the expression of platelet p-selectin (CD62P), CD41/61, CD31, and CD36, which facilitates the binding of platelets to leukocyte cell surfaces. Thus, anti-CD9 antibodies can promote multiple cell-cell linkages and thereby facilitate agglutination and removal from solution. Exemplary monoclonal anti-CD9 antibodies include, without limitation, MEM-61 (Murine IgG1 isotype), MEM-62 (Murine IgG1 isotype), MEM-192 (Murine IgM
isotype), FMC-8 (Murine IgG2a isotype), SN4 (Murine IgG1 isotype), 8.10.E7 (Murine IgM isotype; BioE, St. Paul, MN), and BU-16 (Murine IgG2a isotype).
See e.g., Leukocyte typing VI (1995); Leukocyte typing 11 (1984); Von dem Bourne A. E. G. Kr. and Moderman P. N. (1989) In Leukocyte typing IV (ed. W.
Knapp, et al), pp. 989 ¨ 92, Oxford University Press, Oxford; Jennings, L. K., et al. In Leukocyte typing V, ed. S. F. Schlossmann et al., pp. 1249 ¨ 51, Oxford University Press, Oxford (1995); Lanza F. et al., J. Biol. Chem. 266:10638 (1991); Wright et al., Immunology Today 15:588 (1994); Rubinstein E. et al., Seminars in Thrombosis and Hemostasis 21:10 (1995).
In some embodiments, a cell separation composition contains antibodies against CD41, which can selectively agglutinate platelets. In some embodiments, a cell separation composition contains antibodies against CD3, which can selectively agglutinate T-cells. In some embodiments, a cell separation composition contains antibodies against CD2, which can selectively agglutinate T-cells and NK cells. In some embodiments, a cell separation composition contains antibodies against CD72, which can selectively agglutinate B-cells. In some embodiments, a cell separation composition contains antibodies against CD16, which can selectively agglutinate NK cells and neutrophilic granulocytes. The concentration of each of these antibodies can range from 0.01 to 15 mg/L. Exemplary anti-CD41 antibodies include, without limitation, PLT-1 (Murine IgM isotype), CN19 (Murine IgGi isotype), and 8.7.C3 (Murine IgG1 isotype). Non-limiting examples of anti-CD3 antibodies include OKT3 (Murine IgGi), HIT3a (Murine IgG2a isotype), SK7 (Murine IgGi) and BC3 (Murine IgG2a). Non-limiting examples of anti-CD2 antibodies include 7A9 (Murine IgM isotype), T11 (Murine IgGI isotype), and Leu5b (Murine IgG2a Isotype). Non-limiting examples of anti-CD72 antibodies include BU-40 (Murine IgGi isotype) and BU-41 (Murine IgGi isotype). Non-limiting examples of anti-CD16 antibodies include 3G8 (Murine IgG).
As mentioned above, cell separation compositions can be formulated to selectively agglutinate particular blood cells. As an example, a cell separation composition containing antibodies against glycophorin A, CD15, and CD9 can facilitate the agglutination of erythrocytes, granulocytes, NK cells, B cells, and platelets. T cells, NK cells and rare precursor cells such as MLPC then can be recovered from solution. If the formulation also contained an antibody against CD3, T cells also could be agglutinated, and NK cells and rare precursors such as MLPC could be recovered from solution.
Cell separation compositions can contain antibodies against surface antigens of other types of cells (e.g., cell surface proteins of tumor cells).
Those of skill in the art can use routine methods to prepare antibodies against cell surface antigens of blood, and other, cells from humans and other mammals, including, for example, non-human primates, rodents (e.g., mice, rats, hamsters, rabbits and guinea pigs), swine, bovines, and equines.
Typically, antibodies used in the composition are monoclonal antibodies, which are homogeneous populations of antibodies to a particular epitope contained within an antigen. Suitable monoclonal antibodies are commercially available, or can be prepared using standard hybridoma technology. In particular, monoclonal antibodies can be obtained by techniques that provide for the production of antibody molecules by continuous cell lines in culture, including the technique described by Kohler, G. et al., Nature, 1975, 256:495, the human B-cell hybridoma technique (Kosbor et al., Immunology Today 4:72 (1983); Cole et al., Proc. Natl. Acad. Sci. USA 80:2026 (1983)), and the EBV-hybridoma technique (Cole et al., "Monoclonal Antibodies and Cancer Therapy," Alan R. Liss, Inc., pp. 77-96 (1983)).
Antibodies can be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD, and any subclass thereof. Antibodies of the IgG and IgM isotypes are particularly useful in cell separation compositions of the invention.
Pentameric IgM antibodies contain more antigen binding sites than IgG antibodies and can, in some cases (e.g., anti-glycophorin A and anti-CD15), be particularly useful for cell separation reagents. In other cases (e.g., anti-CD9 antibodies), antibodies of the IgG isotype are particularly useful for stimulating homotypic and/or heterotypic agglutination.
Antibodies against cell surface antigens can be provided in liquid phase (i.e., soluble). Liquid phase antibodies typically are provided in a cell separation composition at a concentration between about 0.1 and about 15 mg/1 (e.g., between 0.25 to 10, 0.25 to 1, 0.5 to 2, 1 to 2, 4 to 8, 5 to 10 mg/1).
Antibodies against cell surface antigens also can be provided in association with a solid phase (i.e., substrate-bound). Antibodies against different cell surface antigens can be covalently linked to a solid phase to promote crosslinking of cell surface molecules and activation of cell surface adhesion molecules. The use of substrate-bound antibodies can facilitate cell separation (e.g., by virtue of the mass that the particles contribute to agglutinated cells, or by virtue of properties useful for purification).
In some embodiments, the solid phase with which a substrate-bound antibody is associated is particulate. In some embodiments, an antibody is bound to a latex microparticle such as a paramagnetic bead (e.g., via biotin-avidin linkage, covalent linkage to COO groups on polystyrene beads, or covalent linkage to NH2 groups on modified beads). In some embodiments, an antibody is bound to an acid-etched glass particle (e.g., via biotin-avidin linkage). In some embodiments, an antibody is bound to an aggregated polypeptide such as aggregated bovine serum albumin (e.g., via biotin-avidin linkage, or covalent linkage to polypeptide COO groups or NH2 groups). In some embodiments, an antibody is covalently linked to a polysaccharide such as high molecular weight (e.g., >1,000,000 Mr) dextran sulfate. In some embodiments, biotinylated antibodies are linked to avidin particles, creating tetrameric complexes having four antibody molecules per avidin molecule. In some embodiments, antibodies are bound to biotinylated agarose gel particles (One Cell Systems, Cambridge, MA, U.S.A.) via biotin-avidin-biotinylated antibody linkages. Such particles typically are about 300-500 microns in size, and can be created in a sonicating water bath or in a rapidly mixed water bath.
Cell-substrate particles (i.e., particles including cells and substrate-bound antibodies) can sediment from solution as an agglutinate. Cell-substrate particles also can be removed from solution by, for example, an applied magnetic field, as when the particle is a paramagnetic bead. Substrate-bound antibodies typically are provided in a cell separation composition at a concentration between about 0.1 and about 50.0 x 109 particles/1 (e.g., between 0.25 to 10.0 x 109, 1 to 20.0 x 109, 2 to 10.0 x 109, 0.5 to 2 x 109, 2 to 5 x 109, 5 to 10 x 109, and 10 to 30 x 109 particles/1), where particles refers to solid phase particles having antibodies bound thereto.
Cell separation compositions also can contain divalent cations (e.g., Ca+2 and Mg+2). Divalent cations can be provided, for example, by a balanced salt solution (e.g., Hank's balanced salt solution). Ca+2 ions reportedly are important for selectin-mediated and integrin-mediated cell-cell adherence.
Cell separation compositions also can contain an anticoagulant such as heparin. Heparin can prevent clotting and non-specific cell loss associated with clotting in a high calcium environment. Heparin also promotes platelet clumping. Clumped platelets can adhere to granulocytes and monocytes and thereby enhance heterotypic agglutination more so than single platelets.
Heparin can be supplied as a heparin salt (e.g., sodium heparin, lithium heparin, or potassium heparin).
Populations and Clonal Lines of MLPC
MLPC can be purified from human fetal blood using a cell separation composition described above. As used herein, "purified" means that at least 90% (e.g., 91, 92, 93, 94, 95, 96, 97, 98, or 99%) of the cells within the population are MLPC. As used herein, "MLPC" refers to fetal blood cells that are positive for CD9 and typically display a constellation of other markers such as CD13, CD73, and CD105. "MLPC population" refers to the primary culture obtained from the human fetal blood and uncloned progeny thereof. "Clonal line" refers to a cell line derived from a single cell. As used herein, a "cell line"
is a population of cells able to renew themselves for extended periods of times in vitro under appropriate culture conditions. The term "line," however, does not indicate that the cells can be propagated indefinitely. Rather, clonal lines described herein typically can undergo 75 to 100 doublings before senescing.
Typically, an MLPC population is obtained by contacting a fetal blood sample with a cell separation composition described above and allowing the sample to partition into an agglutinate and a supernatant phase.
For example, the sample can be allowed to settle by gravity or by centrifugation. Preferably, MLPC are purified from an umbilical cord blood sample that is less than 48 hours old (e.g., less than 24, 12, 8, or 4 hours post-partum). After agglutination, unagglutinated cells can be recovered from the supernatant phase. For example, cells in the supernatant phase can be recovered by centrifugation then washed with a saline solution and plated on a solid substrate (e.g., a plastic culture device such as a chambered slide or culture flask), using a standard growth medium with 10% serum (e.g., DMEM with 10% serum; RPMI-1640 with 10% serum, or mesenchymal stem cell growth medium with 10% serum (catalog # PT-3001, Cambrex, Walkersville, MD). MLPC attach to the surface of the solid substrate while other cells, including T cells, NK cells and CD34+ HSC, do not and can be removed with washing. The MLPC change from the leukocyte morphology to the fibroblastic morphology between 3 days and 2 weeks post initiation of culture after which the cells enter logarithmic growth phase and will continue growing logarithmically as long as cultures are maintained at cell concentrations of less than about 1.5 x105 cells/cm2.
Clonal lines can be established by harvesting the MLPC then diluting and re-plating the cells on a multi-well culture plate such that a single cell can be found in a well. Cells can be transferred to a larger culture flask after a concentration of 1 to 5 x 105 cells/75cm2 is reached.
Cells can be maintained at a concentration between 1 x 105 and 5 x 105 cells/75cm2 for logarithmic growth.
MLPC can be assessed for viability, proliferation potential, and longevity using techniques known in the art. For example, viability can be assessed using trypan blue exclusion assays, fluorescein diacetate uptake assays, or propidium iodide uptake assays. Proliferation can be assessed using thymidine uptake assays or MTT cell proliferation assays. Longevity can be assessed by determining the maximum number of population doublings of an extended culture.
MLPC can be immunophenotypically characterized using known techniques. For example, the cell culture medium can be removed from the tissue culture device and the adherent cells washed with a balanced salt solution (e.g., Hank's balanced salt solution) and bovine serum albumin (e.g., 2% BSA).

Cells can be incubated with an antibody having binding affinity for a cell surface antigen such as CD9, CD45, CD13, C73, CD105, or any other cell surface antigen. The antibody can be detectably labeled (e.g., fluorescently or enzymatically) or can be detected using a secondary antibody that is detectably labeled. Alternatively, the cell surface antigens on MLPC can be characterized using flow cytometry and fluorescently labeled antibodies.
As described herein, the cell surface antigens present on MLPC can vary, depending on the stage of culture. Early in culture when MLPC display a leukocyte-like morphology, MLPC are positive for CD9 and CD45, SSEA-4 (stage-specific embryonic antigen-4), CD34, as well as CD13, CD29, CD41, CD44, CD73, CD90, CD105, stem cell factor, STRO-1 (a cell surface antigen expressed by bone marrow stromal cells), SSEA-3 (galactosylgloboside), and CD133, and are negative for CD15, CD38, glycophorin A (CD235a), and lineage markers CD2, CD3, CD4, CDS, CD7, CD8, CD10, CD11b, CD16, CD19, CD20, CD21, CD22, CD33, CD36, CD41, CD61, CD62E, CD72, HLA-DR, and CD102. After transition to the fibroblastic morphology, MLPC remain positive for CD9, CD13, CD29, CD73, CD90, and CD105, and become negative for CD34, CD41, CD45, stem cell factor, STRO-1, SSEA-3, SSEA-4, and CD133.
At all times during in vitro culture, the undifferentiated MLPC are negative for CD15, CD38, glycophorin A (CD235a), lineage markers CD2, CD3, CD4, CD5, CD7, CD8, CD10, CD11b, CD16, CD19, CD20, CD21, CD22, CD33, CD36, CD41, CD61, CD62E, CD72, HLA-DR, and CD102.
Bone marrow-derived MSC and MAPC as well as the cord blood-derived USSC have been described as being derived from a CD451CD34" cell population. MLPC are distinguished from those cell types as being a CD45/CD34 + derived cell. Additionally, the presence and persistence of CD9 on the fetal blood-derived MLPC at all stages of maturation further distinguishes MLPC from MSC and MAPC, which do not possess CD9 as a marker. MLPC, which share the hematopoietic markers CD45, CD133, CD90 and CD34 during their leukocyte morphology phase, can be distinguished from HSC by their obligate plastic adherence and the presence of rnesenchymal associated markers CD105, CD29, CD73, CD13 and embryonic associated markers SSEA-3 and SSEA-4. Additionally using currently available technology, HSC are unable to be cultured in vitro without further differentiation while MLPC can be expanded for many generations without differentiation. MLPC also differ from MSC and USSC by their more gracile in vitro culture appearance, thread-like cytoplasmic projections and their preference for low density culture conditions for optimal growth.
MLPC can be cryopreserved by suspending the cells (e.g. 5 x 106 to 2 x 107 cells/mL) in a cryopreservative such as dimethylsulfoxide (DMSO, typically 1 to 10%) or in fetal bovine serum, human serum, or human serum albumin in combination with one or more of DMSO, trehalose, and dextran. For example, (1) fetal bovine serum containing 10% DMSO; (2) human serum containing 10%
DMSO and 1% Dextran; (3) human serum containing 1% DMSO and 5%
trehalose; or (4) 20% human serum albumin, 1% DMSO, and 5% trehalose can be used to cryopreserve MLPC. After adding cryopreseivative, the cells can be frozen (e.g., to -90 C). In some embodiments, the cells are frozen at a controlled rate (e.g., controlled electronically or by suspending the cells in a bath of 70%
ethanol and placed in the vapor phase of a liquid nitrogen storage tank. When the cells are chilled to ¨90 C, they can be placed in the liquid phase of the liquid nitrogen storage tank for long term storage. Cryopreservation can allow for long-term storage of these cells for therapeutic use.
Differentiation of MLPC
MLPC are capable of differentiating into a variety of cells, including cells of each of the three embryonic germ layers (i.e., endoderm, ectoderm, and mesoderm). As used herein, "capable of differentiating" means that a given cell, or its progeny, can proceed to a differentiated phenotype under the appropriate culture conditions. For example, MLPC can differentiate into cells having an osteocytic phenotype, cells having an adipocytic phenotype, cells having a neurocytic phenotype, cells having a myocytic phenotype, cells having an endothelial phenotype, cells having a hepatocytic/pancreatic precursor phenotype (also known as an oval cell) as well as other cell types.
Differentiation can be induced using one or more differentiation agents, including without limitation, Ca2+, an epidermal growth factor (EGF), a platelet derived growth factor (PDGF), a keratinocyte growth factor (KGF), a transforming growth factor (TGF), cytokines such as an interleukin, an interferon, or tumor necrosis factor, retinoic acid, transferrin, hormones (e.g., androgen, estrogen, insulin, prolactin, triiodothyronine, hydrocortisone, or dexamethasone), sodium butyrate, TPA, DMSO, NMF (N-methyl formamide), DMF (dimethylformamide), or matrix elements such as collagen, laminin, heparan sulfate).
Determination that an MLPC has differentiated into a particular cell type can be assessed using known methods, including, measuring changes in morphology and cell surface markers (e.g., by flow cytometry or immunohistochemistry), examining morphology by light or confocal microscopy, or by measuring changes in gene expression using techniques such as polymerase chain reaction (PCR) or gene-expression profiling.
For example, MLPC can be induced to differentiate into cells having an osteocytic phenotype using an induction medium (e.g., Osteogenic Differentiation Medium, catalog # PT-3002, from Cambrex) containing dexamethasone, L-glutamine, ascorbate, andf3-glycerophosphate (Jaiswal et al., J. Biol. Chem. 64(2):295-312 (1997)). Cells having an osteocytic phenotype contain deposits of calcium crystals, which can be visualized, for example, using Alizarin red stain.
MLPC can be induced to differentiate into cells having an adipocytic phenotype using an induction medium (e.g., Adipogenic Differentiation Medium, catalog # PT-3004, from Cambrex) containing insulin, L-glutamine, dexamethasone, indomethacin, and 3-isobuty1-1-methyl-xanthine. Cells having an adipocytic phenotype contain lipid filled liposomes that can be visualized with Oil Red stain. Such cells also contain trigycerides, which fluoresce green with Nile Red stain (Fowler and Greenspan, Histochem. Cytochem. 33:833-836 (1985)).
MLPC can be induced to differentiate into cells having a myocytic phenotype using an induction medium (e.g., SkGMTm, catalog # CC-3160, from Cambrex) containing EGF, insulin, Fetuin, dexamethasone, and FGF¨basic (Wernet, et al., U.S. patent publication 20020164794 Al). Cells having a myocytic phenotype express fast skeletal muscle myosin and alpha actinin.
MLPC can be induced to differentiate into cells having a neural stem cell phenotype (neurospheres) using an induction medium (e.g., NPMMTm ¨ Neural Progenitor Maintenance medium, catalog #CC-3209, from Cambrex) containing human FGF-basic, human EGF, NSF-1, and FGF-4 and a culture device pre-coated with poly-D-lysine and laminin (e.g., from BD Biosciences Discovery Labware, catalog #354688). Once cells have been differentiated into neuro spheres, they can be further differentiated into motor neurons with the addition of brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), astrocytes with the addition of leukemia inhibitory factor (LW), retinoic acid and ciliary neurotrophic factor, and oligodendrocytes with the addition of 3,3',5-triiodo-L-thyronine (T3). Neurocytic differentiation can be confirmed by the expression of nestin, class III beta-tubulin (tubulin f3-4), glial fibrillary acidic protein (GFAP), and galactocerebroside (GalC). Neurospheres are positive for all such markers while some differentiated cell types are not. Differentiation into oligodendrocytes can be confirmed by positive staining for myelin basic protein (MBP).
MLPC can be induced to differentiate into cells having an endothelial phenotype using an endothelial growth medium (e.g., EGMTm -MV, catalog #
CC-3125, from Cambrex) containing heparin, bovine brain extract, epithelial growth factor (e.g., human recombinant epithelial growth factor), and hydrocortisone. Endothelial differentiation can be confirmed by expression of E-selectin (CD62E) and ICAM-2 (CD102).
MLPC can be induced to differentiate into cells having a hepatocyte/pancreatic precursor cell phenotype using an induction medium (e.g., HCMTm -hepatocyte culture medium, catalog # CC-3198, from Cambrex) containing ascorbic acid, hydrocortisone, transferrin, insulin, EGF, hepatocyte growth factor, FGF-basic, fibroblast growth factor-4, and stem cell factor.
Liver and pancreas cells share a common progenitor. Hepatocyte differentiation can be confirmed by expression of hepatocyte growth factor and human serum albumin. Pancreatic cell differentiation can be confirmed by production of insulin and pro-insulin.

Modified Populations of MLPC
MLPC can be modified such that the cells can produce one or more polypeptides or other therapeutic compounds of interest. To modify the isolated cells such that a polypeptide or other therapeutic compound of interest is produced, the appropriate exogenous nucleic acid must be delivered to the cells.
In some embodiments, the cells are transiently transfected, which indicates that the exogenous nucleic acid is episomal (i.e., not integrated into the chromosomal DNA). In other embodiments, the cells are stably transfected, i.e., the exogenous nucleic acid is integrated into the host cell's chromosomal DNA. The term "exogenous" as used herein with reference to a nucleic acid and a particular cell refers to any nucleic acid that does not originate from that particular cell as found in nature. In addition, the term "exogenous" includes a naturally occurring nucleic acid. For example, a nucleic acid encoding a polypeptide that is isolated from a human cell is an exogenous nucleic acid with respect to a second human cell once that nucleic acid is introduced into the second human cell. The exogenous nucleic acid that is delivered typically is part of a vector in which a regulatory element such as a promoter is operably linked to the nucleic acid of interest.
Cells can be engineered using a viral vector such as an adenovirus, adeno-associated virus (AAV), retrovirus, lentivirus, vaccinia virus, measles viruses, herpes viruses, or bovine papilloma virus vector. See, Kay et al.
(1997) Proc. Natl. Acad. Sci. USA 94:12744-12746 for a review of viral and non-viral vectors. A vector also can be introduced using mechanical means such as liposomal or chemical mediated uptake of the DNA. For example, a vector can be introduced into an MLPC by methods known in the art, including, for example, transfection, transformation, transduction, electroporation, infection, microinjection, cell fusion, DEAE dextran, calcium phosphate precipitation, liposomes, LIPOFECTINTm, lysosome fusion, synthetic cationic lipids, use of a gene gun or a DNA vector transporter.
A vector can include a nucleic acid that encodes a selectable marker.
Non-limiting examples of selectable markers include puromycin, adenosine deaminase (ADA), aminoglycoside phosphotransferase (neo, G418, APH), dihydrofolate reductase (DHFR), hygromycin-B-phosphtransferase, thymidine kinase (TK), and xanthin-guanine phosphoribosyltransferase (XGPRT). Such markers are useful for selecting stable transformants in culture.
MLPC also can have a targeted gene modification. Homologous recombination methods for introducing targeted gene modifications are known in the art. To create a homologous recombinant MLPC, a homologous recombination vector can be prepared in which a gene of interest is flanked at its 5' and 3' ends by gene sequences that are endogenous to the genome of the targeted cell, to allow for homologous recombination to occur between the gene of interest carried by the vector and the endogenous gene in the genome of the targeted cell. The additional flanking nucleic acid sequences are of sufficient length for successful homologous recombination with the endogenous gene in the genome of the targeted cell. Typically, several kilobases of flanking DNA
(both at the 5' and 3' ends) are included in the vector. Methods for constructing homologous recombination vectors and homologous recombinant animals from recombinant stem cells are commonly known in the art (see, e.g., Thomas and Capecchi, 1987, Cell 51:503; Bradley, 1991, Curr. Opin. Bio/Technol. 2:823-29;
and PCT Publication Nos. WO 90/11354, WO 91/01140, and WO 93/04169.
Methods of Using MLPC
The MLPC can be used in enzyme replacement therapy to treat specific diseases or conditions, including, but not limited to lysosomal storage diseases, such as Tay-Sachs, Niemann-Pick, Fabry's, Gaucher's, Hunter's, and Hurler's syndromes, as well as other gangliosidoses, mucopolysaccharidoses, and glycogenoses.
In other embodiments, the cells can be used as carriers in gene therapy to correct inborn errors of metabolism, adrenoleukodystrophy, cystic fibrosis, glycogen storage disease, hypothyroidism, sickle cell anemia, Pearson syndrome, Pompe's disease, phenylketonuria (PKIJ), porphyrias, maple syrup urine disease, homocystinuria, mucoplysaccharide nosis, chronic granulomatous disease and tyrosinemia and Tay-Sachs disease or to treat cancer, tumors or other pathological conditions.

MLPC can be used to repair damage of tissues and organs resulting from disease. In such an embodiment, a patient can be administered a population of MLPC to regenerate or restore tissues or organs which have been damaged as a consequence of disease. For example, a population of MLPC can be administered to a patient to enhance the immune system following chemotherapy or radiation, or to repair heart tissue following myocardial infarction.
The cells also can be used in tissue regeneration or replacement therapies or protocols, including, but not limited to treatment of corneal epithelial defects, cartilage repair, facial dermabrasion, mucosal membranes, tympanic membranes, intestinal linings, neurological structures (e.g., retina, auditory neurons in basilar membrane, olfactory neurons in olfactory epithelium), burn and wound repair for traumatic injuries of the skin, or for reconstruction of other damaged or diseased organs or tissues.
MLPC also can be used in therapeutic transplantation protocols, e.g., to augment or replace stem or progenitor cells of the liver, pancreas, kidney, lung, nervous system, muscular system, bone, bone marrow, thymus, spleen, mucosal tissue, gonads, or hair.
Compositions and Articles of Manufacture The invention also features compositions and articles of manufacture containing purified populations of MLPC or clonal lines of MLPC. In some embodiments, the purified population of MLPC or clonal line is housed within a container (e.g., a vial or bag). In some embodiments, the clonal lines have undergone at least 3 doublings in culture (e.g., at least 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50 doublings). In other embodiments, a culture medium (e.g., MSCGMTm medium) is included in the composition or article of manufacture. In still other embodiments, the composition or article of manufacture can include one or more cryopreservatives or pharmaceutically acceptable carriers. For example, a composition can include serum and DMSO, a mixture of serum, DMSO, and trehalose, or a mixture of human serum albumin, DMSO, and trehalose.
Purified populations of MLPC or clonal MLPC lines can be combined with packaging material and sold as a kit. The packaging material included in a kit typically contains instructions or a label describing how the purified populations of MLPC or clonal lines can be grown, differentiated, or used.
Components and methods for producing such kits are well known.
The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
EXAMPLES
Example 1: Separating blood cells.
This example describes the general method by which cells were separated using the cell separation reagents described below. Equal volumes of a cell separation reagent (see Table 1) and an acid citrate dextrose (ACD), CPDA (citrate, phosphate, dextrose, adenine) or heparinized umbilical cord blood sample were combined (25 ml each) in a sterile closed container (e.g., a ml conical tube). Samples containing white blood cell counts greater than 20 x 106 cells / ml were combined one part blood with two parts cell separation reagent. Tubes were gently mixed on a rocker platform for 20 to 45 minutes at room temperature. Tubes were stood upright in a rack for 30 to 50 minutes to permit agglutinated cells to partition away from unagglutinated cells, which remained in solution. A pipette was used to recover unagglutinated cells from the supernatant without disturbing the agglutinate. Recovered cells were washed in 25 ml PBS and centrifuged at 500 x g for 7 minutes. The cell pellet was resuspended in 4 ml PBS +2% human serum albumin.
Table 1 Cell Separation Reagent Dextran (average molecular weight 413,000) 20 g/1 Dulbecco's phosphate buffered saline (10X) 100 m1/1 Sodium Heparin (10,000 units/10 1 m1/1 Hank's balanced salt solution (pH 7.2-7.4) 50 m1/1 Anti-human glycophorin A (murine IgM monoclonal 0.1 - 15 mg/L
antibody, clone 2.2.2.E7) (preferably about 0.25 mg/L) Anti-CD15 (murine IgM monoclonal antibody, clone 0.1 - 15 mg/L
324.3.B9) (preferably about 2.0 mg/L) Anti-CD9 (murine IgM monoclonal antibody, clone 0.1 - 15 mg/L
8.10.E7) (preferably about 2.0 mg/L) Cells also were recovered from the agglutinate using a hypotonic lysing solution containing EDTA and ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA). Agglutinated cells were treated with 25 ml VitaLyse (BioE, St. Paul, MN) and vortexed. After 10 minutes, cells were centrifuged at 500 x g for 7 minutes and the supernatant was removed. Cells were resuspended in 4 ml PBS.
Recoveries of erythrocytes, leukocytes, lymphocytes, monocytes, granulocytes, T cells, B cells, NK cells, hematopoietic stem cells, and non-hematopoietic stem cells were determined by standard flow cytometry and immunophenotyping. Prior to flow cytometry, leukocyte recovery (i.e., white blood cell count) was determined using a Coulter Onyx Hematology Analyzer.
Cell types were identified and enumerated by combining hematology analysis with flow cytometry analysis, identifying cells on the basis of light scattering properties and staining by labeled antibodies.
As shown in Table 2, 99.9% of erythrocytes were removed, 99.8%
monocytes and granulocytes, 74% of B cells, 64.9% of NK cells, and 99.4% of the platelets were removed from the cord blood.
Table 2 Recovery of Cells Before separation After separation Erythrocytes per ml 4.41 x 109 0.006x 109 Leukocytes per ml 5.9 x 106 1.53 x 106 Lymphocytes (%) 28.7 99.0 Monocytes (%) 8.69 0.12 Granulocytes (%) 62.5 .083 T Cells (CD3+) 19.7 83.2 B Cells (CD19+) 4.46 8.10 NK Cells (CD16+) 3.15 8.43 Platelets per ml 226 x 106 1.4 x 106 Example 2: Purification of MLPC.
The cell separation reagent of Table 3 was used to isolate MLPC from the non-agglutinated supernatant phase. See FIG 1 for a schematic of the purification.
Table 3 Cell Separation Reagent Dextran (average molecular weight 413,000) 20 g/1 Dulbecco's phosphate buffered saline (10X) 100 m1/1 Sodium Heparin (10,000 units/nil) 1 m1/1 Hank's balanced salt solution (pH 7.2-7.4) 50 m1/1 Anti-human glycophorin A (murine IgM monoclonal 0.1 - 15 mg/L
antibody, clone 2.2.2.E7) (preferably about 0.25 mg/L) Anti-CD15 (murine IgM monoclonal antibody, clone 0.1 - 15 mg/L
324.3.B9) (preferably about 2.0 mg/L) Anti-CD9 (murine IgM monoclonal antibody, clone 0.1 - 15 mg/L
8.10.E7) (preferably about 2.0 mg/L) Briefly, 50-150 ml of CPDA anti-coagulated umbilical cord blood (<48 hours old) was gently mixed with an equal volume of cell separation composition described in Table 3 for 30 minutes. After mixing was complete, the container holding the blood/cell separation composition mixture was placed in an upright position and the contents allowed to settle by normal 1 x g gravity for 30 minutes. After settling was complete, the non-agglutinated cells were collected from the supernatant. The cells were recovered from the supernatant by centrifugation then washed with PBS. Cells were resuspended in complete MSCGMTm (Mesenchymal stem cell growth medium, catalog # PT-3001, Cambrex, Walkersville, MD) and adjusted to 2-9 x 106 cells/ml with complete MSCGMTm. Cells were plated in a standard plastic tissue culture flask (e.g., Coming), chambered slide, or other culture device and allowed to incubate overnight at 37 C in a 5% CO2 humidified atmosphere. All subsequent incubations were performed at 37 C in a 5% CO2 humidified atmosphere unless otherwise noted. MLPC attached to the plastic during this initial incubation.
Non-adherent cells (T-cells, NK-cells and CD34+ hematopoietic stem cells) were removed by vigorous washing of the flask or well with complete MSCGMTm .
MLPC cultures were fed periodically by removal of the complete MSCGMTm and addition of fresh complete MSCGMTm . Cell were maintained at concentrations of 1 x 105 ¨ 1 x 106 cells/75cm2 by this method. When cell cultures reached a concentration of 8 x 105 -1 x 106 cells/75cm2, cells were cryopreserved using 10% DMSO and 90% serum or expanded into new flasks.
Cells were recovered from the adherent cultures by removal of the complete MSCGMTm and replacement with PBS + 0.1% EGTA. Cells were incubated for 15-60 minutes at 37 C then collected from the flask and washed in complete MSCGMTm . Cells were then replated at 1 x 105 cells/mL. Cultures that were allowed to achieve confluency where found to have diminished capacity for both proliferation and differentiation. Subsequent to this finding, cultures were not allowed to achieve higher densities than 1 x 106 cells/75 cm2.
Example 3: Morphology of MLPC and Development to Fibroblastic Morphology Cord blood derived MLPC isolated and cultured according to Examples 1 and 2 were cultured in standard MSCGMTm until confluency.
Depending on the donor, MLPC cultures achieved confluency in 2-8 weeks.
The morphology of these cells during growth and cultural maturation is shown in FIG 2A-2D.
In the early stage shown in FIG 2A, the cells are dividing very slowly and resemble circulating leukocytes but with dendritic cytoplasmic extensions. Many cells still exhibit the small round cell morphology that these cells would exhibit in circulation. As culture continues, the leukocyte-like cells start to change their morphology from the leukocyte-like appearance to a flatter, darker more fibroblast-like appearance (see FIG 2B).
When cells are dividing, they round up, divide, and then reattach to the culture vessel surface and spread out again. This slowly continues until the cells fill the available surface. FIG 2C shows the morphology of cell cultures during logarithmic growth. FIG 2D shows the morphology of a fully confluent culture of MLPC. With the exception of the two cells in active division seen in the lower left corner of the picture, all of the cells have a fibroblast-like morphology.
In summary, early during culture, cells appeared small and round, but had cytoplasmic projections, both finger-like and highly elongate projections, which help distinguish them from the other blood cells. Shortly after the initiation of the culture, the cells began to spread and flatten, taking on a morphology consistent with fibroblasts. Eventually, upon confluency, the cells grew in largely parallel orientation. Repeated growth of cultures to continency resulted in their having diminished proliferation and differentiating capacity.
Example 4: Inununophenotyping of Cells by Inununofluorescent Microscopy In order to determine the surface markers present on MLPC, freshly isolated cells were plated in 16 well chamber slides and grown to confluency.
At various times during the culture (from 3 days post plating to post confluency), cells were harvested and stained for the following markers: CD45-FITC
(BD/Pharmingen), CD34-PE (BD/Pharmingen), CD4-PE (BioE), CD8-PE
(BioE), anti-HLA-DR-PE (BioE), CD41-PE (BioE), CD9-PE (Aileen), CD105-PE (Ancell), CD29-PE (Coulter), CD73-PE (BD/Pharmingen), CD9O-PE
(BD/Pharmingen), anti-hu Stem Cell Factor-FITC (R&D Systems), CD14-PE
(BD/Pharmingen), CD15-FITC (Ancell), CD38-PE (BD/Pharmingen), CD2-PE
(BD/Pharmingen), CD3-FITC (BD/Pharmingen), CDS-PE (BD/Pharmingen), CD7-PE (BD/Pharmingen), CD16-PE (BD/Pharmingen), CD2O-FITC
(BD/Pharmingen), CD22-FITC (BD/Pharmingen), CD19-PE (BD/Pharmingen), CD33-PE (BD/Pharmingen), CD1O-FITC (BD/Pharmingen), CD61-FITC
(BD/Pharmingen), CD133-PE (R&D Systems), anti-STRO-1 (R&D Systems) and Goat anti-mouse IgG(H+L)-PE (BioE), SSEA-3 (R&D Systems) and goat anti-rat IgG (H+L)-PE (BioE), SSEA-4 (R&D Systems) and goat anti-mouse IgG(H+L)-PE (BioE). The cell surface markers also were assessed in bone marrow MSC (Cambrex, Walkersville, MD) and cord blood HSC (obtained from the non-adherent cells described above).
Briefly, cell culture medium was removed from the wells and the cells were washed 3X with Hank's Balanced Salt Solution +2% BSA. Cells were then stained with the antibodies for 20 minutes in the dark at room temperature.
After incubation, the cells were washed 3X with Hank's Balanced Salt Solution + 2% BSA and the cells were directly observed for fluorescence by fluorescent microscopy. Results obtained comparing cord blood derived MLPC with bone marrow-derived MSC's and cord blood derived hematopoietic stem cells (HSC) are outlined in Table 4.

Cell Early MLPC Mature MLPC Cord Bone Marker (Leukocyte (Fibroblast Blood Marrow morphology) morphology) HSC MSC
CD2 Negative Negative Negative Negative CD3 Negative Negative Negative Negative CD4 Negative Negative Negative Negative CD5 Negative Negative Negative Negative CD7 Negative Negative Negative Negative CD8 Negative Negative Negative Negative CD9 Positive Positive Negative Negative CD10 Negative Negative Negative Negative CD13 Positive Positive Negative Positive CD14 Negative Negative Negative Negative CD15 Negative Negative Negative Negative CD16 Negative Negative Negative Negative CD19 Negative Negative Negative Negative CD20 Negative Negative Negative Negative CD22 Negative Negative Negative Negative CD29 Positive Positive Positive Positive .
CD33 Negative Negative Variable Negative CD34 Positive Negative Positive Negative CD36 Negative Negative Negative Negative CD38 Negative Negative Variable Negative CD41 Negative Negative Negative Negative CD45 Positive Negative Positive Negative CD61 Negative Negative Variable Negative CD73 Positive Positive Negative Positive Anti- Negative Negative Variable Negative HLA-DR
CD90 Positive Positive Positive Positive CD105 Positive Positive Negative Positive STRO-1 Positive Negative Negative Negative SSEA-3 Positive Negative Negative Negative SSEA-4 Positive Negative Negative Negative SCF Positive Negative Negative Negative Glycoph Negative Negative Negative Negative orin A
CD133 Positive Negative Positive Negative Example 5: Clonal MLPC Cell Lines After the second passage of MLPC cultures from Example 2, the cells were detached from the plastic surface of the culture vessel by substituting PBS containing 0.1% EGTA (pH 7.3) for the cell culture medium. The cells were diluted to a concentration of 1.3 cells/ml in complete MSCGMTm and distributed into a 96 well culture plate at a volume of 0.2 ml/well, resulting in an average distribution of approximately 1 cell/3 wells. After allowing the cells to attach to the plate by overnight incubation at 37 C, the plate was scored for actual distribution. Only the wells with 1 cell/well were followed for growth. As the cells multiplied and achieved concentrations of 1-5 x 105 cells/75cm2, they were transferred to a larger culture vessel in order to maintain the cells at a concentration between 1 x 105 and 5 x 105 cells/75cm2 to maintain logarithmic growth.
Cells were cultured at 37 C in a 5% CO2 atmosphere.
At least 52 clonal cell lines have been established using this procedure and were designated: UM081704-1-E2, UM081704-1-B6, UM081704-1-G11, UM081704-1-G9, UM081704-1-E9, UM081704-1-E11, UM081704-1-G8, UM081704-1-H3, UM081704-1-D6, UM081704-1-H11, UM081704-1-B4, UM081704-1-H4, UM081704-1-C2, UM081704-1-G1, UM081704-1-E10, UM081704-1-B7, UM081704-1-G4, UM081704-1-F12, UM081704-1-H1, UM081704-1-D3, UM081704-1-A2, UM081704-1-B11, UM081704-1-D5, UM081704-1-E4, UM081704-1-C10, UM081704-1-A5, UM081704-1-E8, UM081704-1-C12, UM081704-1-E5, UM081704-1-Al2, UM081704-1-05, UM081704-1-A4, UM081704-1-A3, MH091404-2 #1-1.G10, UM093004-1-A3, UM093004-1-B7, UM093004-1-F2, UM093004-1-Al2, UM093004-1-G11, UM093004-1-G4, UM093004-1-B12, UM093004-2-A6, UM093004-2-A9, UM093004-2-B9, UM093004-2-05, UM093004-2-D12, UM093004-2-H3, UM093004-2-H11, UM093004-2-H4, UM093004-2-A5, UM093004-2-C3, and 1JM093004-2-C10. The surface markers of clonal cell line UM081704-1-E8 were assessed according to the procedure outlined in Example 4 and found to be the same as the "mature MLPC" having fibroblast morphology, as shown in Table 4.

Example 6: Osteocytic Differentiation of MLPC
A population of MLPC and clonal cell line UM081704-1-E8 each =
were cultured in complete MSCGMTm and grown under logarithmic growth conditions outlined above. Cells were harvested by treatment with PBS +
0.1% EGTA and replated at 5 x 103 to 2 x 104/m1 in complete MSCGMTm.
The cells were allowed to adhere overnight and then the medium was replaced with Osteogenic Differentiation Medium (catalog # PT-3002, Cambrex,) consisting of complete MSCGMTm supplemented with dexamethasone, L-glutamine, ascorbate, and P-glycerophosphate. Cells were cultured at 37 C in a 5% CO2 atmosphere and fed every 3-4 days for 2-3 weeks. Deposition of calcium crystals was demonstrated by using a modification of the Alizarin red procedure and observing red staining of calcium mineralization by phase contrast and fluorescent microscopy.
Example 7: Adipocytic Differentiation of MLPC
A population of MLPC and clonal cell line UM081704-1-E8 each were plated in complete MSCGMTm at a concentration of 1 x 104 to 2 x 105 cells/mL medium and cultured at 37 C in a 5% CO2 atmosphere. Cells were allowed to re-adhere to the culture plate and were fed every 3-4 days until the cultures reached confluency. At 100% confluency, cells were differentiated by culture in Adipogenesis differentiation medium (catalog #PT-3004, Cambrex) consisting of complete MSCGMTm supplemented with hu-insulin, L-glutamine, dexamethasone, indomethacin, and 3-isobuty1-1-methyl-xanthine, for at least 14 days.
To assess differentiation, the cells were stained with Oil Red stain specific for lipid. Confluent cultures of MLPC display a fibroblast-like morphology and do not display any evidence of liposome development as assessed by Oil Red staining. In contrast, MLPC differentiated with Adipogenic medium for 3 weeks exhibit liposomes that are characteristic of adipocytes (i.e., bright white vessels in cytoplasm) and that stain red with the Oil Red stain. MLPC differentiated with Adipogenic medium also fluoresce green with Nile Red stain specific for trigycerides.

Undifferentiated cells retain their fibroblast-like morphology and do not stain.
Example 8: Myocytic Differentiation of MLFC
MLPC (both a population and clonal cell line UM081704-1-E8) were plated in complete MSCGMTm at a concentration of 1.9 x 104 cells/well within a 4-chamber fibronectin pre-coated slide and allowed to attach to the plate for 24-48 hr at 37 C in a 5% CO2 atmosphere. Medium was removed and replaced with 10 M 5-azacytidine (catalog #A1287, Sigma Chemical Co.) and incubated for 24 hours. Cells were washed twice with PBS and fed with SkGMTm Skeletal Muscle Cell Medium (catalog #
CC-3160, Cambrex) containing recombinant human epidermal growth factor (huEGF), human insulin, Fetuin, dexamethasone, and recombinant human basic fibroblast growth factor (100 ng/mL) (huFGF-basic, catalog #
F0291, Sigma Chemical Co., St. Louis, MO). Cells were fed every 2-3 days for approximately 21 days. Control wells were fed with MSCGMTm while experimental wells were fed with SkGMTm (as described above).
Cultures were harvested 7 days post initiation of myocytic culture.
Culture supernatant was removed and cells were fixed for 2 hours with 2%
buffered formalin. Cells were permeabilized with PermaCyte TM (BioE, St. Paul, MN) and stained with mouse monoclonal antibody specific for human fast skeletal myosin (MY-32, catalog #ab7784, Abeam, Cambridge, MA) or mouse monoclonal antibody specific for alpha actinin (BM 75.2, catalog #ab11008, Abeam). Cells were incubated with the primary antibody for 20 minutes, washed with PBS and counter stained with goat anti-mouse IgG (H+L)-PE (BioE, St. Paul, MN). The myocytic culture contained fast skeletal muscle myosin and alpha actinin, which is indicative of the transdifferentiation of MLPC to skeletal muscle cells.
Example 9: Neurocytic Differentiation of MLPC
Bone marrow derived hMSC (Cambrex), cord blood MLPC, and MLPC clonal cell line were grown under logarithmic growth conditions described above. Cells were harvested as described above and replated at 0.8 x 104 cells per chamber in 4-chamber slides that were pre-coated with poly-D-lysine and laminin (BD Biosciences Discovery Labware, catalog #354688) in 0.5 mL of NPMMTm (catalog #CC-3209, Cambrex) containing huFGF-basic, huEGF, brain-derived neurotrophic factor, neural survival factor-1, fibroblast growth factor-4 (20 ng/mL), and 200 mM GlutaMax I
Supplement (catalog #35050-061, Invitrogen, Carlsbad, CA). The medium was changed every 2-3 days for 21 days. Neurospheres developed after 4 to 20 days. Transformation of MLPC to neural lineage was confirmed by positive staining for nestin (monoclonal anti-human nestin antibody, MAB1259, clone 196908, R&D Systems), class III beta-tubulin (tubulin b-4) (monoclonal anti-neuron-specific class III beta-tubulin antibody, MAB1195, Clone TuJ-1, R&D Systems), glial fibrillary acidic protein (GFAP) (monoclonal anti-human GFAP, HG2b-GF5, clone GF5, Advanced Immunochemical, Inc.), and galactocerebroside (GalC) (mouse anti-human GalC monoclonal antibody MAB342, clone mGalC, Chemicon).
Cells were further differentiated into neurons by the addition of 10 ng/mL BDNF (catalog #B3795, Sigma Chemical Co.) and 10 ng/mL NT3 (catalog #N1905, Sigma Chemical Co.) to the neural progenitor maintenance medium and further culturing for 10-14 days. Neurospheres were further differentiated into astrocytes by the addition of 10-6M retinoic acid (catalog #R2625, Sigma Chemical Co.), 10 ng/mL LIF (catalog #L5158, Sigma Chemical Co.) and 10 ng/mL CNTF (catalog #C3710, Sigma Chemical Co.) to the neural progenitor maintenance medium and further culturing for 10-14 days. Neurospheres were further differentiated into oligodendrocytes by the addition of 10-6M T3 (catalog #T5516, Sigma Chemical Co.) to the neural progenitor maintenance medium and further culturing for 10-14 days. Differentiation to oligodendrocytes was confirmed by positive staining for myelin basic protein (MBP) (monoclonal anti-MBP, catalog #ab8764, clone B505, Abeam).
Example 10: Endothelial Differentiation of MLPC
MLPC were plated at 1.9 x 104 cells per well within a 4-chamber slide (2 cm2). Cells were fed with 1 ml of endothelial growth medium-microvasculature (EGMTm-MV, catalog #CC-3125, Cambrex) containing heparin, bovine brain extract, human recombinant epithelial growth factor and hydrocortisone. The cells were fed by changing the medium every 2-3 days for approximately 21 days. Morphological changes occurred within 7-days. Differentiation of MLPC '5 to endothelial lineage was assessed by 5 staining for CD62E [E-selectin, mouse anti-human CD62E monoclonal antibody, catalog #551145, clone 68-5H11, BD Pharmingen] and CD102 [ICAM-2, monoclonal anti-human ICAM-2, MAB244, clone 86911, R&D
Systems]. Control MLPC cultures grown in MSCGM for 14 days were negative for CD62E staining and CD102, while differentiated cultures were 10 positive for both CD62E and CD102.
Example 11: Differentiation of MLPC into Hepatocyte/Pancreatic Precursor Cells MLPC were plated at a concentration of 1 x 105 cells/cm2 in vitro in HCM' medium (catalog #CC-3198, Cambrex) containing ascorbic acid, hydrocortisone, transferrin, insulin, huEGF, recombinant human hepatocyte growth factor (40 ng/mL), huFGF-basic (20 ng/mL), recombinant human fibroblast growth factor-4 (20 ng/mL), and stem cell factor (40 ng/mL).
Cells were cultured for 29 or more days to induce differentiation to precursor cells of both hepatocytes and pancreatic cells lineage. MLPC
changed from a fibroblast morphology to a hepatocyte morphology, expressed cell surface receptors for Hepatocyte Growth Factor, and produced both human serum albumin, a cellular product of hepatocytes, and insulin, a cellular product of pancreatic islet cells, both confirmed by intracellular antibody staining on day 30.
OTHER EMBODIMENTS
While the invention has been described in conjunction with the foregoing detailed description and examples, the foregoing description and examples are intended to illustrate and not to limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the claims.

Claims (55)

CLAIMS:
1. A purified population of human fetal blood multi-lineage progenitor cells (MLPC), wherein said MLPC are positive for CD9, CD13, CD29, CD44, CD73, CD90 and CD105, and negative for CD10, CD34, CD41, CD45, Stro-1, Stage Specific Embryonic Antigen-3 (SSEA-3) and SSEA-4.
2. The purified population of MLPC of claim 1, wherein said MLPC display a fibroblast morphology.
3. The purified population of MLPC of claim 1 or 2, wherein said MLPC are obtained from cord blood.
4. The purified population of MLPC of any one of claims 1 to 3, wherein said MLPC are further negative for CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD16, CD19, CD20, CD22, CD33, CD36, CD38, CD61, CD62E, CD133, glycophorin-A, stem cell factor, and HLA-DR.
5. The purified population of MLPC of any one of claims 1 to 4, wherein said MLPC
adhere to a plastic surface when cultured.
6. The purified population of MLPC of any one of claims 1 to 5, wherein said MLPC are capable of differentiating into cells from all three embryonic germ layers.
7. The purified population of MLPC of any one of claims 1 to 5, wherein said MLPC are capable of differentiating into one or more of: cells having an osteocytic phenotype, cells having an adipocytic phenotype, cells having a neural stem cell phenotype, cells having a myocytic phenotype, cells having an endothelial phenotype, cells having a hepatocytic phenotype, and cells having a pancreatic phenotype.
8. The purified population of MLPC of any one of claims 1 to 7, wherein said MLPC
comprise an exogenous nucleic acid.
9. The purified population of MLPC of claim 8, wherein said exogenous nucleic acid encodes a polypeptide.
10. A clonal line of human fetal blood multi-lineage progenitor cells (MLPC), wherein said MLPC are positive for CD9, CD13, CD29, CD44, CD73, CD90 and CD105, and negative for CD10, CD34, CD41, CD45, Stro-1, Stage Specific Embryonic Antigen-3 (SSEA-3) and SSEA-4.
11. The clonal line of claim 10, wherein said MLPC display a fibroblast morphology.
12. The clonal line of claim 10 or 11, wherein said MLPC are obtained from cord blood.
13. The clonal line of any one of claims 10 to 12, wherein said MLPC are further negative for CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD16, CD19, CD20, CD22, CD33, CD36, CD38, CD61, CD62E, CD133, glycophorin-A, stem cell factor, and HLA-DR.
14. The clonal line of any one of claims 10 to 13, wherein said MLPC adhere to a plastic surface when cultured.
15. The clonal line of any one of claims 10 to 14, wherein said MLPC are capable of differentiating into cells from all three embryonic germ layers.
16. The clonal line of any one of claims 10 to 14, wherein said MLPC are capable of differentiating into one or more of: cells having an osteocytic phenotype, cells having an adipocytic phenotype, cells having a neural stem cell phenotype, cells having a myocytic phenotype, cells having an endothelial phenotype, cells having a hepatocytic phenotype, and cells having a pancreatic phenotype.
17. The clonal line of any one of claims 10 to 16, wherein said MLPC
comprise an exogenous nucleic acid.
18. The clonal line of claim 17, wherein said exogenous nucleic acid encodes a polypeptide.
19. The clonal line of any one of claims 10 to 18, wherein said MLPC have undergone at least 5 doublings in culture.
20. The clonal line of any one of claims 10 to 18, wherein said MLPC have undergone at least 8 doublings in culture.
21. The clonal line of any one of claims 10 to 18, wherein said MLPC have undergone at least 10 doublings in culture.
22. The clonal line of any one of claims 10 to 18, wherein said MLPC have undergone at least 25 doublings in culture.
23. A composition comprising the purified population of MLPC of any one of claims 1 to 9, or the clonal line of any one of claims 10 to 22 and a culture medium.
24. The composition of claim 23, wherein said composition further comprises a cryopreservative.
25. The composition of claim 24, wherein said cryopreservative is dimethylsulfoxide (DMSO).
26. The composition of claim 25, wherein said cryopreservative is 1 to 10%
DMSO.
27. The composition of claim 24, wherein said cryopreservative is fetal bovine serum, human serum, or human serum albumin in combination with one or more of the following:
DMSO, trehalose, and dextran.
28. The composition of claim 27, wherein said cryopreservative is human serum, DMSO, and trehalose.
29. The composition of claim 27, wherein said cryopreservative is fetal bovine serum and DMSO.
30. An article of manufacture comprising the purified population of MLPC of any one of claims 1 to 9, or the clonal line of any one of claims 10 to 22, wherein said purified population of MLPC or said clonal line is housed within a container.
31. The article of manufacture of claim 30, wherein said container is a vial.
32. The article of manufacture of claim 30, wherein said container is a bag.
33. The article of manufacture of claim 30, wherein said container further comprises a cryopreservative.
34. A
method for purifying a population of multi-lineage progenitor cells (MLPC) from human fetal blood, said method comprising:
a) contacting a human fetal blood sample with a composition, said composition comprising:
i) dextran;
ii) anti-glycophorin A antibody;
iii) anti-CD15 antibody; and iv) anti-CD9 antibody;
b) allowing said sample to partition into an agglutinate and a supernatant phase;
c) recovering cells from said supernatant phase;
d) purifying MLPC from the recovered cells by adherence to a solid substrate, wherein said MLPC are positive for CD9 and positive for CD45; and e) culturing said MLPC such that said MLPC obtain a fibroblast morphology, wherein said MLPC after obtaining said fibroblast morphology are positive for CD9, CD13, CD29, CD44, CD73, CD90 and CD105, and negative for CD10, CD34, CD41, CD45, Stro-1, Stage Specific Embryonic Antigen-3 (SSEA-3) and SSEA-4.
35. The method of claim 34, wherein said fetal blood sample is cord blood.
36. The method of claim 34 or 35, wherein said solid substrate is a plastic substrate.
37. The method of any one of claims 34 to 36, wherein said MLPC, after obtaining said fibroblast morphology, are further negative for CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD16, CD19, CD20, CD22, CD33, CD36, CD38, CD61, CD62E, CD133, glycophorin-A, stem cell factor, and HLA-DR.
38. The method of any one of claims 34 to 37, further comprising testing said MLPC for CD9.
39. The method of claim 38, further comprising testing said MLPC for CD29, CD45, CD73, and CD90.
40. A method for cryopreserving MLPC, said method comprising:
a) contacting said purified population of MLPC of any one of claims 1 to 9, or said clonal line of any one of claims 10 to 22 with a cryopreservative; and b) freezing said purified population of MLPC or said clonal line.
41. The method of claim 40, wherein said cryopreservative is DMSO.
42. The method of claim 40, wherein said cryopreservative is fetal bovine serum, human serum, or human serum albumin in combination with one or more of the following: DMSO, trehalose, and dextran.
43. The method of claim 42, wherein said cryopreservative is human serum, DMSO, and trehalo se.
44. The method of claim 42, wherein said cryopreservative is fetal bovine serum and DMSO.
45. The method of any one of claims 40 to 44, wherein said purified population of MLPC
or said clonal line is suspended in said cryopreservative at a concentration between 1 x 10 5 and 5 x 10 7 cells/mL.
46. The method of any one of claims 40 to 45, wherein said purified population of MLPC
or said clonal line is frozen at a controlled rate.
47. The method of claim 46, wherein the freezing rate is controlled electronically.
48. The method of any one of claims 40 to 47, wherein said purified population of MLPC
or said clonal line is frozen by placement in an ethanol bath in the vapor phase of a liquid nitrogen cryogenic storage tank.
49. A method of producing a population of differentiated cells, said method comprising culturing the purified population of MLPC of any one of claims 1 to 9, or the clonal line of any one of claims 10 to 22 with an agent effective to induce differentiation of said MLPC.
50. The method of claim 49, wherein said agent comprises insulin, glutamine, dexamethasone, indomethacin, and 3 -isobutyl-1-methyl-xanthine.
51. The method of claim 49, wherein said agent comprises dexamethasone, glutamine, ascorbate, and .beta.-glycerophosphate.
52. The method of claim 49, wherein said agent comprises epithelial growth factor, insulin, fetuin, dexamethasone, and fibroblast growth factor-basic.
53. The method of claim 49, wherein said agent comprises fibroblast growth factor-basic, epidermal growth factor, NSF-1, and retinoic acid.
54. The method of claim 49, wherein said agent comprises heparin, bovine brain extract, epithelial growth factor, and hydrocortisone.
55. The method of claim 49, wherein said agent comprises ascorbic acid, hydrocortisone, transferrin, insulin, epidermal growth factor, hepatocyte growth factor, fibroblast growth factor-basic, fibroblast growth factor-4, and stem cell factor.
CA2563518A 2004-04-23 2005-04-20 Multi-lineage progenitor cells Expired - Fee Related CA2563518C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US56468704P 2004-04-23 2004-04-23
US60/564,687 2004-04-23
PCT/US2005/013244 WO2005108559A2 (en) 2004-04-23 2005-04-20 Multi-lineage progenitor cells

Publications (2)

Publication Number Publication Date
CA2563518A1 CA2563518A1 (en) 2005-11-17
CA2563518C true CA2563518C (en) 2014-09-02

Family

ID=35320798

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2563518A Expired - Fee Related CA2563518C (en) 2004-04-23 2005-04-20 Multi-lineage progenitor cells

Country Status (8)

Country Link
US (2) US7670596B2 (en)
EP (1) EP1747265B1 (en)
CN (1) CN101080486B (en)
AT (1) ATE506431T1 (en)
AU (1) AU2005241008C1 (en)
CA (1) CA2563518C (en)
DE (1) DE602005027557D1 (en)
WO (1) WO2005108559A2 (en)

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2563518C (en) 2004-04-23 2014-09-02 Bioe, Inc. Multi-lineage progenitor cells
JP5174656B2 (en) 2005-04-21 2013-04-03 マサチューセッツ インスティテュート オブ テクノロジー Materials and methods for modifying an immune response against exogenous and endogenous immunogens, including syngeneic or non-syngeneic cells, tissues or organs
GB0509500D0 (en) * 2005-05-10 2005-06-15 Revealcyte Method of fetal cell enrichment
CN102743792B (en) * 2005-06-21 2014-09-24 夏尔再生医学公司 Methods and compositions for enhancing vascular access
WO2007048813A2 (en) * 2005-10-28 2007-05-03 Universität Zürich Tissue engineering using pure populations of isolated non-embryoblastic fetal cells
WO2007080590A2 (en) 2006-01-11 2007-07-19 Technion Research & Development Foundation Ltd. Human embryonic stem cell-derived connective tissue progenitors for tissue engineering
EP2019858B1 (en) * 2006-04-17 2012-06-13 BioE LLC Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US7875453B2 (en) * 2006-06-14 2011-01-25 Bioe Llc Differentiation of multi-lineage progenitor cells to hepatocytes
ES2583371T3 (en) 2006-10-11 2016-09-20 The General Hospital Corporation Compositions, methods, and devices to treat liver diseases
US7883698B2 (en) * 2007-01-17 2011-02-08 Maria Michejda Isolation and preservation of fetal hematopoietic and mesencymal system cells from non-controversial materials and/or tissues resulting from miscarriages and methods of therapeutic use
US8574567B2 (en) 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
EP2607477B1 (en) 2007-05-03 2020-09-23 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US20090053182A1 (en) * 2007-05-25 2009-02-26 Medistem Laboratories, Inc. Endometrial stem cells and methods of making and using same
CA2693827A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of multi-lineage progenitor cells to chondrocytes
KR101046188B1 (en) 2007-11-30 2011-07-04 주식회사 알앤엘바이오 Cell therapy of urinary incontinence containing decidual or adipose derived stem cells
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
US10220128B1 (en) 2008-11-06 2019-03-05 Allan R. Robinson Implanted cardiac device to treat heart failure
WO2010059848A2 (en) * 2008-11-24 2010-05-27 Bioe, Inc. Implantable compositions for repairing osteochondral defects
US20110008764A1 (en) * 2009-06-02 2011-01-13 Davinci Biosciences Llc Human gonadal stem cells
KR20120093407A (en) * 2009-12-02 2012-08-22 리서치 디벨럽먼트 파운데이션 Selection of stem cell clones with defined differentiation capabilities
WO2011106521A1 (en) * 2010-02-25 2011-09-01 Abt Holding Company Modulation of macrophage activation
US9260697B2 (en) 2010-07-09 2016-02-16 The Gid Group, Inc. Apparatus and methods relating to collecting and processing human biological material containing adipose
WO2013106655A1 (en) 2012-01-11 2013-07-18 The Gid Group, Inc. Method for processing adipose tissue and processing apparatus
US9296984B2 (en) 2010-07-09 2016-03-29 The Gid Group, Inc. Tissue processing apparatus and method for processing adipose tissue
US9206387B2 (en) 2010-07-09 2015-12-08 The Gid Group, Inc. Method and apparatus for processing adipose tissue
WO2012019002A2 (en) * 2010-08-04 2012-02-09 StemBios Technologies, Inc. Somatic stem cells
US8795149B2 (en) 2010-11-23 2014-08-05 Theodore J. Lillehei Pneumatic or hydraulic cardiac assist devices
WO2013177460A1 (en) 2012-05-23 2013-11-28 Lillehei Theodore J Pneumatic or hydraulic cardiac assist devices
EP3896145A1 (en) 2012-09-06 2021-10-20 GID BIO, Inc. Tissue processing apparatus and method for processing adipose tissue
WO2014093845A1 (en) 2012-12-14 2014-06-19 Bhc Technology Holdings Llc Point of care isolation and concentration of blood cells
US11261429B2 (en) 2013-05-20 2022-03-01 Icahn School Of Medicine At Mount Sinai Enriched and expanded human cord blood stem cells for treatment of hematological disorders
EP3038629B1 (en) 2013-09-05 2020-11-18 GID BIO, Inc. Method for processing adipose tissue
CN104222069B (en) * 2014-08-27 2016-06-29 中国人民解放军军事医学科学院野战输血研究所 CFU-E frozen stock solution and application thereof
CN105076116B (en) * 2015-09-17 2017-08-08 广州赛莱拉干细胞科技股份有限公司 A kind of cells frozen storing liquid and its application and the cryopreservation methods of megakaryoblast
US10456423B2 (en) 2016-06-13 2019-10-29 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
US10426796B2 (en) 2016-06-13 2019-10-01 SMART SURGICAL, Inc. Compositions for biological systems and methods for preparing and using the same
KR20190046844A (en) 2016-08-30 2019-05-07 라이프셀 코포레이션 System and method for medical device control
CN110072992A (en) 2016-12-14 2019-07-30 株式会社大塚制药工场 Mammalian cell freezen protective liquid
USD851777S1 (en) 2017-01-30 2019-06-18 Lifecell Corporation Canister-type device for tissue processing
WO2019018002A1 (en) 2017-07-18 2019-01-24 The Gid Group, Inc. Adipose tissue digestion system and tissue processing method
CN107557332B (en) * 2017-09-28 2021-02-26 吉林省拓华生物科技有限公司 CD29+Human umbilical cord-derived mesenchymal stem cells and application thereof in preparation of medicine for treating skeletal muscle atrophy in high-sugar and high-fat environment
CN107502587A (en) * 2017-09-28 2017-12-22 吉林省拓华生物科技有限公司 CD29+People's Mesenchymal Stem Cells from Umbilical Cord and its purposes in the Seeding Cells in Bone Tissue Engineering for preparing treatment bone injury
CN109619090A (en) * 2018-12-30 2019-04-16 深圳光彩生命工程技术有限公司 A kind of freeze drying process saving cell factor vigor

Family Cites Families (129)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5130144B1 (en) * 1984-02-06 1995-08-15 Univ Johns Hopkins Human stem cells and monoclonal antibodies
US5510254A (en) * 1986-04-18 1996-04-23 Advanced Tissue Sciences, Inc. Three dimensional cell and tissue culture system
US5744347A (en) * 1987-01-16 1998-04-28 Ohio University Edison Biotechnology Institute Yolk sac stem cells and their uses
US5192553A (en) 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5004681B1 (en) * 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5061620A (en) * 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US6967086B2 (en) 1991-04-18 2005-11-22 Becton Dickinson And Company Method for determining the presence or absence of respiring cells on a three-dimensional scaffold
WO1994000484A1 (en) 1992-06-22 1994-01-06 Young Henry E Scar inhibitory factor and use thereof
US5453357A (en) * 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US6432711B1 (en) * 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US5972703A (en) 1994-08-12 1999-10-26 The Regents Of The University Of Michigan Bone precursor cells: compositions and methods
US5789147A (en) * 1994-12-05 1998-08-04 New York Blood Center, Inc. Method for concentrating white cells from whole blood by adding a red cell sedimentation reagent to whole anticoagulated blood
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5580714A (en) 1995-03-08 1996-12-03 Celox Laboratories, Inc. Cryopreservation solution
US5830651A (en) 1995-06-01 1998-11-03 Signal Pharmaceuticals, Inc. Human oligodendroglial progenitor cell line
US5908782A (en) * 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US5877299A (en) * 1995-06-16 1999-03-02 Stemcell Technologies Inc. Methods for preparing enriched human hematopoietic cell preparations
US6117985A (en) 1995-06-16 2000-09-12 Stemcell Technologies Inc. Antibody compositions for preparing enriched cell preparations
US6306575B1 (en) 1995-06-16 2001-10-23 Stemcell Technologies, Inc. Methods for preparing enriched human hematopoietic cell preparations
US5753505A (en) * 1995-07-06 1998-05-19 Emory University Neuronal progenitor cells and uses thereof
CA2235069C (en) 1995-10-19 2010-12-14 Advanced Reproduction Technologies, Inc. Methods and compositions to improve germ cell and embryo survival and function
US5827740A (en) 1996-07-30 1998-10-27 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
GB9617058D0 (en) 1996-08-14 1996-09-25 Sandoz Ltd Organic compounds
US6890724B2 (en) * 1996-09-06 2005-05-10 California Institute Of Technology Methods and compositions for neural progenitor cells
AU5436998A (en) 1996-11-15 1998-06-03 Osiris Therapeutics, Inc. MSC-megakaryocyte precursor composition and method of isolating MSCs asso ciated with isolated megakaryocytes by isolating megakaryocytes
AU6869198A (en) * 1997-03-25 1998-10-20 Morphogenesis, Inc. Universal stem cells
US5968829A (en) 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
WO1999020741A1 (en) 1997-10-23 1999-04-29 Geron Corporation Methods and materials for the growth of primate-derived primordial stem cells
CA2311729A1 (en) * 1998-01-23 1999-07-29 Imclone Systems Incorporated Purified populations of stem cells
US6962698B1 (en) * 1998-02-17 2005-11-08 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
EP1062321B1 (en) * 1998-03-13 2004-12-29 Osiris Therapeutics, Inc. Uses for humane non-autologous mesenchymal stem cells
ES2258329T3 (en) 1998-03-18 2006-08-16 Osiris Therapeutics, Inc. MESENQUIMATIC MOTHER CELLS FOR THE PREVENTION AND TREATMENT OF IMMUNE ANSWERS IN TRANSPLANTS.
US6368636B1 (en) * 1998-03-18 2002-04-09 Osiris Therapeutics, Inc. Mesenchymal stem cells for prevention and treatment of immune responses in transplantation
US6835377B2 (en) 1998-05-13 2004-12-28 Osiris Therapeutics, Inc. Osteoarthritis cartilage regeneration
ES2292245T3 (en) * 1998-05-29 2008-03-01 Osiris Therapeutics, Inc. MESENCHIMATIC MOTHER CELLS CD45 + AND / OR FIBROBLASTO + HUMANAS.
CA2329519A1 (en) * 1998-06-08 1999-12-16 Osiris Therapeutics, Inc. In vitro maintenance of hematopoietic stem cells
CA2279474C (en) 1998-07-31 2011-01-04 Stemcell Technologies Inc. Novel antibody composition for debulking blood and bone marrow samples from cml patients
US5958767A (en) * 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
US6767737B1 (en) * 1998-08-31 2004-07-27 New York University Stem cells bearing an FGF receptor on the cell surface
US6184035B1 (en) 1998-11-18 2001-02-06 California Institute Of Technology Methods for isolation and activation of, and control of differentiation from, skeletal muscle stem or progenitor cells
PT1975243E (en) * 1998-12-07 2010-06-24 Univ Duke Bodipy aminoacetaldehyde diethyl acetal
US6242666B1 (en) * 1998-12-16 2001-06-05 The Scripps Research Institute Animal model for identifying a common stem/progenitor to liver cells and pancreatic cells
KR20020013496A (en) * 1999-02-04 2002-02-20 추후제출 Method and apparatus for maintenance and expansion of hemopoietic stem cells and(or) progenitor cells
US6946293B1 (en) 1999-02-10 2005-09-20 Es Cell International Pte Ltd. Progenitor cells, methods and uses related thereto
US6777231B1 (en) * 1999-03-10 2004-08-17 The Regents Of The University Of California Adipose-derived stem cells and lattices
US20030082152A1 (en) 1999-03-10 2003-05-01 Hedrick Marc H. Adipose-derived stem cells and lattices
WO2000073794A2 (en) * 1999-05-28 2000-12-07 Stemcell Technologies Inc. Method for separating cells using immunorosettes
US6750326B2 (en) * 1999-05-28 2004-06-15 Stemcell Technologies Inc. Method for separating cells using immunorosettes
US6761883B2 (en) * 1999-06-29 2004-07-13 The Board Of Trustees Of The Leland Stanford Junior University Mammalian myeloid progenitor cell subsets
EP1194035B1 (en) 1999-06-29 2007-09-19 The Board Of Trustees Of The Leland Stanford Junior University Mammalian myeloid progenitor cell subsets
US20050158289A1 (en) * 1999-07-07 2005-07-21 Simmons Paul J. Mesenchymal precursor cell and use thereof in the repair of bone defects and fractures in mammals
AUPQ147799A0 (en) 1999-07-07 1999-07-29 Medvet Science Pty. Ltd. Mesenchymal precursor cell
US7670628B2 (en) * 1999-07-07 2010-03-02 Angioblast Systems, Inc. Mesenchymal precursor cell
NZ516738A (en) * 1999-07-20 2004-01-30 Univ Southern California Identification of pluripotent pre-mesenchymal, pre-hematopoietic progenitor cells expressing a unique molecular marker
US7015037B1 (en) * 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US8075881B2 (en) * 1999-08-05 2011-12-13 Regents Of The University Of Minnesota Use of multipotent adult stem cells in treatment of myocardial infarction and congestive heart failure
US6280718B1 (en) * 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
IL149933A0 (en) 1999-12-06 2002-11-10 Gen Hospital Corp Pancreatic stem cells and their use in transplantation
WO2001042425A2 (en) 1999-12-07 2001-06-14 Childrens Hospital Los Angeles Research Institute Lung stem cells and lung regeneration
US7468277B2 (en) * 1999-12-23 2008-12-23 Cornell Research Foundation, Inc. Enriched preparation of human fetal multipotential neural stem cells
US7160724B2 (en) * 2000-03-09 2007-01-09 University Of South Florida Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US7011828B2 (en) 2000-03-14 2006-03-14 Es Cell International Pte. Ltd. Implanting neural progenitor cells derived for human embryonic stem cells
JP4889902B2 (en) * 2000-03-14 2012-03-07 イーエス・セル・インターナショナル・プライヴェート・リミテッド Method for producing human neural progenitor cells from human embryonic stem (hES) cells, method for producing neurons using the method, method for producing oligodendrocytes or astrocytes
US6436704B1 (en) 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
US7473555B2 (en) * 2000-04-27 2009-01-06 Geron Corporation Protocols for making hepatocytes from embryonic stem cells
US20020045196A1 (en) * 2000-05-12 2002-04-18 Walt Mahoney Methods of isolating trophoblast cells from maternal blood
US6645727B2 (en) 2000-05-26 2003-11-11 Stemcell Technologies Inc. Antibody compositions for preparing enriched mesenchymal progenitor preparations
US6936281B2 (en) * 2001-03-21 2005-08-30 University Of South Florida Human mesenchymal progenitor cell
US7049072B2 (en) 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
US7442390B2 (en) * 2000-06-05 2008-10-28 University Of South Florida Method for enhancing engraftment of cells using mesenchymal progenitor cells
JP4043703B2 (en) 2000-09-04 2008-02-06 株式会社ルネサステクノロジ Semiconductor device, microcomputer, and flash memory
US7560280B2 (en) * 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
AU2002243263A1 (en) * 2000-11-15 2002-07-24 Roche Diagnostics Corporation Methods and reagents for identifying rare fetal cells in the material circulation
AU2001297880B2 (en) 2000-11-30 2007-05-31 Stemron Inc. Isolated homozygous stem cells differentiated cells derived therefrom and materials and methods for making and using same
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
AU2020902A (en) * 2000-12-06 2002-06-18 Robert J Hariri Method of collecting placental stem cells background of the invention
EP1367899A4 (en) * 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
IL157350A0 (en) * 2001-02-14 2004-02-19 Anthrogenesis Corp Post-partum mammalian placenta, its use and placental stem cells therefrom
EP1916297B1 (en) * 2001-04-10 2010-12-29 BioE LLC Cell separation compositions and methods
US6841386B2 (en) 2001-04-10 2005-01-11 Viacell, Inc. Modulation of primary stem cell differentiation using an insulin-like growth factor binding protein
DK2105497T3 (en) * 2001-04-24 2019-06-24 Dolores Baksh Progenitor cell populations, their expansion, and growth of non-hematopoietic cell types and tissues therefrom
WO2003015802A1 (en) 2001-08-14 2003-02-27 Medipost, Co., Ltd. Composition for treatment of articular cartilage damage
US6740793B2 (en) * 2001-09-12 2004-05-25 Albert Einstein College Of Medicine Of Yeshiva University Transgenic animal having a disrupted PDE7A gene and uses thereof
US6949355B2 (en) * 2001-10-11 2005-09-27 Aviva Biosciences Methods, compositions, and automated systems for separating rare cells from fluid samples
US7166443B2 (en) * 2001-10-11 2007-01-23 Aviva Biosciences Corporation Methods, compositions, and automated systems for separating rare cells from fluid samples
WO2003033697A1 (en) * 2001-10-18 2003-04-24 Ixion Biotechnology, Inc. Conversion of liver stem and progenitor cells to pancreatic functional cells
US20040259254A1 (en) 2001-10-30 2004-12-23 Osamu Honmou Method for inducing differentiation mesodermal stem cells, es cells or immortalized cells into nervous system cells
EP1442115B9 (en) * 2001-11-15 2009-12-16 Children's Medical Center Corporation Methods of isolation, expansion and differentiation of fetal stem cells from chorionic villus, amniotic fluid, and placenta and therapeutic uses thereof
US8105580B2 (en) * 2001-12-07 2012-01-31 Cytori Therapeutics, Inc. Methods of using adipose derived stem cells to promote wound healing
BR0214772A (en) * 2001-12-07 2007-01-09 Macropore Biosurgery Inc systems and methods for treating patients with processed liposuction cells
US20030113910A1 (en) * 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
AU2002350352A1 (en) * 2001-12-21 2003-07-15 Mount Sinai Hospital Cellular compositions and methods of making and using them
CN1671835A (en) * 2001-12-28 2005-09-21 塞拉提斯股份公司 A method for the establishment of a pluripotent human blastocyst-derived stem cell line
WO2003068937A2 (en) * 2002-02-13 2003-08-21 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
US20030161818A1 (en) * 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
PL372619A1 (en) * 2002-03-15 2005-07-25 University Of North Carolina At Chapel Hill Primitive and proximal hepatic stem cells
AU2002315259A1 (en) * 2002-03-20 2003-09-29 Miltenyi Biotec Gmbh A method for generating human nervous system cells, tissues, or neural stem cell progenitors from haematopoietic stem cells
CA2481385A1 (en) * 2002-04-12 2003-10-23 Celgene Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
CA2483010A1 (en) 2002-04-19 2003-10-30 University Of Pittsburgh Of The Commonwealth System Of Higher Education Placental derived stem cells and uses thereof
US20040161419A1 (en) * 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
KR20050008757A (en) * 2002-05-30 2005-01-21 셀진 코포레이션 Methods of using jnk or mkk inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US7838290B2 (en) * 2002-07-25 2010-11-23 The Scripps Research Institute Hematopoietic stem cells and methods of treatment of neovascular eye diseases therewith
ATE539146T1 (en) * 2002-09-13 2012-01-15 Univ Leland Stanford Junior MAMMAL MEGAKARYOCYTE STEM CELL
US9969977B2 (en) * 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
AU2003275260B2 (en) * 2002-09-27 2008-01-24 Bioe, Inc. Cell separation compositions and methods
US20040121464A1 (en) * 2002-09-30 2004-06-24 Rathjen Peter David Method for the preparation of cells of mesodermal lineage
KR101042448B1 (en) 2002-11-26 2011-06-16 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
CA2515594A1 (en) 2003-02-13 2004-08-26 Anthrogenesis Corporation Use of umbilical cord blood to treat individuals having a disease, disorder or condition
EP1596659A2 (en) 2003-02-28 2005-11-23 Duke University Progenitor cells and methods of using same
US20060134636A1 (en) * 2003-03-13 2006-06-22 Stanton Lawrence W Standardization of growth conditions for human embryonic stem cells intended for use in regenerative medicine
US20040203142A1 (en) 2003-04-14 2004-10-14 Reliance Life Sciences Pvt. Ltd. Growth of neural precursor cells using umbilical cord blood serum and a process for the preparation thereof for therapeutic purposes
US7413734B2 (en) * 2003-06-27 2008-08-19 Ethicon, Incorporated Treatment of retinitis pigmentosa with human umbilical cord cells
EP1506997A1 (en) * 2003-08-14 2005-02-16 NeuroProgen GmbH Leipzig Method of generating neural stem cells
WO2005052138A1 (en) 2003-11-19 2005-06-09 Wisconsin Alumni Research Foundation Cryopreservation of pluripotent stem cells
US7534606B2 (en) * 2004-01-12 2009-05-19 National Health Research Institutes Placental stem cell and methods thereof
US20050176139A1 (en) * 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
WO2005085422A1 (en) 2004-02-27 2005-09-15 Michigan State University Adult stem cells and uses thereof
CA2563518C (en) 2004-04-23 2014-09-02 Bioe, Inc. Multi-lineage progenitor cells
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
WO2005113754A1 (en) 2004-05-20 2005-12-01 New York Medical College Pluripotent adult stem cells
ES2527293T3 (en) * 2004-08-16 2015-01-22 Cellresearch Corporation Pte Ltd Isolation of stem / progenitor cells of amniotic membrane of the umbilical cord
WO2006088867A2 (en) * 2005-02-15 2006-08-24 Medistem Laboratories, Incorporated Method for expansion of stem cells
EP2019858B1 (en) * 2006-04-17 2012-06-13 BioE LLC Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US7875453B2 (en) * 2006-06-14 2011-01-25 Bioe Llc Differentiation of multi-lineage progenitor cells to hepatocytes
CA2693827A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of multi-lineage progenitor cells to chondrocytes
CA2724839A1 (en) 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells

Also Published As

Publication number Publication date
CN101080486A (en) 2007-11-28
EP1747265A4 (en) 2007-12-26
WO2005108559A2 (en) 2005-11-17
EP1747265B1 (en) 2011-04-20
DE602005027557D1 (en) 2011-06-01
ATE506431T1 (en) 2011-05-15
US20050255592A1 (en) 2005-11-17
US20100028851A1 (en) 2010-02-04
EP1747265A2 (en) 2007-01-31
AU2005241008C1 (en) 2010-11-04
US7670596B2 (en) 2010-03-02
US8163275B2 (en) 2012-04-24
AU2005241008B2 (en) 2010-05-13
CN101080486B (en) 2012-05-16
CA2563518A1 (en) 2005-11-17
WO2005108559A3 (en) 2007-05-03
AU2005241008A1 (en) 2005-11-17

Similar Documents

Publication Publication Date Title
CA2563518C (en) Multi-lineage progenitor cells
US20120077271A1 (en) Differentiation of multi-lineage progenitor cells to chondrocytes
US7875453B2 (en) Differentiation of multi-lineage progenitor cells to hepatocytes
US7622108B2 (en) Multi-lineage progenitor cells
US7727763B2 (en) Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
EP2374871B1 (en) Pluripotent stem cells, method for preparation thereof and uses thereof
EP1458854B1 (en) Cellular compositions and methods of making and using them
EP2862925A1 (en) Identification and isolation of multipotent cells from non osteochondral mesenchymal tissue
US20110274729A1 (en) Implantable compositions for repairing osteochondral defects
KR20020013492A (en) Human liver progenitors
SG187114A1 (en) Culture medium composition for culturing amnion-derived mesenchymal stem cell, and method for culturing amnion-derived mesenchymal stem cell by using same
US20090291494A1 (en) Differentiation of Multi-Lineage Progenitor Cells to Pancreatic Cells
US20210009947A1 (en) Methods and materials for maintaining and expanding stem cell-derived hepatocyte-like cells

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20220420