US20040137612A1 - Progenitor cell populations , expansions thereof, and growth of non-hematopoietic cell types and tissues therefrom - Google Patents

Progenitor cell populations , expansions thereof, and growth of non-hematopoietic cell types and tissues therefrom Download PDF

Info

Publication number
US20040137612A1
US20040137612A1 US10/475,861 US47586103A US2004137612A1 US 20040137612 A1 US20040137612 A1 US 20040137612A1 US 47586103 A US47586103 A US 47586103A US 2004137612 A1 US2004137612 A1 US 2004137612A1
Authority
US
United States
Prior art keywords
cells
progenitor
cell
population
progenitor cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/475,861
Other languages
English (en)
Inventor
Dolores Baksh
John Davies
Peter Zandstra
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of US20040137612A1 publication Critical patent/US20040137612A1/en
Priority to US11/828,575 priority Critical patent/US9644182B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]

Definitions

  • the present invention relates to the identification and subsequent expansion of a new compartment of progenitor cells which give rise through differentiation to a wide variety of cell types that form tissues including bone, cartilage, fat, muscle, endothelium, adipose and neural cells, among others.
  • the invention further relates to methods by which such cell populations are expanded. Further, the invention relates to methods by which such cell types and tissues are formed, to the cell and tissue types resulting therefrom, and to applications of such progenitor and differentiated cells in cell-based therapy and tissue engineering.
  • the bone marrow compartment and particularly the marrow parenchyma, provides an attractive source for such progenitor cells.
  • the marrow parenchyma includes hematopoietic progenitor cells, which differentiate into blood cells of various types.
  • the narrow parenchyma also includes non-hematopoietic progenitor and stem cells, which differentiate into mesenchymal and other cell types, from which various structural and connective human tissues arise.
  • the mesenchymal progenitor and stem cells within the marrow parenchyma give rise, through differentiation, to osteogenic cell types that form bone, to chondrogenic cell, types that form cartilage, and to myogenic cell types that form muscle and other connective tissues such as bone, marrow stroma.
  • Parenchymal progenitors further include cells that give rise to other non-hematopoietic cell types and tissues, which include adipocytes that form adipose tissue, neurons and other neural cells that form nerve tissue, and cells capable of giving rise to endothelium and related vasculature.
  • the present invention is thus concerned with progenitor and stem cells of the type that are extractable, for instance, from marrow parenchyma, and which give rise to non-hematopoietic cell types. These cells can be referred to as non-hematopoietic progenitors, or NHP cells.
  • NHP cells include the mesenchymal progenitor cells (MPCs) and progenitor cells that give rise to other non-blood cell types, and thus as a class, give rise to a wide variety of cell types and tissues of therapeutic interest including nerve tissue, connective tissue, muscle, tendon/ligament, bone, cartilage, adipose tissue and vascular endothelium.
  • MPCs mesenchymal progenitor cells
  • progenitor cells that give rise to other non-blood cell types, and thus as a class, give rise to a wide variety of cell types and tissues of therapeutic interest including nerve tissue, connective tissue, muscle, tendon/ligament, bone, cartilage, ad
  • the NHPs including the MPCs can be expanded without differentiating.
  • the population can be increased from the small numbers first obtained from the bone marrow or other sources, to the large numbers required for their medical use, without concomitantly inducing the cells to differentiate or become committed to development toward a particular cell lineage.
  • research has accordingly focused on finding novel approaches for ax vim expansion of progenitor cells including MPCs.
  • U.S. Pat. No. 6,087,113 issued Jul. 11, 2000 entitled “Monoclonal antibodies for human mesenchymal stem cells” describes a method for isolating a population of human cells that based on their expression of specific cell surface antigens detectable using monoclonal antibodies, are enriched in cells capable of giving rise to a variety of mesenchymally-derived tissues.
  • mesenchymal stem cells can be identified and isolated by positive binding to such monoclonal antibodies, what is not considered is that these cells have been cultured on an adherent surface and then positively selected, a process that has the potential to change both the phenotype and functional properties of the marrow-derived cell population.
  • non-hematopoietic stem and progenitor cells including the mesenchymal progenitor cells can be expanded more rapidly.
  • non-hematopoietic progenitor cells can usefully be expanded in non-static suspension culture. More particularly, it has been found that non-hematopoietic progenitor cells of the type recoverable from bone marrow and comparable sources can be expanded significantly in stirred suspension bioreactors. Moreover, the progenitor cells expanded in non-static suspension culture retain the potential to differentiate into a variety of cell types other than blood cells, including neuronal and endothelial vascular tissue as well as the mesenchymal tissues including bone, cartilage, connective tissue, muscle, tendon and adipose tissue.
  • the present method enables progenitor cell expansion at a rate and efficiency that far surpasses known expansion techniques that rely on and are limited by two dimensional surfaces such as tissue culture plates and tissue flask surfaces.
  • the present method of culturing the exacted progenitor cells in suspension avoids the known techniques in which progenitor cells destined for expansion are first selected and then maintained in an adherence-dependent environment that influences and limits the phenotype of cells that can be expanded by those known techniques.
  • an input cell population comprising non-hematopoietic progenitor cells is first obtained, and the input cell population is expanded by the step of maintaining that input population in non-static suspension culture for a period, sufficient to expand cells with properties of the input population.
  • the input cell population can comprise non-hematopoietic progenitor (NHP) cells of various classes or types, and particularly includes mesenchymal progenitor cells (MPCs).
  • NHP non-hematopoietic progenitor
  • MPCs mesenchymal progenitor cells
  • the input cell population is not subjected to prior selection for adherent or anchorage-dependent cells.
  • the present invention provides an input cell population which is amenable to expansion by non-static suspension culturing thereof, wherein the input cell population comprises a cell population that is enriched for non-hematopoietic progenitor cells.
  • the input population is enriched for marrow parenchymal cell progenitors, and especially for mesenchymal progenitor cells.
  • such enriched input populations are characterized by the substantial absence of at least one hematopoietic cell type, i.e., a cell type bearing a surface marker specific to hematopoietic cells.
  • the input population is prepared by enriching a progenitor cell population for mesenchymal progenitor cells.
  • the input population is prepared by enriching for non-hematopoietic marrow parenchymal progenitors other than mesenchymal progenitors.
  • the input cell population can be extracted from bone marrow, particularly human bone marrow that is either cadaveric or, more desirably from a fresh source, or from any comparable source such as embryos, umbilical cord blood and the like, in accordance with extraction techniques established for progenitor stem cells.
  • the extract is suitably subjected to fractionation using, for instance, commercially available density gradient media that such as Ficoll-PaqueTM (Sigma).
  • the extracted progenitor cells can be subjected to a selection procedure that allows for subtraction (or depletion), from the exact, of cells having a phenotype that is not desired, such as a hematopoietic progenitor cell phenotype.
  • a selection procedure that allows for subtraction (or depletion), from the exact, of cells having a phenotype that is not desired, such as a hematopoietic progenitor cell phenotype.
  • Such enrichment can be achieved, for instance, using the fluorescence-activated cell sorting device and technique, or by application of the established rosetting technique.
  • Both techniques utilize antibodies directed at a surface marker unique to a cell type of interest. In essence, both techniques utilize the binding interaction between the antibody/marker to allow cells of that phenotype to be extracted from the cell population undergoing expansion.
  • the input population comprises an enriched input population of progenitors that is substantially devoid of at least one hematopoietic cell type, which cell type is characterized by the presence of at least one surface maker selected from CD3, CD14, CD38, CD66 and CD119.
  • the input cell population is enriched for cells that lack one or more surface markers associated with the major histocompatibility complex type I (MHC-I) and/or lack one or more surface markers associated with the major histocompatibility complex type II (MHC-II).
  • the input cell population is desirably enriched for cells that lack one or more, and desirably all, of the following surface makers: HLA-A, HLA-B, HLA-C, HLA-DR, HLA-DP and HLA-DQ.
  • the present invention provides a novel input cell population for suspension culturing to provide an expanded cell population from which non-hematopoietic and particularly mesenchymal cell types and tissues can be grown, the input cell population comprising a progenitor cell population that has not been adherence-selected and which is enriched fir non-hematopoietic progenitor cells.
  • enriched input progenitor cell population is characterized by the substantial absence of cells expressing one or more hematopoietic progenitor cell surface markers.
  • the novel input cell population is characterized by the substantial absence of cells bearing at least one surface marker selected firm CD3, CD14, CD38, CD66, ad CD119.
  • the novel input cell population is characterized by the substantial absence of cells bearing at least one marker of MHC, selected from HLA-A, HLA-B, HLA-C, HLA-DR, HLA-DP and HLA-DQ.
  • the novel input cell population is characterized by the substantial absence of all MHC marker-bearing cells.
  • the input cell population is expanded by the technique of non-static suspension culturing.
  • the input cell population is considered “expanded” when the input population has proliferated, and the numbers of coils with one or more of the functional and phenotypic properties of the input population has increased, relative to the number of input cells seeded in suspension culture.
  • Suspension culturing wider non-static conditions is performed in a bioreactor or other suitable environment that allows for the 3-dimensional culturing of cells in liquid medium under conditions of temperature, pressure and nutrition amenable to their expansion.
  • Non-static suspension culturing further involves agitation of the culturing environment, as a means to maintain the cultured cells in suspension as individual cells during their expansion.
  • Such agitation typically entails mechanical agitation such as stirring, but may also utilize other fluidizing techniques such as percolation, aspiration, and the like.
  • the bioreactor suitably is treated, e.g., siliconized, to inhibit cell adherence to the internal surfaces of the bioreactor.
  • the suitable medium may be a hematopoietic culture environment and the suspension may be in a stirred bioreactor.
  • the medium may be adapted to exclude nutrients of a type typically required for the culturing of hematopoietic progenitor cells and which are necessary only for the maintenance of such hematopoietic cells. For instance, typically the nutrient liquid medium does not require hydrocortisone, and similar supplements that have been utilized predominantly for hematopoietic cell maintenance.
  • expansion of the functional capacity of the input cell population is performed in a medium supplemented with cytokines and/or growth factors, or cellular sources thereof, useful to maintain cell viability over the culturing period.
  • cytokines and/or growth factors include IL-3 and stem cell actor (SCF, known also as steel factor (SF)).
  • SCF stem cell actor
  • Also particularly useful are such grown factors as platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF), which are valuable in expanding input cell populations capable of giving rise to neuronal cell types.
  • PDGF platelet-derived growth factor
  • FGF fibroblast growth factor
  • Still other growth factors and cytokines and other supplements can be utilized in accordance with culturing practices established for cell types of particular interest.
  • Expansion of the functional capacity of the input cell population can be achieved after non-static suspension culture for a matter of days, and culturing periods in the 1-3 week range and longer are also suitable.
  • the progenitor cells maintained in suspension are multipotential and include mesenchymal and other progenitor cells. Accordingly, the expanded population may be used to form differentiated phenotypes including bone, chondrocytes, adipocytes, endothelial cells, neural cells, myoblast cells and cartilage.
  • some or all of the cells can be placed in the appropriate differentiation environment to grow the desired tissue.
  • the present invention further provides an isolated population of expanded human progenitor cells, characterized in that:
  • the population phenotype is SH4 negative
  • the population results from culturing in the absence of at least one hematopoietic stem cell-specific culturing supplement.
  • the present invention provides an expanded population of human mesenchymal progenitor cells which result from the non-static suspension culturing of an input population enriched for non-hematopoietic progenitor cells, as described above.
  • the present invention provides a therapeutic composition, useful for delivering human mesenchymal progenitor cells to an environment conducive to the formation of differentiated cells therefrom, the composition comprising a population or subpopulation of human progenitor cells as described above, and a physiologically acceptable vehicle for delivering said cells to said environment
  • the vehicle is one suitable for delivering the cells to a bone site at which repair or regeneration is desired.
  • the vehicle used to deliver the cells is a matrix material that is compatible with bone and may itself comprise osteoinductive material
  • the present invention provides an expanded progenitor cell population as described above in combination with factors suitable for inducing the differentiation thereof.
  • Such a combination is manifest as an assay environment as a tissue growth environment of the type used to prepare tissue for implantation or of the type established following treatment of a human or other patient to provide for tissue repair or regeneration.
  • the present invention provides a method for producing differentiated, non-hematopoietic cell types, comprising the steps of obtaining an expanded progenitor cell population of the type described above, or a subpopulation thereof, and then delivering that population of cells either per se or in a suitable delivery vehicle to an environment which supports differentiation thereof into a desired cell type.
  • the cells are delivered to an environment suitable for inducing their differentiation into a parenchymal cell or tissue selected from bone, cartilage, muscle, adipose, tendon, cartilage, and neurons.
  • the mesenchymal progenitor cells awe exploited for the purpose of repairing or regenerating bone, and are therefore delivered to a bone site at which such endpoints are desired.
  • FIG. 1A is a graphical representation illustrating the change in total cell number with time relative to input numbers from density gradient separated human bone marrow-derived cells maintained in a stirred suspension bioreactor (see also FIG. 8),
  • FIG. 1B shows the expansion profile of the colony forming units (CFU) generated from the cells maintained in each treatment group upon plating in a CFU-F assay;
  • FIG. 1C is a graphical representation of total cell (panels A, D), CFU-F (B, E) and CFU-O (C, F) expansion calculated from cells grown in stirred suspension bioreactors (A-C) and static control cultures (D-F). Bars represent the mean of three different experiments ⁇ standard deviation (see also FIG. 8).
  • FIG. 1D shows various types of differentiated cells induced from the parenchymal progenitor tell population, grown in the presence of defined cytokine cocktails.
  • the cytokine concentrations used were 10 ng/ml (stems cell factor) (SCF), 2 ng/ml interleukin-3 (IL3), 100 ng/mJ macrophage colony stimulating factor (MCSF), and 30 ng/ml platelet-derived growth factor (PDGF).
  • SCF stems cell factor
  • IL3 interleukin-3
  • MCSF macrophage colony stimulating factor
  • PDGF platelet-derived growth factor
  • FIGS. 2 A- 2 D are optical micrographs taken of CPU-F cultures of suspension cells isolated from bioreactors at Day 5 with differeing combinations of cytokines;
  • FIGS. 2 E- 2 H are CFU-F assay results of cells removed from suspension bioreactors at Day 20 with differing combinations of cytokines;
  • FIGS. 3A, 3B, 3 C and 3 D depict tetracycline labeled osteogenic cultures of Day 5 and Day 10 suspension cells maintained in the presence of various cytokines. Mineralized nodular areas first appeared after 3 weeks and cultures were terminated at 5 weeks. Fluorescence intensity corresponded to tetracycline that had chelated to the mineralized areas.
  • the images in (A-B) reveal tetracycline signals that corresponded to nodule-like structures that formed from cells that were removed, at 5 and 10 days from suspension bioreactors supplemented with various cytokine cocktails. All bone nodule assays were initiated with 1 ⁇ 10 4 cells/cm 2 . The tetracycline signal from each bone nodule culture initiated with cells derived from bioreactors experiments were quantified using the Chemilmager 5500 software (Alpha Innotech Corporation, San Leandro, Calif.);
  • FIG. 3F illustrates the expansion of CFU-O progenitors detected as bone nodule colonies over the time period in those conditions presented in Figures A-D;
  • FIGS. 4A and 4B show dark field micrographs of developing nodular areas (3 weeks) generated from cells derived from the no cytokine and SCF+IL3 bioreactor treatment groups after 7 days of suspension culturing;
  • FIGS. 4C and 4D show no nodular areas associated with the cells retrieved from the SCF+PDGF treatment
  • FIG. 4E is a graphical representation showing the tetracycline signal associated with the mineralized areas of FIGS. 3A, 3B, 3 C and 3 D quantified and normalized to the input signal of bone nodule cultures initiated with calls removed from the culture suspension on day 5 and day 10.
  • FIGS. 4F and 4G are also graphical representation showing the tetracycline signal from each bone nodule culture initiated with cells derived from both bioreactors and static control (type 1) experiments (see FIG. 8 description for static control type I description). Panels F and G reveal the quantitative results, including results with a combination of SCF+IL-3+PDGF, where the tetracycline positive signal is expressed as an integrated density value (IDV). All bone nodule assays were initiated with 1 ⁇ 10 4 cells/cm 2 . Each bar represents the mean tetracycline positive signal front 3 bone nodule cultures ⁇ SD for each cytokine conditions;
  • FIGS. 5 A- 5 D are scanning electron microscope (SEM) images taken of bone nodule cultures of Day 5 suspension cells that were grown in the various treatment groups studied; and FIG. 5F is a transmission electron microscope (TEM) image showing that the matrix lining the culture dish comprises globular accretions that form the cement line matrix.
  • FIG. 5E shows a differentiating osteogenic cell secreting globular accretions forming the cement line matrix
  • FIGS. 5G and 5H are images taken from cultures initiated with cells removed from bioreactors after 3 weeks. Field widths (F. W: 17 ⁇ m for G and H)
  • FIG. 6 provides flow cytometric dot plots of various cell populations at different time points throughout suspension culturing.
  • FIG. 6A reveals CD34 and CD45 phenotype and progression over culturing time in the presence and absence of cytokines.
  • the input population contained a 65% CD45 and 0.5% CD34 population.
  • FIG. 6B particularly shows dot plots generated from Ficoll-PaqueTM bone marrow incubated with various antibodies against cell surface markers.
  • the dot plots reveal that Ficoll-PaqueTM processed human bone marrow contains populations of cells that are CD45 + , CD34 low , CD50 + , SH2 low , SH4 ⁇ and STRO-1.
  • the cell population also includes a double negative population ( ⁇ 17%) when incubated with antibodies against HLA-A, B, C and -DP, DQ, DR.
  • FIG. 6C shows flow cytometric analysis-plots for expression of HLA-A, 8, C and HLA-DP, DQ, DR markers, which correspond to MHC class I and MHC class II, respectively.
  • Plotted is the expansion of the above discussed mark as on Ficolled Bone marrow cells (A, B and C), the population of cells grown in the presence of SCF+IL3 (D, E and F) and in the absence of cytokines (G) after 21 days of culture.
  • Plot H is the negative control for the input population while plot I is the negative control for the SCF+IL3 and No Cytokines experiments, and
  • FIG. 7 reveals the calculated CD45 ⁇ and CD45 + cell expansion as determined by flow cytometric analysis of cells present in bioreactors at 1, 2 and 3 weeks. Greater CD45 ⁇ and CD45 low cell expansion is achieved in the presence of SCF+IL3. Each bar represents the meal CD45 ⁇ and CD45 + cell number calculated from three independent flow cytometric analysis of bioreactor-derived cells at 1, 2 and 3 weeks.
  • FIG. 8 reveals total cell expansion in stirred suspension bioreactors as a function of cytokine supplementation.
  • B demonstrates the extent of cell growth attained in 3 weeks through suspension culturing as of function of cytokine addition. Two types of static control experiments were performed: Static Control I (C) and II (D).
  • Static Control 1 was designed to function as a control for bioreactor cultures and therefore, was initiated with the same number of cells, suspended in the same growth medium and a one-third medium exchange (including both adherent and suspension cells) was performed weekly.
  • Static Control II involved initiating cultures also with the same number of cells as the bioreactor cultures; however, the cultures were passaged at ⁇ 80-90% confluency ad then split 1:2.
  • C and D represent the extent of cell growth achieved through the two types of static cultures. Note the scale difference on the y-axis in C and D compared to B. Each point represents the mean of 3 independent experiments were the error bars represent S.D. Significant differences (p ⁇ 0.05) in total cells (for both B and C) were observed between the groups at week 3 (denoted with *);
  • FIGS. 9A and 9B are graphic representations of the fold increase in CFU-F numbers relative to input from cells derived from suspension bioreactor treatment groups and adherent cells removed by 0.1% typsin treatment from static controls (type 1), respectively.
  • a significant difference (p ⁇ 0.05) was observed in CFU-F expansion between the No Cytokine and the other treatment groups at wee 3 (denoted with *) in 9 A.
  • Nodular areas were enumerated and used to calculate the fold increase in CFU-O numbers relative to input from cells derived from bioreactor cultures FIG. 9C) and cells removed from the adherent fraction of static control (type 1) experiments ( 9 D).
  • FIG. 9E shows a graphical representation of the positive expression associated with NeuN expressing cells of 2-week old CFU-F cultures that were initiated with cells removed from a SCF+IL3+PDGF supplemented bioreactor at 1, 2 and 3 weeks. The cultures were incubated with mouse anti-NeuN monoclonal antibody (MoAb) and positive expression was detected as a fluorescence signal above background using an anti-mouse MoAb conjugated to Alexa Fluor 594.
  • MoAb mouse anti-NeuN monoclonal antibody
  • FIG. 10A shows a graphical representation of the growth of RosetteSepTM-derived human bone marrow-derived cells cultured in stirred suspension bioreactors in the absence and presence of SCF+IL3.
  • FIG. 10B shows a tetracycline incubated bone nodule assays (5 week) initiated with cells derived from the SCF+IL3 bioreactor treatment group at Day 7, while
  • FIG. 10C is a dark field micrograph (50 ⁇ ) showing a mineralized nodular area.
  • the invention relates to human progenitor cells from which a variety of non-hematopoietic cell types can differentiate. Because of the variety of non-hematopoietic cells that can differentiate from the present progenitor cell population, it is referred to herein as a population of non-hematopoietic progenitor a cells. Such a population comprises mesenchymal progenitor cells (MPCs), as well as other non-hematopoietic progenitor cells. Such progenitor cells may also be referred to as “precursor” cells, and these terms are considered equivalent herein for the reason that both progenitors and precursors are able to give rise to differentiated cell type.
  • MPCs mesenchymal progenitor cells
  • precursor cells are considered equivalent herein for the reason that both progenitors and precursors are able to give rise to differentiated cell type.
  • the present progenitor cell population results from the non-static suspension culturing of cells of the type obtained from bone marrow, using techniques that are described in greater detail in the examples herein. It is to be appreciated that her comparable and known sources of such progenitor cells can also be used, as noted hereinabove, including particularly umbilical cord and placental blood, peripheral blood, skin, adipose, and muscle.
  • the present invention applies a non-static approach to culturing. This is distinct from established methods that are static, and which are designed not to disturb cells undergoing expansion. Rather, in the present method, the cells are cultured under non-static conditions that introduce agitation and thereby produce cultures in which the cells a present as non-adherent individual cells, and not as either large clumps or as confluent layers.
  • the agitation source is typically a mechanical agitation means that either introduces movement either directly within the culturing medium or externally through the surface on which the culturing vessel is placed.
  • the non-static conditions are introduced by stirring means, which can include a magnetized stirring paddle that is placed the bioreactor and is induced by external magnetizing means.
  • stirring means can include a magnetized stirring paddle that is placed the bioreactor and is induced by external magnetizing means.
  • the agitation can be introduced by other means that include fluidized culturing beds, which introduce agitation through the movement of appropriate culturing atmosphere through the cell population.
  • the magnitude of the agitation will be that which is sufficient to maintain the cells in suspension substantially as individual) expandable cells with desired circulation of culturing nutrients, but insufficient to introduce shear forces that will result in cell lysis.
  • the optimum stirring speed is about 40 rpm under the experimental conditions noted herein.
  • the culture system was configured such that i) the impeller was placed in the middle of the bioreactor at a 90°angle to the solution surface to maintain axial flow, ii) a constant mixing speed of 40 rpm was used to maintain the cells in suspension and iii) the agitator was positioned three quarters of the way down the vessel to ensure uniform mixing.
  • the present method introduces a three-dimensional environment for cell expansion, and raises exponentially the volume of the environment with in which progenitor cells can be expanded, thereby offering increases not only in the number of cells that can be expanded at any given time but also accelerating the rate at which the cells can be expanded over time.
  • suitable culturing volumes can range from the milliliters to many litres, for instance from about 0.1 L to more than 200 L, e.g., 0.5 L to 100 L, such as 1 L to 10 L.
  • Suitable culturing media for use in the present method, as well as culturing temperature and atmosphere are those well established in the art for the culturing of progenitor cells. Temperatures suitably lie within the physiological range.
  • the culturing atmosphere is desirably the appropriate blend of O 2 and CO 2 , e.g., humidified 5% CO 2 in air.
  • Culturing medium is any appropriate liquid medium particularly including the commercially available long term culturing (LTC) media, which desirably is supplemented with agents useful in the expansion of non-hematopoietic progenitor bells, such as various growth factors and nutrient supplements.
  • LTC long term culturing
  • each culture received a different cytokine treatment that included the following combinations: no cytokines, SCF, IL3, MCSF, SCF+IL3, SCF+PDGF and SCF+IL3+PDGF.
  • no cytokines SCF, IL3, MCSF, SCF+IL3, SCF+PDGF and SCF+IL3+PDGF.
  • the selection of these growth factors was based on previous report of their effects on the growth of fibroblasts and other associated mesenchymally-derived cells (see for instance Wang, et al, Exp. Hematol. 18, 341-347 (1990); Hirata et al, Acta Haematol. 74, 189-194 (1985), and Yamada et al, J Cell Physiol 184, 351-355 (2000).
  • Suitable culturing periods will be determined largely based on the number of progenitor cells demanded from the expansion process. Under conditions noted in the examples, the numbers of progenitor cells can readily be expanded 5-10 fold over a one week period from initial seeding, with longer term culturing providing further increases in progenitor cell numbers.
  • the present progenitor cell population results from non-static suspension culture of the bone marrow derived input cell population, or an enriched subpopulation thereof, and is produced without an intervening step of selection based on cell adherence or anchorage dependence, which is more common in the art and which in itself is believe to impart certain phenotypic traits to the prior art cell populations that are not reflected in the present cell population.
  • the present population is also distinct from a homogeneous mesenchymal stem or progenitor cell population in its ability to give rise to differentiated cell progeny including endothelial cells, and cells forming neuronal tissue, including those displaying neurite outgrowth.
  • the present progenitor cell population can be sustained in culture media that lacks one or more artificial media supplements used commonly in the culturing of hematopoietic stein cells, such as hydrocortisone.
  • the present invention also provides a progenitor cell population that is enriched for non-hematopoietic cells, relative to the extracted population.
  • enriched input cell populations include cell populations from which one or more hematopoietic cell types have been subtracted, and those input cell populations to which one or more non-hematopoietic progenitor cells have been added.
  • the enriched human mesenchymal progenitor cells are represented by cell populations from which one or more cells of a hematopoietic phenotype are subtracted.
  • cell phenotypes that can be subtracted to arrive at the present enriched populations include cells types bearing one or mote of the following markers: CD3, CD14, CD38, CD19, CD66, CD119, and cells bearing markers of the major histocompatibility complex types I and II which include HLA-A, HLA-B and HLA-C (for MHC-I) and HLA-D, HLA-E and HLA-F (for MHC-II).
  • the generation of such enriched progenitor cell populations can be achieved using the FACS procedure in combination with antibodies to markers on the cell types to be subtracted.
  • Such antibodies, and procedures and devices for performing such subtractions are commercially available.
  • the procedure can be applied either to the cells following their extraction and prior to suspension culturing, or the procedure can also be applied to the expanded population before being subjected to a differentiation environment e.g. prior to its formulation or subsequent use for tissue production.
  • the cell populations generated from the input progenitor cell populations are useful medically in various therapies designed to repair or regenerate tissue of any type into which these populations can differentiate.
  • the expanded mesenchymal progenitor cell population can be formulated for delivery to the site at which differentiation is desired, using any delivery vehicle that is physiologically tolerable to both the recipient and to the viability of the cells formulated therein.
  • Particular formulations and suitable delivery vehicles are known in the art, and will be apparent from the nature of the intended therapy.
  • the formulation further contains agents, such as growth factors and cytokines, tit enhance viability and/or the differentiation of the administered cells.
  • the present invention also provides a method of using specifically differentiated cells for therapy comprising administering the specifically differentiated cells to a patient in need thereof. It further provides for the use of genetically engineered multipotent stem cells to selectively express an endogenous gene or a transgene, and or the use of the progenitor cells either per se or in expanded from in vivo for transplantation/administration into an animal to treat a disease.
  • the cells can be used to engraft a cell into a mammal comprising administering autologous, allogenic or xenogenic cells, to restore or correct tissue specific structural or other function to the mammal.
  • the cells can be used to engraft a cell into a mammal causing the differentiation in vivo of cell types, and for administering the progenitor or differentiated cells into the mammal.
  • the cells, or their in vitro or in vivo differentiated progeny can be used to correct a genetic disease, degenerative disease, neural, or cancer disease process. They can be used to produce gingiva-like material for treatment of periodontal disease. They could be used to enhance muscle such as in the heart.
  • a genetically engineered progenitor cell, or its differentiated progeny can be used to treat a disease with CNS deficits or damage.
  • progenitor cells can be used to treat disease with neural deficits or degeneration including among but not limited to stroke, Alzhemier's, Parkinson's disease, Huntington's disease, AIDS associated dementia, spinal cord injury, metabolic diseases effecting the brain or other nerves.
  • neural deficits or degeneration including among but not limited to stroke, Alzhemier's, Parkinson's disease, Huntington's disease, AIDS associated dementia, spinal cord injury, metabolic diseases effecting the brain or other nerves.
  • the progenitor cells, or cartilage differentiated progeny can be used to treat a disease of the joints or cartilage such as cartilage tears, cartilage thinning, and osteoarthritis.
  • the cells or their osteoblast differentiated progeny can be used to treat bone disorders and conditions, such as bone fractures, osteoarthritis, bone voids caused by surgery or tumors for tissue regeneration in osteoporosis, Paget's disease, and osteomyelitis.
  • progenitor cells and their expanded equivalents, to differentiate is achieved using techniques established in the art, which vary according to the differentiated cell type desired.
  • progenitors can be cultured for about 14-21 days in culturing medium comprising supplements such as dexamethasone, ⁇ -glycerophosphate and ascorbic acid, and optionally including various bone growth factors.
  • the presence of osteoblasts can be confirmed by Von Kossa staining, or antibodies against a bone cell marker such as bone sialoprotein, osteonectin, osteopontin and osteocalcin.
  • the progenitors can be grown in serum-free DMEM supplemented with TGF- ⁇ in suspension culture, for about 14 days or more.
  • Adipocyte differentiation can be detected by examination with light microscopy, staining with oil-red, or detection of lipoprotein ( ⁇ I'm not sure of the spelling of this) lipase (LPL), adipocyte lipid-binding protein ( ⁇ P2), or peroxisome proliferator-activated receptor gamma (PPAR).
  • LPL lipoprotein
  • ⁇ P2 adipocyte lipid-binding protein
  • PPAR peroxisome proliferator-activated receptor gamma
  • Adipocytes can be used for the treatment of Type II diabetes, and in reconstructive or cosmetic surgery, e.g., for breast reconstruction after mastectomy, or for reshaping tissue lost as a result of other surgery.
  • progenitor cells can be treated with 5-azacytidine in expansion medium for a period, and then transferred to LTC medium. Differentiation can be confirmed by detecting sequential activation of Myf-5, Myo-D, Myf-6, myogenin, desmin, skeletal actin and skeletal myosin, either by immunohistochemistry or Western blot analysis. Smooth muscle cells can also be induced by culturing progenitors in serum-free medium, without, growth actors supplemented with high concentrations of platelet-derived growth factor (PDGF). Terminally differentiated smooth muscle cells can be identified by detecting expression of desmin, smooth muscle specific actin, and smooth muscle specific myosin by standard methods. Cardiac muscle differentiation can be accomplished by adding basic fibroblast growth factor (bFGF) to the standard serum-free culture media without growth factors.
  • bFGF basic fibroblast growth factor
  • the progenitor cells of the present invention can be used in cell replacement therapy and/or gene therapy to treat a variety of conditions.
  • the cells can be manipulated to serve as universal donor cells for cell and gene therapy to remedy genetic or other diseases.
  • Particularly useful as donor cells are those progenitors that express neither class of HLA antigen, and which therefore avoid NK-mediated killing when transplanted.
  • tissue layers from cultured cells are known to those of skill in the art (see, for example, Vacanti, et al., U.S. Pat. No. 5,855,610). These methods can be especially effective when used in combination with cells of the present invention, which have a broad range of differentiation.
  • either autologous or allogeneic progenitors of the present invention can be administered to a patient either in differentiated or undifferentiated state, genetically altered or unaltered state, by direct injection to a tissue site, systemically, with an acceptable matrix, or in combination with a pharmaceutically acceptable carrier.
  • the cells can be provided as frozen stocks, alone or in combination with prepackaged medium and supplements for their culture, and can be additionally provided in combination with separately packaged effective concentrations of appropriate factors to induce differentiation to specific cell types.
  • the cells can be provided as frozen stocks containing cells induced to differentiate by the methods described herein above.
  • the expanded progenitor cells and particularly the expanded mesenchymal progenitors are utilized in bone therapy.
  • the cells can be delivered as such or together with a suitable matrix, such as a liquid or gelatinous material, by injection or applied as a paste to a site at which bone formation is desired.
  • the cells can be placed ex vivo in a differentiation environment, exemplified by the CFU-O conditions described herein, and then transplanted to the intended site when their differentiation to bone tissue has reached an appropriately mature stage.
  • hBMDC Human bone marrow-derived cells
  • This medium contains a-medium supplemented with 12.5% (FCS), 12.5% horse serum, 0.1 mM B-2-mercaptoethanol, 2 mM 1-glutamine, 0.16 mM 1-inositol and 0.016 mM folic acid (medium referred to as MyeloCultTM and is available from StemCell Technologies (SCT), Inc, Va, BC, Canada).
  • FCS 12.5%
  • B-2-mercaptoethanol 2 mM 1-glutamine
  • 0.16 mM 1-inositol 0.16 mM 1-inositol
  • 0.016 mM folic acid medium referred to as MyeloCultTM and is available from StemCell Technologies (SCT), Inc, Va, BC, Canada.
  • non-hematopoietic progenitor cells may also be derived from human umbilical cord or other tissues that have been shown to contain cells capable of exhibiting a similar developmental capacity to the bone marrow derived cells, e.g., stromal and mesenchymal, potential that can be expanded in suspension and under the appropriate culture conditions.
  • stromal and mesenchymal e.g., stromal and mesenchymal
  • Suitable enrichment techniques an those in which the extracted cells are depleted of at least one hematopoietic cell type, such as by processes that utilize antibodies to subtract cells bearing surface markers characteristic of hematopoietic progenitors.
  • One particularly useful technique is the commercially available RosetteSepTM procedure, which is designed to enrich for mesenchymal progenitors.
  • RosetteSepTM contains a combination of mouse and rat monoclonal antibodies purified from mouse ascites fluid or hybridoma culture supernatant. These antibodies are bound to specific bispecific antibody complexes, which are directed against cell surface antigens on human hematopoietic cells (CD3, CD14, CD19, CD38 and CD66b—see below for details in Table 1) and glycophorin A on red blood cells.
  • the mouse monoclonal antibody subclass is IgG1.
  • TABLE 1 Surface Marker Description CD3 Reacts with the ⁇ chain of the CD3/T-cell antigen receptor (TCR) complex found on 70-80% of normal human peripheral blood lymphocytes and 10-20% of thymocytes.
  • CD3 plays a role in signal transduction during antigen recognition.
  • CD14 Reacts with a 53-55 kDa glycosylphosphatidylinisitol (GIP)-anchored signal chain glycoprotein expressed at high levels on monocytes. Additionally, CD14 antibody reacts with interfollicular macrophage, reticular dendritic cells and some Langerhans cells.
  • CD19 Reacts with the 95 kDa type I transmembrane glycoprotein expressed during all stages of B-cell differentiation and maturation, except on plasma cells. CD19 is also present on follicular dendritic cells. It is not found on T cells or on normal granulocytes. CD19 plays a role in regulation of B-cell proliferation.
  • CD38 Reacts with the 45 kDa type II single-chain transmembrane glycoprotein present on thymocytes, activated T-cells and terminally differentiation B-cells (plasma cells). Other reactive cells include monocytes, macrophage, dendritic cells and some epithelial cells CD66b Reacts with CD66b, a 100 kDa GPI- linked protein expressed on granulocytes. This molecule was previously clustered as CD67 in the Fourth Human Leukocyte Differentiation Antigen (HLDA) Workshop and renamed CD66b in the Fifth HLDA workshop
  • the RosetteSepTM cocktail enriches for mesenchymal progenitor cells by removing the following cells: peripheral blood lymphocytes, peripheral blood thymocytes, monocytes, macrophage, differentiating B-cells, activated T-cells, plasma cells, dendritic cells, some epithelial cells and granulocytes.
  • RosetteSepTM enrichment for mesenchymal progenitor cells involves adding 50 ⁇ l of RosetteSepTM cocktail (available through StemCell Technologies, Canada) per ml of whole bone marrow cells and mixing well, followed by a 20 minute incubation period at room temperature. The sample is then diluted with an equal volume of Phosphate Buffered Saline (PBS)+2% FBS and then mined gently. The diluted sample is layered on top of density medium (Ficoll-PaqueTM) while minimizing mixing of the density medium and sample. The tube is centrifuged for 20 minutes at 1750 rpm at room temperature, with the brake off. The enriched cells are located at the plasma interface and are removed.
  • RosetteSepTM cocktail available through StemCell Technologies, Canada
  • a 100-ml stirred suspension spinner flask (catalogue A: 1965-0010, Bellco, Vireland, N.J.) was used. The flasks were siliconized (Sigma) prior to use to prevent the attachment of adherent calls.
  • Suspension cultures were initiated with approximately 5 ⁇ 10 3 to 1 ⁇ 10 6 cells/ml in LTC medium, either with or without the addition of various soluble factors such as 2 ng/ml of highly purified recombinant human interleukin 3 (rhIL-3) (Stem Cell Technologies, VA, Canada), 10 ng/ml of highly purified recombinant human Steel Factor (rhSF) (also referred to interchangeably as stem cell factor, or SCF) (Stem Cell Technologies) or 10, 20 or 30 ng/ml of human recombinant platelet derived growth factor (rhPDGF) (Sigma). Cultures were maintained at 37° C. in humidified atmosphere of 5% CO 2 z in air with constant stirring at 40 rpm.
  • rhIL-3 highly purified recombinant human interleukin 3
  • rhSF highly purified recombinant human Steel Factor
  • SCF stem cell factor
  • rhPDGF platelet derived growth factor
  • Static cell cultures (Static Control I) are initiated in T-75 cm 2 flask to function as controls for stirred suspension bioreactor experiments. Each flask is initiated with the same concentration of cells (i.e. 1 ⁇ 10 5 cells/ml) as its parallel stirred suspension bioreactor culture At 7, 14, and 21 days, one third of the suspension cells, including the media and one third of the adherent cells, detached by 0.01% trypsin treatment, are removed and fresh, LTC media with or without the addition of soluble factors is replaced as at the start. Both the suspension and adherent cells recovered from the medium change are counted and analyzed by flow cytometry while only the adherent cells are used in initiating progenitor assays. The remaining two-thirds of the cell culture (with fresh medium) is inoculated into a new T-75 cm 2 flask.
  • CFU-F assay was-used to evaluate the number of marrow mesenchymal cell precursors in cultures maintained in suspension by continuous stirring and the possible effect of the added cytokines on effecting output CFU-P colony numbers and cell composition.
  • CFU-F numbers were determined using a modified version of the method described by Castro-Malaspina et al., 1980. Although having the same essential minerals/vitamins and serum as previously described, the present protocol utilized the medium described below, supplemented with 10% antibiotic solution. The cell suspension obtained from weekly medium changes was washed in Iscove's medium containing 2% FCS (Stem Cell Technologies, VA, Canada).
  • the appropriate cell number was resuspended in MesenCultTM medium (Available through SCT and contains fetal bovine serum (10%) in McCoy's 5A medium (modified) supplemented with L-glutamine (2 mM)).
  • the cells were plated at 1 ⁇ 10 4 cells/cm 2 suspended in MesenCultTM medium and then incubated without being perturbed for 14 days. After 2 weeks, the media was removed and the dishes were fixed and stained with ⁇ -naphthyl acetate esterase followed by a counterstain with Hematoxylin Solution (Sigma). Adherent colonies containing greater than 50 cells with fibroblastic-like morphology were counted at ⁇ 10 magnification.
  • the cell suspension obtained at weekly medium change was placed in a bone nodule assay determine the capacity of the bioreactor-derived cells to form mineralize nodular areas and to calculated the number of colony forming unit-osteogenic (CFU-O) progenitors. Following the wash and centrifugation steps, the pellet was resuspended in bone forming medium containing ⁇ -minimal essential medium, 15% fetal bovine serum by volume, 10% antibiotic solution by volume, and 1% of dexamethasone (10 ⁇ 8 M) and L-ascorbic acid (50 ⁇ g/ml) by volume and plated on tissue culture polystyrene dishes at a cell seeding density of 1 ⁇ 10 4 cells/cm 2 .
  • CFU-O colony forming unit-osteogenic
  • ⁇ -glyercophosphate at a concentration of 3.5 mM was added to the culture. Each week until termination the flasks were re-fed with fresh medium. The cultures were maintained until such time as mineralized nodular areas were observed after which the cultures were fixed and prepared for analysis.
  • Tetracycline (9 ⁇ g/ml) was added to the cultures prior to termination. At termination, the cells were fixed in Karnovosky's fixative overnight and then viewed by UV-excited fluorescence imaging for tetracycline labeling of the mineral component of the nodular areas.
  • Representative samples of bone nodule cultures were prepared for SEM by first placing them in 70%, 80%, 90% and 95% ethanol for 1 hour followed by immersion in 100% ethanol for 3 hours. They were then critical point dried. A layer of gold approximately 3 nm layer was sputter coated with a Polaron SC515 SEM Coating System onto the specimens, which were then examined at various magnifications in a Hitachi S-2000 scanning electron microscope at an accelerating voltage of 15 kV. The images generated were used to demonstrate the presence of morphologically identifiable bone matrix.
  • the procedure for the STRO-1 antibody stain involved resuspending the cell suspension (1 ⁇ 10 5 cells) in 200 ⁇ l of saturating concentrations of the mouse IgM monoclonal antibody STRO-1 for 30 minutes at 4° C. or on ice. This step was preceded with blocking the cell suspension with 1% human goat and mouse sera for 10 minutes. The cells were then washed twice with PBS containing 2% PBS before the addition of 3 ⁇ l/105 or 100 ⁇ l of PE-conjugated rat anti-mouse IgM monoclonal antibody (BD Biosciences, CA). Cell suspensions were washed twice with PBS+2% PBS and then 7 AAD was added to the final suspension.
  • CFU Total Cell and Colony Forming Unit
  • FIG. 1A is a graphic representation illustrating the change in total cell number with time, relative to input cell numbers, firm human bone marrow-derived cells maintained in a stirred suspension bioreactor.
  • FIG. 1B shows the expansion profile of the colony forming units (CFU) generated from the cells maintained in each treatment group.
  • CFU colony forming units
  • FIG. 1C shows the advantage of stirred suspension bioreactors over static control cultures, in expanding the progenitor cell population, and importantly, in producing mesenchymal progenitor cells that differentiate into fibroblast and bone cell colonies.
  • FIG. 1D shows the morphology of cells differentiated from the expanded progenitors.
  • FIG. 8 shows the effect on expansion of various cytokine combinations, measured at weekly intervals. It is apparent that, by 3 weeks of culture, certain treatment groups resulted in greater total cell expansion, while others failed to even maintain input cell numbers (FIG. 8A).
  • the groups with cell expansions of greater than 5-fold after 3 weeks of culture were namely the SCF+IL3 and SCF+IL3+PDGF.
  • Static Control I FIG. 8C
  • Static Control II FIG. 8D
  • FIG. 9 shows the results of testing the functional capacity of our bioreactor grown cells to form bone matrix and colonies of fibroblast-like cells. Colonies of fibroblast-like cells were enumerated from bioreactor-derived cells inoculated in CFU-F assays and the fold increase in CFU-F numbers were calculated relative to input (FIG. 9A). Tetracycline labeling of bone nodule cultures was used to label the newly formed biological mineral phase of bone and as a method of enumerating CFU-O progenitors to determine the extent CFU-O expansion over the time period studied. FIG.
  • FIGS. 9C and 9D are static control I CPU-F and CPU-O expansion results. It is apparent that there is a greater extent of these progenitors expansion achieved through suspension culturing. Light micrographs of cells generated in the CPU-F assays of the PDGF-treated cultures suggested, based on morphology, that the cells maybe of neural origin.
  • FIGS. 2 A-D represents light micrographs taken of CFU-F cultures of suspension cells isolated from bioreactors at Day 5. At 14 days, cultures were with stained with ⁇ -naphthyl acetate esterase followed by a counterstain with Hematoxylin Solution.
  • FIGS. 2 B- 2 H are CFU-F assay results of cells removed from suspension bioreactors at Day 20. Note the similarity in the cells comprising the colonies from Day 5 and Day 20 suspension cells, with the SCF+PDGF treatment having a strikingly different cell composition and morphology from the rest of the treatment groups.
  • Tetracycline labeling of cultures was used for labeling of the newly formed calcium phosphate associated with the biological mineral phase of bone.
  • the tetracycline labeling of the cultures coincide with the mineralized nodular areas, which is visualized by exposing the cultures to UV light
  • FIGS. 3 A- 3 D depict tetracycline labeled bone nodule cultures of Day 5 and Day 10 suspension cells maintained in the various cytokine conditions. These images were generated by UV-excited fluorescence imaging. The CFU-O colonies were enumerated by this method and
  • FIG. 4F illustrates the expansion of CFU-O colonies over the time period from the treatment conditions described in FIGS. 3 A-D.
  • both the no cytokine and SCF+IL3 treatment groups showed quantifiable levels of expansion where as the SCF+PDGF and PDGF treatment groups did not yield any bone colonies.
  • FIGS. 4A and 4B show dark field micrographs of developing nodular areas associated with the no cytokine and SCF+IL3 groups.
  • FIGS. 4C and 4D show no nodular areas associated with the cells retrieved from the SCF+PDGF treatment (such results were also observed for the PDGF treatment, data no shown).
  • the tetracycline signals associated with the mineralized areas were quantified and normalized to the input CFU-O signal, as shown in FIG. 4E.
  • the SCF+PDGF and PDGF groups show a tetracycline signal, however, this signal is associated with the random deposition of mineral throughout the culture dish, which can be described as dystrophic mineralization.
  • Similar experiments were performed on No Cytokine, SCF+IL3 and SCF+IL3+PDGF suspension bioreactors whereby cells were removed instead at 1, 2 and 3 weeks (FIGS. 4F and 4G). It is apparent that the SCF-IL3 and SCF+IL3+PDGF conditions ( 4 F) resulted in a greater tetracycline signal at 3 weeks when compared to controls ( 4 G).
  • FIGS. 5 A-D are SEM images taken of bone nodule cultures of Day 5 suspension cells that were grown in the various treatment groups studied.
  • FIGS. 5A and 5B show cement line matrix covering the underlying substratum. This matrix is the initial matrix laid down by differentiating osteogenic cells (FIG. 5B).
  • FIGS. 5C and 5D shows mineralized collagen, comprising the nodular area.
  • Transmission electron microscopy (FIG. 5F) confirmed that the matrix lining the culture dish comprises globular accretions that form the cement line matrix.
  • bioreactor-derived cells maintained their capacity to form cement line matrix and mineralized collagen as seen in FIG. 5G (No Cytokine condition) and 5 H (SCF+IL3 condition).
  • the antibody was detected with a goat anti-mouse IgG MoAb conjugated with Alexa Fluor 594 (1:200 dilution, Molecular Probes, Eugene, Oreg.).
  • a goat anti-mouse IgG MoAb conjugated with Alexa Fluor 594 (1:200 dilution, Molecular Probes, Eugene, Oreg.).
  • Alexa Fluor 594 1:200 dilution, Molecular Probes, Eugene, Oreg.
  • FIGS. 6 and 7 illustrate the type and distribution, over the culture duration, of specific populations of cells generated from the input cell population.
  • a shift in the distribution of the hematopoietic CD45 cells occurs over time, as a gradual reduction in cells of this type (FIG. 7).
  • the progenitor population has a SH4 negative phenotype.
  • the SH4 marker is alleged to be definitive of mesenchymal stem cells, suggesting that the present progenitor population is not homogeneous for this cell type.
  • FIG. 6C dot plots are provided for the HLA markers. Cells bearing these surface markers are involved in graft tissue rejection. The impact of suspension culturing on cells expressing these markers is elaborated in FIG. 6C. As is evident the input population of FIG. 6B reveals a heterogeneous HLA cell population, which includes a “double negative” phenotype representing 17% of the population. Upon expansion culturing in the presence and absence of SCF+IL3 for 21 days, it is evident that there is an increase in the fraction of double negative cells (FIG. 6C, panels D and G).
  • cells expanded by suspension culturing include those expressing CD50, a marker associated with endothelial cells.
  • non-static suspension culturing is a viable method for the expansion of progenitor cells that are of the HLA double negative phenotype.
  • progenitor cells that are of the HLA double negative phenotype.
  • Such cells will be very useful in the engineering either ex vivo of in vivo of allogeneic tissues given that they have yet to commit to a lineage recognized as “self” by the recipient.
  • non-hematopoietic progenitor cells of the marrow parenchyma including mesenchymal progenitor cells and particularly including osteoblast and adipocyte progenitor cells exist and remain viable in a long-term culture environment that is maintained in a stirred suspension bioreactor, as determined by they ability to form CFU-Fs and CFU-Os.
  • Preliminary factorial design experiments involving the addition of cytokines and combination of cytokines that are potent mitogens for proliferation have demonstrated that cytokine addition can influence the output colony numbers as well the cells comprising the colony forming units.
  • the cytokine supplementation appears to influence potential of the cells maintained in suspension to form bone matrix in vitro when placed in an appropriate bone forming culture environment.
  • the progenitor cells maintained in suspension are multipotential, that is, they can form other differentiated phenotypes including bone, chondrocytes, adipocytes, endothelial cells, fibroblastic cells, neuron cells, bone marrow, myoblast cells and human cartilage.
  • a suitable vehicle for delivery to a site at which differentiation into the desired cell pr tissue type is desired.
  • the cells can be introduced into a tissue-forming environment scaled to be suitable either for assay or for growth of human tissues or cells on a small scale for transplantation.
  • the input population to be cultured in the expansion bioprocess can be the input population of progenitor cells extracted from marrow parenchyma or a comparable source, or the input population can be an enriched, subpopulation thereof from which one or more hematopoietic cell phenotypes have been subtracted. It will be understood that the present invention is not restricted per se to humans and may be used for producing mesenchymal and other marrow parenchymal progenitor cells for non-human applications.
  • Naughton, G K and Naughton B A Three-dimensional cell and tissue culture system.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Neurosurgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Materials For Medical Uses (AREA)
US10/475,861 2001-04-24 2002-04-23 Progenitor cell populations , expansions thereof, and growth of non-hematopoietic cell types and tissues therefrom Abandoned US20040137612A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/828,575 US9644182B2 (en) 2001-04-24 2007-07-26 Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28570101P 2001-04-24 2001-04-24
US32811001P 2001-10-11 2001-10-11
PCT/CA2002/000550 WO2002086104A1 (en) 2001-04-24 2002-04-23 Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/828,575 Continuation US9644182B2 (en) 2001-04-24 2007-07-26 Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom

Publications (1)

Publication Number Publication Date
US20040137612A1 true US20040137612A1 (en) 2004-07-15

Family

ID=26963332

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/475,861 Abandoned US20040137612A1 (en) 2001-04-24 2002-04-23 Progenitor cell populations , expansions thereof, and growth of non-hematopoietic cell types and tissues therefrom
US11/828,575 Active 2024-12-22 US9644182B2 (en) 2001-04-24 2007-07-26 Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/828,575 Active 2024-12-22 US9644182B2 (en) 2001-04-24 2007-07-26 Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom

Country Status (9)

Country Link
US (2) US20040137612A1 (da)
EP (2) EP2105497B1 (da)
JP (1) JP2004531256A (da)
AT (1) ATE433484T1 (da)
CA (2) CA2444959C (da)
DE (1) DE60232585D1 (da)
DK (2) DK2105497T3 (da)
ES (1) ES2328460T3 (da)
WO (1) WO2002086104A1 (da)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050148074A1 (en) * 2003-02-11 2005-07-07 Davies John E. Progenitor cells from wharton's jelly of human umbilical cord
US20050255592A1 (en) * 2004-04-23 2005-11-17 Bioe, Inc., A Minnesota Corporation Multi-lineage progenitor cells
US20060040392A1 (en) * 2004-04-23 2006-02-23 Collins Daniel P Multi-lineage progenitor cells
US20060241376A1 (en) * 2003-04-24 2006-10-26 Koninklijke Philips Electronics N.V. Non-invasive left ventricular volume determination
US20070042491A1 (en) * 2005-08-18 2007-02-22 Karp Jeffrey M Amplification of cell populations from embryonic stem cells
US20070249047A1 (en) * 2006-04-17 2007-10-25 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Respiratory Epithelial Cells
US20080113434A1 (en) * 2004-03-05 2008-05-15 Davies John E Serum-Free Suspension Culture System For Mesenchymal Progenitor Cells
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
US20090181007A1 (en) * 2006-04-14 2009-07-16 Luisa Gennero Culture medium and pharmaceutical composition for regenerating cartilage tissue, a method, uses and products related thereto
US20090275127A1 (en) * 2005-12-22 2009-11-05 Jane Ennis Viable cells from frozen umbilical cord tissue
US20090285842A1 (en) * 2006-05-05 2009-11-19 Davies John E Immune privileged and modulatory progenitor cells
US20090291494A1 (en) * 2008-05-21 2009-11-26 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Pancreatic Cells
US20090311781A1 (en) * 2006-08-02 2009-12-17 Michal Amit Methods of expanding embryonic stem ceels in a suspension culture
US20100111910A1 (en) * 2006-07-12 2010-05-06 Angioblast Systems, Inc. Treatment of excessive neovascularization
US20100221829A1 (en) * 2005-08-29 2010-09-02 Technion Research & Development Foundation Ltd. Media for culturing stem cells
US7807458B2 (en) 2003-01-30 2010-10-05 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Multilineage-inducible cells and uses thereof
US20110104126A1 (en) * 2008-05-14 2011-05-05 Public University Corporation Yokohama City Univer Human Hepatic Stem Cell, Method for Preparation of the Same, Method for Induction of Differentiation of the Same, and Method for Utilization of the Same
US20110177023A1 (en) * 2008-04-21 2011-07-21 Jane Elizabeth Ennis Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US10071158B2 (en) 2005-04-26 2018-09-11 Lindis Biotech Gmbh Combination of the application of antibodies for immunostimulation together with glucocorticoids
WO2020061408A1 (en) * 2018-09-21 2020-03-26 Tarachon Llc Cartilage regeneration by synovial fluid-derived stem cells and their derivatives
US11608486B2 (en) 2015-07-02 2023-03-21 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11634677B2 (en) 2016-06-07 2023-04-25 Terumo Bct, Inc. Coating a bioreactor in a cell expansion system
US11667881B2 (en) 2014-09-26 2023-06-06 Terumo Bct, Inc. Scheduled feed
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11795432B2 (en) 2014-03-25 2023-10-24 Terumo Bct, Inc. Passive replacement of media
US11965175B2 (en) 2016-05-25 2024-04-23 Terumo Bct, Inc. Cell expansion

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2444959C (en) 2001-04-24 2014-07-08 Dolores Baksh Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom
JP4604238B2 (ja) * 2003-09-19 2011-01-05 独立行政法人産業技術総合研究所 燐酸カルシウムと生理活性物質を含有する高分子複合体、その製造方法及びそれを用いた医療用材料
CA2718174A1 (en) * 2008-03-03 2009-09-11 William Marsh Rice University Methods of fabricating enhanced tissue-engineered cartilage
EP2568991B3 (en) 2010-05-12 2018-11-28 ABT Holding Company Modulation of splenocytes in cell therapy
WO2012046065A2 (en) 2010-10-06 2012-04-12 Omnicyte Limited Culture method
CN105338989B (zh) 2013-04-12 2022-04-08 赛维里奥·拉弗朗切西卡 改进用于移植的器官
US20160193386A1 (en) * 2015-01-05 2016-07-07 Histogenics Corporation Neocartilage constructs using universal cells
EP3098304A1 (en) 2015-05-28 2016-11-30 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Hybrid cell detachment-free cell expansion system for adherent cell culturing and related proceeding protocol
NZ745530A (en) 2016-01-21 2023-03-31 Abt Holding Co Stem cells for wound healing
KR102557574B1 (ko) * 2016-05-17 2023-07-20 엘지전자 주식회사 디지털 디바이스 및 그 제어 방법

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6087113A (en) * 1991-06-18 2000-07-11 Case Western Reserve University Monoclonal antibodies for human mesenchymal stem cells

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850748A (en) * 1973-06-28 1974-11-26 Lilly Co Eli Method of producing animal cells in suspension culture
US4495288A (en) 1981-03-13 1985-01-22 Damon Biotech, Inc. Method of culturing anchorage dependent cells
US5763266A (en) * 1989-06-15 1998-06-09 The Regents Of The University Of Michigan Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5197985A (en) * 1990-11-16 1993-03-30 Caplan Arnold I Method for enhancing the implantation and differentiation of marrow-derived mesenchymal cells
US5226914A (en) * 1990-11-16 1993-07-13 Caplan Arnold I Method for treating connective tissue disorders
US6010696A (en) * 1990-11-16 2000-01-04 Osiris Therapeutics, Inc. Enhancing hematopoietic progenitor cell engraftment using mesenchymal stem cells
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5855610A (en) 1995-05-19 1999-01-05 Children's Medical Center Corporation Engineering of strong, pliable tissues
US7514074B2 (en) * 1997-07-14 2009-04-07 Osiris Therapeutics, Inc. Cardiac muscle regeneration using mesenchymal stem cells
CA2324350A1 (en) * 1998-03-23 1999-09-30 Zymogenetics, Inc. Cardiac-derived stem cells
RU2272839C2 (ru) 1998-11-09 2006-03-27 Консорцио Пер Ла Джестионе Дель Чентро Ди Биотекнолоджие Аванцате Бессывороточная среда для культуры клеток, используемых для реконструкции костных и хрящевых сегментов (варианты)
DK1226233T3 (da) * 1999-08-05 2011-10-03 Abt Holding Co Multipotente voksne stamceller og fremgangsmåder til isolering heraf
CA2444959C (en) 2001-04-24 2014-07-08 Dolores Baksh Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom
KR100534215B1 (ko) 2003-11-11 2005-12-08 (주)히스토스템 냉동 보관된 제대혈로부터 중간엽 줄기세포의 분리 및 배양 방법

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6087113A (en) * 1991-06-18 2000-07-11 Case Western Reserve University Monoclonal antibodies for human mesenchymal stem cells

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7807458B2 (en) 2003-01-30 2010-10-05 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Multilineage-inducible cells and uses thereof
US20090269318A1 (en) * 2003-02-11 2009-10-29 Tissue Regeneration Therapeutics, Inc. Progenitor cells from wharton's jelly of human umbilical cord
US9567564B2 (en) 2003-02-11 2017-02-14 Tissue Regeneration Therapeutics Inc. Progenitor cells from Wharton's jelly of human umbilical cord
US9611456B2 (en) 2003-02-11 2017-04-04 Tissue Regeneration Therapeutics Inc. Progenitor cells from wharton'S jelly of human umbilical cord
US8481311B2 (en) 2003-02-11 2013-07-09 Tissue Regeneration Therapeutics Inc. Progenitor cells from wharton's jelly of human umbilical cord
US20050148074A1 (en) * 2003-02-11 2005-07-07 Davies John E. Progenitor cells from wharton's jelly of human umbilical cord
US7547546B2 (en) 2003-02-11 2009-06-16 Tissue Regeneration Therapeutics Inc. Progenitor cells from wharton's jelly of human umbilical cord
US20060241376A1 (en) * 2003-04-24 2006-10-26 Koninklijke Philips Electronics N.V. Non-invasive left ventricular volume determination
US20080113434A1 (en) * 2004-03-05 2008-05-15 Davies John E Serum-Free Suspension Culture System For Mesenchymal Progenitor Cells
US20100028851A1 (en) * 2004-04-23 2010-02-04 Bioe, Inc. Multi-Lineage Progenitor Cells
US20050255592A1 (en) * 2004-04-23 2005-11-17 Bioe, Inc., A Minnesota Corporation Multi-lineage progenitor cells
US20060040392A1 (en) * 2004-04-23 2006-02-23 Collins Daniel P Multi-lineage progenitor cells
US7622108B2 (en) 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
US8163275B2 (en) 2004-04-23 2012-04-24 Bioe Llc Multi-lineage progenitor cells
US7670596B2 (en) 2004-04-23 2010-03-02 Bioe, Inc. Multi-lineage progenitor cells
US10071158B2 (en) 2005-04-26 2018-09-11 Lindis Biotech Gmbh Combination of the application of antibodies for immunostimulation together with glucocorticoids
US10576149B2 (en) 2005-04-26 2020-03-03 Lindis Biotech Gmbh Combination of the application of antibodies for immunostimulation together with glucocorticoids
US20070042491A1 (en) * 2005-08-18 2007-02-22 Karp Jeffrey M Amplification of cell populations from embryonic stem cells
US20100221829A1 (en) * 2005-08-29 2010-09-02 Technion Research & Development Foundation Ltd. Media for culturing stem cells
US11512283B2 (en) 2005-08-29 2022-11-29 Technion Research & Development Foundation Limited Media for culturing stem cells
US8476070B2 (en) 2005-08-29 2013-07-02 Technion Research & Development Foundation Limited Media for culturing stem cells
US9404079B2 (en) 2005-08-29 2016-08-02 Technion Research & Development Foundation Limited Media for culturing stem cells
US10385312B2 (en) 2005-08-29 2019-08-20 Technion Research & Development Foundation Limited Media for culturing stem cells
US8278102B2 (en) 2005-12-22 2012-10-02 Jane Ennis Viable cells from frozen umbilical cord tissue
US8790923B2 (en) 2005-12-22 2014-07-29 Jane E. Ennis Viable cells from frozen umbilical cord tissue
US20090275127A1 (en) * 2005-12-22 2009-11-05 Jane Ennis Viable cells from frozen umbilical cord tissue
US20090181007A1 (en) * 2006-04-14 2009-07-16 Luisa Gennero Culture medium and pharmaceutical composition for regenerating cartilage tissue, a method, uses and products related thereto
US20070249047A1 (en) * 2006-04-17 2007-10-25 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Respiratory Epithelial Cells
US7727763B2 (en) 2006-04-17 2010-06-01 Bioe, Llc Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
US8277794B2 (en) 2006-05-05 2012-10-02 Davies John E Immune privileged and modulatory progenitor cells
US8900573B2 (en) 2006-05-05 2014-12-02 John E. Davies Immune privileged and modulatory progenitor cells
US20090285842A1 (en) * 2006-05-05 2009-11-19 Davies John E Immune privileged and modulatory progenitor cells
US20100111910A1 (en) * 2006-07-12 2010-05-06 Angioblast Systems, Inc. Treatment of excessive neovascularization
US20090311781A1 (en) * 2006-08-02 2009-12-17 Michal Amit Methods of expanding embryonic stem ceels in a suspension culture
US9040297B2 (en) * 2006-08-02 2015-05-26 Technion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
US9834749B2 (en) 2006-08-02 2017-12-05 Technion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
US20180066227A1 (en) * 2006-08-02 2018-03-08 Technion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
US10968427B2 (en) * 2006-08-02 2021-04-06 Teehnion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
US9005599B2 (en) 2008-04-21 2015-04-14 Tissue Regeneration Therapeutics Inc. Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US20110177023A1 (en) * 2008-04-21 2011-07-21 Jane Elizabeth Ennis Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US9498544B2 (en) 2008-04-21 2016-11-22 Tissue Regeneration Therapeutics Inc. Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US20110104126A1 (en) * 2008-05-14 2011-05-05 Public University Corporation Yokohama City Univer Human Hepatic Stem Cell, Method for Preparation of the Same, Method for Induction of Differentiation of the Same, and Method for Utilization of the Same
US20090291494A1 (en) * 2008-05-21 2009-11-26 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Pancreatic Cells
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11746319B2 (en) 2010-10-08 2023-09-05 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11773363B2 (en) 2010-10-08 2023-10-03 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11708554B2 (en) 2013-11-16 2023-07-25 Terumo Bct, Inc. Expanding cells in a bioreactor
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11795432B2 (en) 2014-03-25 2023-10-24 Terumo Bct, Inc. Passive replacement of media
US11667881B2 (en) 2014-09-26 2023-06-06 Terumo Bct, Inc. Scheduled feed
US11608486B2 (en) 2015-07-02 2023-03-21 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11965175B2 (en) 2016-05-25 2024-04-23 Terumo Bct, Inc. Cell expansion
US11634677B2 (en) 2016-06-07 2023-04-25 Terumo Bct, Inc. Coating a bioreactor in a cell expansion system
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11999929B2 (en) 2016-06-07 2024-06-04 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
WO2020061408A1 (en) * 2018-09-21 2020-03-26 Tarachon Llc Cartilage regeneration by synovial fluid-derived stem cells and their derivatives

Also Published As

Publication number Publication date
JP2004531256A (ja) 2004-10-14
CA2444959C (en) 2014-07-08
WO2002086104A1 (en) 2002-10-31
DK2105497T3 (da) 2019-06-24
ATE433484T1 (de) 2009-06-15
EP1383869A1 (en) 2004-01-28
ES2328460T3 (es) 2009-11-13
EP2105497A1 (en) 2009-09-30
EP1383869B1 (en) 2009-06-10
EP2105497B1 (en) 2019-04-03
US9644182B2 (en) 2017-05-09
CA2843001A1 (en) 2002-10-31
DK1383869T3 (da) 2009-10-05
CA2444959A1 (en) 2002-10-31
US20080020459A1 (en) 2008-01-24
DE60232585D1 (de) 2009-07-23

Similar Documents

Publication Publication Date Title
US9644182B2 (en) Progenitor cell populations, expansion thereof, and growth of non-hematopoietic cell types and tissues therefrom
Lindroos et al. The potential of adipose stem cells in regenerative medicine
JP6257559B2 (ja) 脈管周囲の間葉系前駆細胞
JP5732011B2 (ja) 非骨軟骨性の間葉組織由来の多能性細胞の同定および単離
CA2600653C (en) Pluripotent stem cell derived from cardiac tissue
EP1725656B1 (en) Serum-free suspension culture system for mesenchymal progenitor cells
KR20040094910A (ko) 개선된 지방세포 분화된 지방 유래 성체 줄기세포 및 이의용도
KR20070100908A (ko) 내피 세포의 특성을 나타내는 지방 유래 성인 스트로마세포
JP3762975B2 (ja) 単球由来多能性細胞momc
Baksh et al. A non‐contact suspension culture approach to the culture of osteogenic cells derived from a CD49elow subpopulation of human bone marrow‐derived cells
US20080241111A1 (en) Pluripotent Stem Cell Derived from Cardiac Tissue
US10400211B1 (en) Cell composition for tissue regeneration
AU2002250754A1 (en) Progenitor cell populations, expansion thereof, and growth of non-hematopietic cell types and tissues therefrom
JP3928881B2 (ja) 単球由来多能性細胞momc
Thitiset et al. Isolation and cellular properties of mesenchymal stem cells from human periosteum
Mannerstrom et al. The Potential of Adipose Stem Cells in Regenerative Medicine
Rosocha et al. Mesenchymal Stem Cells: Potential for Tissue Regeneration

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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