WO2003004626A1 - A bioprocess for the generation of cells derived from spheroid-forming cells - Google Patents

A bioprocess for the generation of cells derived from spheroid-forming cells Download PDF

Info

Publication number
WO2003004626A1
WO2003004626A1 PCT/CA2002/001022 CA0201022W WO03004626A1 WO 2003004626 A1 WO2003004626 A1 WO 2003004626A1 CA 0201022 W CA0201022 W CA 0201022W WO 03004626 A1 WO03004626 A1 WO 03004626A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
sep
spheroid
aggregation
Prior art date
Application number
PCT/CA2002/001022
Other languages
French (fr)
Other versions
WO2003004626B1 (en
WO2003004626A3 (en
WO2003004626A9 (en
Inventor
Peter W Zandstra
Stephen Dang
Original Assignee
Peter W Zandstra
Stephen Dang
Cardion Ag
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 Peter W Zandstra, Stephen Dang, Cardion Ag filed Critical Peter W Zandstra
Priority to EP02745012A priority Critical patent/EP1407002A2/en
Priority to AU2002317110A priority patent/AU2002317110B2/en
Priority to CA002488550A priority patent/CA2488550A1/en
Publication of WO2003004626A1 publication Critical patent/WO2003004626A1/en
Publication of WO2003004626A9 publication Critical patent/WO2003004626A9/en
Publication of WO2003004626A3 publication Critical patent/WO2003004626A3/en
Publication of WO2003004626B1 publication Critical patent/WO2003004626B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/08Flask, bottle or test tube
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/10Petri dish
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M25/00Means for supporting, enclosing or fixing the microorganisms, e.g. immunocoatings
    • C12M25/01Drops
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M27/00Means for mixing, agitating or circulating fluids in the vessel
    • C12M27/02Stirrer or mobile mixing elements
    • 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
    • C12N5/0012Cell encapsulation
    • 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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/70Polysaccharides
    • C12N2533/76Agarose, agar-agar

Abstract

The present inventors identified aggregation of embryonic stem cells and embryoid bodies (EBs) as the cause of the difficulty in generating large numbers of the embryonic stem cells (ES) cell-derived tissues. To counter this, the invention provides a novel bioprocess where aggregation of spheroid-forming cells, such as embryonic stem cells and spheroids, such as EBs is controlled, such as by encapsulation of within a matrix. As a result, EBs can be generated with high efficiency and cultured in high cell density, well-mixed systems. Well-mixed conditions facilitate measurement and control of the bulk media conditions and allow for the use of scalable bioreactor systems for clinical production of tissue. Therefore, the invention enables generation of ES cell-derived tissue on a clinical scale. The invention is also applicable to any spheroid-forming cells and other types of pluripotent cells.

Description


  



  TITLE : A BIOPROCESS FOR THE GENERATION OF CELLS FROM
SPHEROID-FORMING CELLS
CROSS REFERENCE TO RELATED APPLICATIONS
This application hereby claims the benefit of priority from Canadian
Patent Application No. 2,351, 156 filed July 4,2001 and United States
Provisional Patent Application No. 60/302,706 filed July 5,2001, both entitled, "A Bioprocess For The Generation Of Pluripotent Cell Derived Cells And
Tissues". Both of these references are herein incorporated by reference.



  FIELD OF THE INVENTION
The invention relates to a process for generating cells and tissues, preferably cells and tissues derived from pluripotent cells or spheroid-forming cells. In one embodiment the pluripotent cells are embryonic stem cells.



  Products and applications resulting from said process are also encompassed within the scope of this application.



  BACKGROUND OF THE INVENTION
Tissue engineering, transplantation therapy, gene therapy, and drug discovery promise to revolutionize the medical paradigm and herald the coming of the"biotechnology era". However, development and implementation of these technologies depend upon cell-based approaches that require a plentiful supply of human cells : a supply that unfortunately does not currently exist. Therefore, identifying sources of human cells that can be practically obtained in sufficient quantities is of paramount importance.



  Spheroid-forming cells, preferably pluripotent cells such as human embryonic stem cells (Thomson, Itskovitz-Eldor et   al.    1998), neural stem cells   (Clarke,   
Johansson, et   al.    2000), multipotent adult progenitor cells (Schwartz, Reyes, et   al.    2002), and others may be able to provide tissue in this manner.



   The potential utility of pluripotent cells, in particular, the utility of embryonic stem (ES) cells, is derived from their demonstrated ability to differentiate into any other cell type in the body (Evans and Kaufman, 1981).



  Derived from the early embryo, ES cells can be grown and manipulated in vitro. ES   cells are pluripotent cells    that have the ability to differentiate into any cell type in the body (Evans and Kaufman 1981). They also have the ability to grow and multiply indefinitely while maintaining their   pluripotentiality.   



  Theoretically, these traits make ES cells an unlimited source of any cell type and therefore a very attractive tissue source for other biotechnological applications (Zandstra and Nagy 2001).



   Generation of various differentiated cell types (cardiac myocytes, hematopoietic cells, neurons, hepatocytes, and others) from murine ES cells   (e. g. , Wang, Clark et al. 1992; Rohwedel, Maltsev et al. 1994; Bain, Kitchens    et al. 1995;   Palacios,      Golunski    et   al.    1995; Choi, Kennedy et   al.    1998;   Fleischmann,    Bloch et   al.    1998; Tropepe,   Sibilia    et   al.    1999) as well as from   human ES cells (e. g. , Jones and Thomson 2000; Thomson et al. 1998;   
Mummery et al 2002; Levenberg et   al.    2002; Reubinoff et   al.    2001; Assady et al 2001) has been extensively reported.



   In general, production of these various cell types from ES cells require a complex multi-step process, starting with the expansion of ES cells to adequate numbers. Next, ES cells would be differentiated to the desired cell type (s). Finally, the desired cells would be selected and purified from the remaining cells before they could be utilized for various applications.



   In vitro, differentiating ES cells follow a reproducible temporal pattern of development that in many ways recapitulates early embryogenesis (Keller 1995). As ES cells and their derivatives proliferate and differentiate, they typically form spherical tissue-like structures (spheroids) called Embryoid
Bodies (EBs). Other pluripotent stem cells such as neural stem cells have also been described to form spheroids. In the case of neural stem cells these spheroids are termed neurospheres   (Kalyani,    Hobson, and Rao, 1997). In the case of ES cells, over time, EBs increase in cell number and complexity as cells from the three embryonic germ layers are formed (Keller 1995).

   The ability to control and manipulate the formation of spheroids from pluripotent cells and other types of cells that are derived directly, or indirectly from human tissue, as well as the interactions between the spheroids and between the   spheroid-forming cells (i. e. , prevent or inhibit the aggregation of spheroids)    would allow for the control of interactions between these cells during proliferation and differentiation, and thus be an important development for the design of technologies for the in vitro differentiation of pluripotent cells.

   For example, the generation of"chimeric spheroids"or"chimeric EBs"defined as multicellular, spherical structure comprising a mixture of differentiated cells and pluripotent cells (Perkins, 1998), has been shown to be an effective approach to influence the development of the cell types in from pluripotent cells (Clarke, Johansson et al. 2000).



   Current ES cell differentiation systems include cultures initiated with an
ES cell suspension in liquid media (LSC), or methylcellulose (semi-solid) media (MC), or attached to a surface in liquid media, or with multi-ES cell aggregates formed in"hanging drop"cultures (HD) where ES cells are aggregated in hanging drops for 2 days before transfer to liquid culture (see
Figure 1). These systems are adequate for small-scale laboratory purposes but are not amenable to clinical production because of deficiency in three important areas: a) the ability to measure and control the   extracellular    environment,   b)    scalability, and c) cell density.



   These systems are static and batch-style cultures that result in formation of spatial and temporal gradients in nutrients and metabolic products. These gradients make measurement and process control difficult because these processes depend on sampling measurements that need to reflect the conditions throughout the culture. The inability to control the cell culture environment will affect product purity and reproducibility, as well as the   cell types that can be readily generated [e. g. , for oxygen (Maltepe, Schmidt, et    al. 1997), glucose (Soira, 2001) and many other biological and physicochemical factors (Zandstra and Nagy, 2001)]. In the case of methylcellulose culture, the semi-solid media also hinders measurement and manipulation.

   Regulatory approval for the use of any biotechnology product demands consistency in the method of production and in the final product itself (http://www. fda. gov/cdrh/tisseng/te6.   html).    This is not possible when measurement and control are impeded or absent.



   Second, clinical or industrial scale production of cells from the current culture systems is not practical. The current systems are typically carried out in petri dishes where cells grow in only a thin layer of media. These systems are essentially two-dimensional with respect to the EBs; therefore, unrealistically large surface areas would be required for industrial scale production. It can be estimated that several billion cells would be need for applications such as cardiac cell therapy (Zandstra and Nagy, 2001); this scale of cell production is highly impractical, if not impossible using current technologies.



   Third, current liquid suspension cultures (LSC) require EBs to be cultured at relatively low density. Higher cell densities can be achieved in methylcellulose cultures, where semi-solid media reduces the likelihood of cell aggregation, however media supplementation of methylcellulose cultures is troublesome and increased cell density would demand more frequent supplementation.



   Therefore, there is a need to develop methods for efficient growth and differentiation of spheroid-forming cells, preferably pluripotent cells, such as
ES cells.



  SUMMARY OF THE INVENTION
Because of the limitations reviewed above, one ideal differentiation culture system for clinical scale production of EBs or other spheroids would be the stirred tank reactor or other scalable, well-mixed and controlled bioreactor systems such as a fluidized bed reactor. While the standard methods are limited to two-dimensional growth because of static conditions, a controlled bioreactor could suspend the cells evenly throughout the volume (in threedimensions). This configuration of system can be easily scaled in size to accommodate the need for increased production of cells. In addition, stirring ensures homogenous media conditions throughout the reactor that would facilitate measurement and control of the   extracellular    environment.



   Despite the importance of stirred or other well-mixed bioreactors, to date there have been no reports of successful use of these systems for ES cell differentiation culture. ES cells added directly to stirred culture quickly aggregate into large cell clumps within 24 hours and cell growth and differentiation within these clumps are severely impaired (Wartenberg et al.   



  (1998) ). However, it was not known whether aggregation was the cause or    result of a different problem. Differentiation of ES cells in stirred culture is therefore a non-trivial task; however the ability to do so would greatly facilitate clinical and industrial scale production of cells for therapeutic purposes. ES cells cultured in stirred bioreactors fail to generate EBs in an efficient manner.



   This patent application is based upon a novel technology that overcomes one or more of the limitations of the prior art: a) the ability to measure and control the   extracellular    environment,   b)    scalability, and c) cell density and allows for the scalable generation of cells and/or tissue derived from spheroid-forming, preferably pluripotent, preferably ES cells.



   The present inventors have determined that ES cells cultured in stirred bioreactors fail to generate EBs in an efficient manner because the cells aggregate forming large cell clumps that results in poor cell proliferation and incomplete cell differentiation and that controlling aggregation can overcome this problem. As such, in one aspect, the present invention provides a process to control cellular and spheroid aggregation of spheroid-forming cells, such as
ES cells or neuronal stem cells, to enable the use of a stirred tank reactor or other scalable, well-mixed and controlled bioreactor systems such as a fluidized bed reactor or other liquid suspension system. The invention can also have benefits in other systems, especially liquid systems such as LSC.



   In one embodiment, the invention provides a method of culturing spheroid-forming cells, preferably pluripotent cells, preferably ES cells, under controlled cell aggregation conditions that enable spheroid formation. In another embodiment the method is done in a bioreactor system by controlling cellular aggregation. In one embodiment the cells are differentiating cells. In yet another embodiment, the invention provides a method of generating or obtaining cells derived from spheroid-forming cells cultured under controlled aggregation conditions. Although a preferred embodiment of the invention is with ES cells, the invention is not limited to ES cell derived cells and tissues.



  It is equally applicable to any spheroid-forming cell type, preferably pluripotent cell type that can differentiate as a cell cluster or aggregate, or form spheroid bodies, such as adult pluripotent cells (Schwartz, Reyes, et al. 2002; Clarke,
Johansson et al. 2000) embryonic germ cells (EG cells) (reviewed in
Thomson and Odorico, 2000), early primitive ectoderm-like cells (EPL) (Pelton and Sharma, 2002), and neuronal stem cells. In one embodiment, the invention can also apply to other cell types where cell aggregation or control of the cell mircoenvironment is indicated.

   As such the embodiments of the invention as outlined below in relation to embryonic stem cells also apply to other pluripotent cells where controlling aggregation of the cells at various points of expansion and or differentiation can enhance the efficiency of said expansion, spheroid formation or differentiation and are intended to be encompassed within the scope of the present invention.



   In one embodiment, the invention provides a method of generating cells derived from spheroid-forming or pluripotent cells comprising culturing the cells, for example pluripotent cells, such as ES cells, EG cells, EPL cells, and adult pluripotent cells (Schwartz, Reyes, et al. 2002), while controlling cellular and spheroid aggregation. In a preferred embodiment, the method is conducted under conditions that permit cell differentiation and proliferation. In another embodiment, the cells are encapsulated to prevent aggregation with neighboring cells, spheroids or cells contained in separate capsules. In another embodiment the aggregation between specific cell types is controlled (to enable aggregation necessary for spheroid formation, where applicable) and the aggregation between spheroids of these cells is prevented.

   In yet another embodiment the kinetics of aggregation is controlled.



   In one embodiment, the invention provides a method of generating cells from spheroid-forming cells, preferably embryonic stem cells, comprising culturing the spheroid-forming, preferably embryonic stem cells under conditions that enable spheroid or embryoid body formation and cell differentiation while controlling cell aggregation.



   In a preferred embodiment, the spheroid-forming cells, such as embryonic stem cells are encapsulated to control cell aggregation such that each capsule will transiently contain and give rise to one spheroid or embryoid body. In another embodiment, each capsule contains a predetermined or desired number of spheroid-forming or ES cells that are permitted to aggregate and together give rise to a single spheroid or EB. Preferably, the undifferentiated spheroid-forming cells, such as embryonic stem cells are first encapsulated to prevent aggregation between the cells contained within separate capsules. The encapsulated cells are then cultured under conditions that enable cell proliferation and optionally differentiation, leading to spheroid or embryoid body formation.

   In a more preferred embodiment, emulsification of cells with agar in inert silicon oil is used to encapsulate the cells. In a most preferred embodiment, this emulsification results in the generation of agarose microcapsules containing cells. In yet another embodiment the process of encapsulation allows for the control of spheroid-forming cell aggregation by interfering with cell surface receptor binding. In one embodiment, the process of encapsulation allows for the control of spheroid-forming or ES cell aggregation by interfering with E-cadherin mediated cell aggregation.



   In another embodiment, the invention provides a method wherein the embryonic stem cells are cultured under conditions wherein embryoid bodies and/or differentiated embyronic stem cells can be formed. For example, in some ES cell lines multiple ES cells may be preferred for EB formation.



  Following these permissive steps, cells are transferred to conditions that prevent aggregation of embryoid bodies. In one embodiment, the method further comprises a step wherein the differentiated embryonic stem cells and/or tissues of interest are selected and harvested. In a preferred embodiment, the cells and/or tissues of interest are cardiomyocytes or cardiac tissue or hematopoietic cells or tissue or endothelial cells or tissues.



   In another embodiment, the invention provides an embryonic stem cell derived cell culture obtained using the method of the invention. In a further embodiment, the invention provides embryonic stem cell derived cells and tissues obtained using the method of the invention.



   In yet another embodiment the invention provides a method to identify factors, e. g. any variant, condition, substance, that affect embryonic stem cell differentiation and/or embryoid body formation, said method comprising culturing the embryonic stem cells while preventing embryoid body and embryonic stem cell aggregation, in the presence of the factor to be tested and then detecting the effect of the variant on embryonic stem cell differentiation and embryoid body formation. In a further embodiment, of said method, the effect on embryonic stem cell differentiation and embryoid body formation is compared to a control culture, preferably a negative control wherein embryonic stem cells are cultured under the same conditions except in the absence of the factor to be tested.

   The invention also provides a method of identifying factors that affect cell proliferation or differentiation in any cell culture by encapsulating said cells. The invention also encompassed a method of preventing cell aggegation in any cell type by encapsulation.



   Applications and uses of the method and initiating and resulting cells, tissues, cultures and bioreactors or culture systems are also encompassed within the scope of the present invention.



   Other features and advantages of the present invention will become apparent from the following detailed description. For instance where reference is made to embryoid bodies or embryonic stem cells, the invention can also be applied to spheroid-forming cells and spheroids. It should be understood, however, that the detailed description and the specific examples while indicating preferred embodiments of the invention are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.



  BRIEF DESCRIPTION OF THE DRAWINGS
The invention will now be described in relation to the non-limiting drawings in which:
Figure 1 is a schematic drawing of the current technologies used to culture pluripotent stem cells. (A) Hanging drop (HD) cultures typically consist of a defined number of cells allowed to aggregate in small fluid volumes that hang from the tops of tissue culture dishes. Liquid suspension cultures (LSC) (B) are typically low-density suspension cultures of individual cells or aggregates of cells. When individual ES cells are put into the bottoms of 96 well plates to measure EB developments these cultures are termed single cell liquid suspension cultures (scLSC) (C).

   Methycellulose (MC) (or similar solid or semi solid cultures) are cultures where cells are enveloped in semi-or semisolid media that prevents aggregation, for example by preventing the collision of cells or cell aggregates due to its high viscosity (D). 



  Figure 2 is a schematic diagram of one embodiment of the method to control spheroid-forming   cell and/or plurioptent cell    and spheroid aggregation. Test cell populations (either individual cells or controlled aggregates of cells) are encapsulated, put inside a controlled stirred suspension bioreactor   (Encapsulated Stirred Culture (ESC) ), or in any type of encapsulated liquid    suspension culture (ELSC) where, because of the encapsulation technology, they are prevented from aggregating, and allowed to proliferate and differentiate into the desired cell type.



     Figure 3 is a graph illustrating the affect of initial ES cell density (i. e. , number    of ES cells per unit volume) on EB efficiency (ratio of number of cell aggregates to number of input ES cells) over time for A: liquid suspension culture (LSC) and B: methylcellulose (MC) culture. Cultures were initiated with ES cell densities: 102,   103,      104,    105 cells/ml.



  Figure 4 is a linear graph illustrating cells per EB (y-axis) over time (x-axis).



  EBs were initiated with: 1 (scLSC), 100 (100 cells per drop in the hanging drop cultures,   HD100),    and 1000 (1000 cells per drop in the hanging drop cultures, HD1000) ES cells/EB. Despite initial differences in   cells/EB,    all EBs grew to approximately the same size by day 12.



  Figure 5 (A) illustrates the method of quantifying degree of aggregation (DOA) between two EBs, defined as the ratio of the diameter of the interface to the overall length of the two EB system. DOA is calculated by dividing the interface diameter by the overall length of the two EB system, yielding a scale between 0 (no aggregation) and 1 (fully aggregated). (B) A representative photomicrograph series of two EBs aggregating over time is shown with corresponding DOA measurements at each time interval. Two types of ES cell derived EBs were used in this experiment, one that was not fluorescent and one that was constitutively fluorescent (light EB in the photomicrograph)). 



  Figure 6 is a linear graph illustrating the effects of the   chemicar additives    cytochalasin D (CytoD), mitomycin C (MitoC), E-cadherin blocking peptide (ECadAB), and a control, on the aggregation kinetics, measure using the
DOA, of EBs. A representative photo taken at 24 hours of culture is shown for each treatment group.



  Figure 7 panel A is a bar graph illustrating percent E-cadherin expressing cells (y-axis) versus time (x-axis). Figure 7 panel B is a linear graph illustrating aggregation kinetics of EBs differentiated for 0,2, 4, and 6 days.



  Figure 8 panel A are photographs of ES cells encapsulated inside agarose microcapsules designed to have specific diameters. Shown are capsules of mean diameter 35 50, and 85   um.    The scale is indicated by the straight lines at the bottom of each photograph which each represent   100, um. In    some capsules single ES cells were encapsulated   ( (35    and 50   Fm capsules), while    in other instances, a plurality of ES cells were encapsulated (85   jim    capsules).



  Figure 8 panel B is a photograph series of differentiating ES cells forming EBs inside the agarose microcapsules. Day 0 shows encapsulated ES cells, day 2 shows day 2 EBs, day 4 shows EBs emerging, as desired, from the agarose microcapsules.



  Figure 9 is a series of flow cytometry plots showing phenotypic detection of endothelial cells generated from encapsulated ES cells grown in stirred suspension bioreactors using flow cytometric based cell surface expression of various endothelial cell markers. Figure 9 panel A shows cells expressing   Flk-1    (y-axis) and CD34 (x-axis). Cells expressing either   Flk-1    or CD34 were gated and the percent of cells expressing these markers is indicated. Figure 9 panel B shows CD31 expression by ungated cells. Figure 9 panel C shows
CD31 expression by gated cells. CD34 and/or   Flk-1    expression together with
CD31 expression identifies endothelial cells.



  Figure 10 Panel A shows kinetic changes of the percentage of the MF-20 positive cells during the course of differentiation and selection with G418 of transgene-containing ES cells cultured in stirred suspension bioreactors.



  Figure 10 panel B shows morphologic and structural analysis of bioreactorgrown ES cell-derived cardiomyocytes. The cells exhibit a mature sarcomeric organization and Z-banding (arrow). Figure 10 panel C shows a cross section of a"cardiac body" (a spheroid of ES cell derived cardiac cells) generated in bioreactors.



  DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS "Adult stem cell"as used herein means an undifferentiated cell found in a tissue in an adult organism that can renew itself and can (with certain limitations) differentiate to yield specialized cell types of the tissue from which it originated or to yield specialized cell types in other tissues and organs.



  "Pluripotent   cell"as    used herein refers to a cell that can self renew and differentiate as a cell cluster or aggregate into one or more types of cells   and/or    tissue. Examples of such cells include but are not limited to embryonic stem cells, embryonic germ cells, early primitive ectoderm-like cells and derivatives, multiple adult progenitor cells, adult or embryonic neural stem cells (NSC) (Toma and Akhavan, 2001; Hitoshi, Tropepe, Sibilia, et al. 1999) adult   mesenchymal    stem cells (MSC), mesenchymal adult pluripotent stem cells, sometimes referred to as MAPC (Schwartz, Reyes, et al. 2002),   ductal    stem cells (Suzuk, Zheng et al. 2002), muscle derived stem cells (McKinney
Freeman, Jackson et al. 2002).



  "Embryonic stem cell"as used herein refers to pluripotent cells derived from the inner cell mass of a blastocyst, or derived by other methodologies (Jones   and Thomson (2000) ) and shown to be able to contribute to development of    multiple tissues.



  "Embryonic stem cell derived cells and tissues"as used herein refers to any cells or tissues that are derived from embryonic stem cells. The term   "pluripotent    cell derived cells and tissues"as used herein refers to any cells or tissues that are derived from pluripotent cells or tissues. The term"spheroid forming cell derived cells and tissues"as used herein refers to any cells or tissues that are derived from spheroid-forming cells or tissues.



  "Spheroid-forming   cell"as    used herein means cells that form multi-cellular aggregates consisting of more then a single cell when cultured in suspension "Spheroid"as used herein means a cellular structure consisting of more then a single cell that has initially developed from a single or from multiple cells.



  "Embryoid Body"as used herein means a"Spheroid"which is derived from embryonic stem cells.



  "Clonal Spheroid"as used herein means a cellular structure consisting of more then a single cell but which has been developed from a single cell.



  "Stem cell"as used herein means an"adult stem cell"or a"pluripotent cell".



  "Chimeric spheroid"is a"spheroid"made up of at least two distinct cell types. For example, in one embodiment it has been shown that the mixing ES cells with adult pluripotent cells can broaden the differentiation capacity of the adult pluripotent cells, for instance a chimeric spheroid can comprise two different   pluripotent cell    types, one an adult pluripotent cell (e. g.   neuronal      pluripotent cell)    and the other an ES cell. In one embodiment the ES cell can induce the other cell type to differentiate into other cell types such as cardiac cells.



  DESCRIPTION
The present invention is directed to a new method for generating spheroid-forming cell and/or pluripotent cell derived cells and tissues, such as for example cardiomyocytes, hematopoietic cells, endothelial cells, insulin producing beta cells, neuronal cells, glial cells, kidney cells, hepatocytes, vascular progenitor cells or derivatives (such as hematopoietic stem cells or endothelial cells), to name a few.



   The present inventors have shown that as cell density increases, cell aggregation occurs more readily, resulting in lower cell expansion and impaired cell differentiation (also see Dang, et   al.    2002). The present inventors have also hereby shown that cellular and spheroid aggregation is indeed the cause of the inability to culture spheroid-forming cells in a high density culture, such as in a liquid suspension culture (e. g. a stirred liquid bioreactor system). As such the inventors have shown that controlling cellular and/or spheroid aggregation can increase spheroid-forming efficiency.



   Thus in one embodiment, the invention provides a method of generating cells and/or tissues derived from spheroid-forming cells, preferably pluripotent cells comprising culturing spheroid-forming cells under controlled cell aggregation conditions. In another embodiment, such conditions enable spheroid formation in liquid suspension.



   In a preferred embodiment, the invention provides a method for efficient formation of spheroids or EBs and the culture of spheroid or EBs in suspension at higher cell densities. This is done by controlling cell aggregation. Thus, in another embodiment of the method of the invention, the spheroid-forming cells can be cultured under high density conditions.



  Although the invention will work at low cell density (even seeding the initial culture with 1 cell), it can also work with an initial cell density of about 102 to about 106   cells/ml, preferably    about   103    to about 106   cells/ml,    more preferably from about   104    to about   106 cells/ml.    The invention is not intended to be limited by such cell density values and are provided as a suitable example.



  These cells can then be cultured under controlled aggregation conditions to yield to form spheroids. As one can start with a culture with more cells than the prior art, the resulting number of spheroids formed will be greater than in cultures commenced at a lower cell density. In one embodiment, each spheroid may be cultured to about 30,000 to 35,000 cells per spheroid, preferably about 30,000 to 46,000 cells per spheroid. It should be noted that this is but one embodiment, and the invention is not limited to such numbers.



  A person skilled in the art could control conditions (such as time, temperature, media conditions, other environmental factors) to get lower or potentially higher numbers. Thus the present method permits generation of cells at an industrial scale.



   In one embodiment, the invention provides a scalable method for generating spheroid-forming cell or pluripotent cell derived cells and tissues, such as spheroid or ES derived cells and tissues for various uses, such as for transplantation. 



   In another embodiment, the invention provides a method of culturing spheroid-forming cells, such as   pluripotent    cells in a bioreactor system where the culture conditions can be measured and controlled. In yet another embodiment, the invention provides a scalable and controllable culture of spheroid-forming cells, such as pluripotent cells by allowing them to be cultured in liquid suspension cultures, such as stirred bioreactors, such as stirred liquid bioreactors. This is done by controlling cell aggregation. This improves the efficiency of generation of differentiated cells.



   In one embodiment, the bioreactor or culture system used in the invention is one that keeps the cells and/or spheroids in liquid suspension. In another embodiment, this culture system keeps the cells   and/or    spheroids in liquid suspension by stirring, but other methods or means can be used, such as by agitation of the system, use of various media or other environmental conditions.



   The term"controlling cell aggregation"refers to the process of effectively modulating the extent and kinetics of cell aggregation such that an endpoint, not effectively attainable without such control, is achieved. As used herein in relation to cell and/or spheroid or EB aggregation,"controlling cell aggregation"means that cell aggregation can be permitted or prevented as desired. For example, in one aspect of the invention, aggregation of spheroid-forming cells, such as ES cells, sufficient to induce spheroid or EB formation is permitted, and aggregation between cells within a spheroid or EB is permitted. However aggregation between spheroid-forming cells or ES cells beyond that determined to induce spheroid or EB formation is prevented.



  Aggregation between separate EBs is also prevented.



   Further, benefits of the invention (i. e. increasing efficiency of generating cells from spheroid-forming, pluripotent or ES cells) can be obtained with any degree of prevention of cellular and/or spheroid aggregation. Controlling aggregation may, in some application consist of preventing aggregation where the term"preventing"includes any inhibition of (i. e. that may prevent some, but not necessarily all) of spheroid-forming cell or pluripotent cell and/or spheroid or EB aggregation in a particular culture system. It would be appreciated by those skilled in the art upon reading the description herein that any prevention of spheroid, EB and/or spheroidforming cell or pluripotent cell aggregation would improve cell yields in the culture system and/or cellular differentiation.



     In one embodiment, "preventing, spheroid and/or ES cell and/or    spheroid or EB aggregation"means that after spheroid-forming cells or ES cells are cultured to form spheroids or embryoid bodies, there may be cellular aggregation between cells within a spheroid or embryoid body but not between cells of different spheroids or embryoid bodies or between spheroid or embryoid body themselves.



   In another aspect, the invention provides a method of generating cells derived from spheroid-forming cells wherein the spheroid-forming cells are cultured under conditions that enable spheroid formation, said conditions comprising culturing said spheroid-forming cells in liquid suspension under non-aggregating conditions.



   Thus in one embodiment, the invention provides a method of culturing spheroid-forming cells or ES cells comprising culturing the cells under conditions that promote spheroid or EB formation, including controlled aggregation of spheroid-forming or ES cells, then preventing cell and spheroid or EB aggregation.



   In another embodiment, the invention provides a method of controlling the production of spheroids from spheroid-forming cells by controlling the aggregation of such cells. In one embodiment, the spheroid-forming cells are pluripotent cells such as neural stem cells that form neurospheres, or are embryonic stem cell which form EBs.



   In a preferred embodiment the method of the invention does not adversely affect spheroid or EB formation, cell growth, or cell differentiation.



  Thus in another preferred embodiment, the method does not adversely effect the delivery of desired nutrients, oxygen, or cytokines. In yet another embodiment, the method can further comprise culturing the spehroid-forming or ES cells under conditions that promote differentiation of the cells and spheroid or EB formation. For instance, ES cells can be cultured in a suitable base media (such as ES cell media) without LIF. In another embodiment, the cells can be cultured under conditions that do not promote cell differentiation or further differentiation. For instance ES cells can be cultured in a suitable base media (Such as ES cell media) with LIF. Thus the method of the invention, by adjusting culture conditions can generate differentiated cells or be used to expand the existing cell culture initially or at any point of the method.

   For instance, in one embodiment culture conditions can change be changed during the method to control differentiation conditions. Examples of culture conditions are described further below.



   Examples of conditions that can be used in the present invention for culturing spheroid-forming cells, such as ES cells to control spheroid or EB and/or cell aggregation include but are not limited to:  (a) Methods for preventing physical association between cell aggregation molecules (i. e. encapsulation, such as described herein or Sefton et al.



   1997).



   (b) Methods that use reduced inorganic salt concentration as described in
Boraston et al. 1992 and Ko et al. 2001.



   (c) Methods that block surface aggregation molecules with specific peptides or other molecules, such as described in Burdsal et al. 1993  (d) Using genetically modified spheroid-forming or ES cells such as E    cadherin-null    ES cells such as described in Larue et al. 1996 or methods that inhibit E-cadherin expression or antagonize or inhibit E cadherin activity.



   (e) Addition of agents that prevent cell aggregation such as dextran sulfate or other sulfate polyanions as described in Dee et al. 1997
Methods that prevent cell aggregation by physically separating one ore more spheroid-forming cells to enable spheroid formation in distinct compartments such as described in the single cell liquid suspension culture (scLSC) method in this patent without limitation to a single cell.



   In one embodiment of the invention, the above methods can be used alone or in combination, ie, more than one method can be used to prevent spheroid, EB and/or spheroid-forming or ES   cell'and/or non-clonal    spheroidforming or non-clonal ES cell aggregation.



   The selected spheroid-forming or ES cell culturing strategy preferably does not affect the development and differentiation of pluripotent cells, preferably ES cells, or EBs in any undesired way.



   In a preferred method of the invention, the spheroid-forming or ES cells are encapsulated. In one embodiment, the average number of initial cells encapsulated can be predetermined or conditions can be adjusted to get a desired number of cells per capsule (for instance the capsule matrix, cell density, media or other environmental or capsule forming conditions). For instance, in one embodiment, the number of cells per capsule is 1-10,5-10, 15,5, or 1, but the invention is not limited by such examples. In one embodiment, the spheroid-forming or ES cell or cells can be encapsulated using methods such as described in Weaver et al. 1988 or Turcanu et   al.   



  2001 and then cultured under conditions that promote spheroid or EB formation and cellular differentiation, such as described in Keller et al, 1995 or
O'Shea et al. 1999. In another embodiment, the capsule comprises cells of at least two different cell types, such as an ES cell and an adult pluripotent cell to produce a chimeric spheroid.



   A variety of methods can be used to encapsulate cells. Although, the
Examples described herein use the Gel Microdrop Technique (GMD), patented and owned by One Cell Systems, Boston Massachusetts. The GMD technique was designed to encapsulate cells in agarose gel for the purpose of isolating individual cells with specific protein secretion profiles; however, the present invention adapted the process for encapsulation of spheroid-forming or ES cells and controlling cell and spheroid or EB aggregation. It is important to note that the microencapsulation of spheroid-forming cells such as ES cells for the purposes of the present invention does not depend upon the GMD technique as any method of encapsulation could be used.



   In one embodiment the size of the capsule can be controlled. In one aspect, this can determine the time at which a spheroid or EB emerges from the capsule. Preferably the size of the capsule is such that maintains the spheroid-forming cells and/or sperhoids in the capsule until aggregation cell surface markers or other aggregating factors (molecules, proteins, etc..) are no longer expressed. For instance, under certain conditions, as noted in the present examples, for ES cells, EBs that emerge after about 4 days no longer express E-cadherin and thus do not have a tendency to aggregate. In one approach, varying the rate of emulsification during the encapsulation process can control the size of the microcapsule. The different capsule sizes produced under each of the conditions tested are shown in Table 3.

   Other factors that may control the size of the microcapsules include the addition of surfactants to the encapsulation solution, the ratio of encapsulation gel to liquid, and others factors that would be familiar to those practiced in the art upon reading this description. The efficiency of cell production could be affected by the timing of the emerging EB, as once the EB emerges from the capsule, that particular method of prevention of EB aggregation would be gone. The effect of any subsequent EB aggregation would depend on the stage of development of the emerged EB and any limitations in size (numbers of cells) of an EB. The size of the capsule could also effect efficiency of delivery of various growth or differentiation factors or any other factors or nutrients to the cells.

   For instance larger capsules would have greater surface in which such factors could pass through, or provide a substrate onto which one could functionalize bioactive molecules   (Irvine    et al, 1998). This could also be accomplished by controlling the porosity of the encapsulation matrix.



  In one embodiment, the method of the invention enables the culture of differentiating ES cells in conventional commercially available bioreactor systems that include the use of standard bioprocess equipment (probes, filters, etc).



   The matrix of the capsule can be composed of a variety of substances.



  In one embodiment it is an agarose capsule. In another embodiment, the matrix should be biocompatible in that it should not have a detrimental effect on cell proliferation   and/differentiation    as desired. In yet another embodiment, the matrix should such that permits the passage or exposure of desired media components to the spheroid-forming cells or spheroid. This can be done by keeping certain components in the capsule and others out or by permitting the passage of certain components through the capsule matrix or by including certain components in the capsule matrix.



   The encapsulation material chosen should prevent aggregation of cells contained in different capsules. Suitable material would include but is not limited to agarose, alginate, polymers such as poly HEMMA   (Valbacka    et al (2001) and others or matrices described in Weaver et   al.    (1988) and Sefton et al (1977).



   The material selected for encapsulation should permit adequate delivery of any desired nutrients, cell culture media, growth factors, cytokines, or any other factors to promote desired spheroid-forming or ES cell growth,   proliferation, differentiation or spheroid or EB formation, e. g. , through      functionalization    of the encapsulation matrix, or by diffusion through the pores.



  The culture media can also include other factors such as low molecular weight molecules or drugs, such as retinoic acid or DMSO, as desired. Factors can also be included in the media for testing their effect on the cells. The material selected may depend on the factors that are to be delivered. Example of matrices that could be used are agarose, alginate, and polymers that support cell growth. Examples of suitable matrices are described in Weaver et al.



  (1988) and Sefton et al (1977). However, a person skilled in the art would be familiar with other suitable matrices upon reading this description.



   Examples of culturing conditions that promote spheroid-forming cell or
ES cell differentiation and spheroid or EB formation include but are not limited to conditions described in Keller et   al.    1995 or O'Shea et al. 1999 or those outlined in Tables 10 or 11 and references noted therein to enhance generation of particular cell types. The spheroid-forming cell and other cells used in the invention can be genetically modified cells. The genetic modification can be for a variety of purposes including, to enhance cell selection by conferring particular identifying characteristic to the desired cell, to generate cells for gene or cell therapy, transplantation (for instance to reduce or prevent graft versus host disease and/or rejection) diagnostic and/or screening purposes. Other applications would be apparent to those skilled in the art. 



   More particularly, the conditions under which the ES cells are cultured for differentiation will depend on the type of ES cell derived cells and tissues desired. For instance, to obtain hematopoietic stem cells one would preferably culture the ES cells under certain conditions, such as described in Wiles and
Johansson 1999; Rathjen et al. 1998; or Keller et al, 1993.

   On the other hand, if one wishes to obtain cardiomyocytes, the cells would be cultured under specific conditions, such as in the presence of specific cytokines, i. e.   TGF-     (Dickson et al. 1993), or using a selectable genetic marker such as described for cardiac myocytes in Klug et al   (1996).    In vitro, ES cells have been shown to differentiate into a variety of cell types and tissues, including beta cells, cardiomyocytes, hepatic cells, kidney cells, neuronal cells, and hematopoietic cells.

   Various cell types and culturing conditions are described in Zandstra and Nagy, 2001 for general approaches; Wang et   al,    1992 for endothelial cells ; Rohwedel et   al.,    1994, Fleishman et   al,    1998 and Mummery et al. 2002 for cardiac and muscle cells ; Bain et al, 1995, Tropepe et al, 1999 and
Reubinoff et al. 2001 for neuronal cells ; Palacios et al, 1995 and Kyba et   al.,    2002 for hematopoietic stem cells ; Choi et al, 1998 and Levenberg et al. 2002 for hematopoietic and endothelial cells. Assady et al. 2001 for insulinproducing cells. For other examples of culture conditions see Tables 10 and 11.

   Since the differentiation of the ES cells results in a complex mixture of all possible cell types an efficient enrichment protocol would be necessary to obtain a homogeneous population of specific cell types or tissues to be utilized for various purposes including the generation of surrogate cells for human therapies or for drug screen testing. This enrichment could be done by the introduction of a specific construct into ES cells, comprising a cell or tissue type specific promoter controlling the expression of a marker gene. The resulting transgenic ES cell (s) can be expanded, differentiated and selected to generate the desired cell type of interest using techniques known in the art, for instance as described in Klug et al (1996) for cardiac cells (WO 95/14079, 26 May 1995; U. S. Patent Nos. 5,733, 727 and 6,015, 671) or Soria et al (2000) or Soria et al (2001).

   The method is widely applicable to any cell type for which the tissue specific promoter is known. Table 9 provides some examples of promoters useful for the selection of specific cell lineages without limiting the number of useful promoters to that table.



   Particular cell types can be selected for and/or harvested using a variety of methods, including cell surface receptors or other labeling, tagging or monitoring or selection methods known in the art, such as expression of particular types of genes, proteins or levels of certain molecules.



   In a further embodiment, the invention provides a method for the selection of specific cell types from spheroid-forming cells comprising the steps of: i. introducing a reporter gene expressing vector into at least one spheroid-forming cell whereby a cell-type specific promoter is combined with an reporter gene, such that the reporter gene is expressed under the control of the cell-type specific promoter;

   ii. culturing the spheroid-forming cell (s), for example to expand the cells iii. differentiating spheroid-forming cell (s), for instance in accordance with the method of the present invention under controlled aggregation conditions. iv. isolating and harvesting the specific cell-type based on the reporter gene expression
In one embodiment, the reporter gene is an antibiotic resistance gene and the cell-type of interest is isolated by the addition of an appropriate antibiotic in step (iii) or (iv).



   In yet another embodiment, the method for the selection according to the reporter gene is selected from the group consisting of the Hygro mycin resistence gene (hph), the Zeocin resistence gene (Sh   ble),    the Puromycin resistance gene (pacA), and the Gentamycin of G418 resistance gene (aph).



   In yet another embodiment, in the method for the selection the reporter gene is selected from the group consisting of luciferase, green fluorescence protein, red fluorescence protein, and yellow fluorescence protein and the cells of interest are selected by fluorencent activated cell sorting (FACS). 



   In yet another embodiment, in the method for the selection the reporter gene is selected from the group consisting of   luciferase,    green fluorescence protein, red fluorescence protein, yellow fluorescence protein, or a his-, myc-, or flag-tag ligated to a   heterologous    gene, or any   heterologous    gene which when expressed is inserted into the cell surface and the cells of interest is isolated from the cultured cells by affinity purification.



   In yet another embodiment, in the method for the selection, the cell type specific promoter is selected from those listed in Table 9 for cell types listed therein.



   The initial undifferentiated spheroid-forming or ES cells used in the method can be obtained by methods known in the art. For instance one can use ES cells obtained directly from the inner cell mass of the embryo, or one can use ES cells that were cultured, i. e. expanded under conditions that preferably did not promote differentiation, such as described in Klug et   al.   



  1996.



   In another embodiment the method of the invention enables culture of differentiating spheroid-forming or ES cells in stirred 3-dimensional reactors with scalable volume.



   In another aspect the invention provides a culture bioreactor for industrial production of cells derived from spheroid-forming cells comprising: a. culturing spheroid-forming cells in accordance with the method of the invention under controlled aggregation, wherein the cells are cultured in a spheroid-forming cell suitable media under conditions that promote spheroid formation; while b. inhibiting or preventing spheroid aggregation.



   Such suitable media conditions are any conditions that promote spheroid formation, examples of which are provided herein.



   In another embodiment, the invention provides a bioreactor for generating cells from spheroid-forming cells comprising a means for controlling conditions suitable for spheroid formation, such conditions comprising a means for controlling cellular aggregation and means for maintaining said cells and generated cells in suspension, such as means for preventing cellular aggregation.



   In yet another embodiment the invention allows for measurement and control of the culture environment by enabling the use of stirred or other wellmixed bioreactors.



   As a result of being able to better control the ES cell culture environment, the method of the invention enables one to better study various conditions to obtain particular differentiated ES cells and tissue (i. e. hematopoietic cells, cardiomyocytes, nerve cells, beta cells, hepatic cells, kidney cells, any other cell types differentiated or not, known to develop from embryonic stem cells) and to better optimize these conditions. As such, the method of the invention can be used to derive a cell culture with a higher density of desired differentiated cells. The ability to do this may determine whether or not the process is economically viable and therefore is critical to the translation of these technologies from the lab scale.



   The method of the invention can also be used to identify factors (e. g., any variant, such as growth factors, differentiating factors, such as cytokines, or other conditions, such as pH, temperature, oxygen or factor concentration levels) and conditions that effect and preferably optimize spheroid-forming cell or ES cell differentiation and spheroid or EB formation. Examples of some factors, especially for studying spheroid-forming cell or ES cell differentiation to cardiomyocytes include but are not limited to fibroblast growth factors (FGFs), vascular endothelial growth factor (VEGF), cardiotrophin-1 (CT-1), leukemia inhibitory factor   (LIF), endothelin-1    (ET-1), stem cell factor (SF), opiod peptide (or supplemental DMSO), bone morphogenic protein (BMP) family members transforming growth factor beta (TGF-beta), retinoic acid (RA).

   Factors preferably used to study beta-cell differentiation include but are not limited to, glucose levels, nicotinamide, KGF (karatinocyte growth factor),
EGF (epithelial growth factor, NGF (neural growth factor) and TGF-beta, the above factors, as well as others typically used on adult hematopoietic stem cells (Zandstra et al 1997) can also be used to elicit the development of hematopoietic and endothelial cells from pluripotent cells. This method would comprise culturing the spheroid-forming cells or ES cells under conditions that control cell aggregation in the presence of the particular factor to be tested and then detecting the effect of the variant on cell proliferation and differentiation. The effect of various different concentrations or formulations of the factor can also be tested.

   Preferably a negative control is used, whereby the spheroid-forming cells or ES cells are cultured in the absence of the factor to be tested.



   The methods and products of the invention can also be used to produce cells useful for drug therapy testing, to identify targets for gene and cell therapy, and in assays for such methods.



   Spheroid-forming cells, such as ES cells and the cells and tissues derived thereform can also be used in the treatment of various cancers, leukemias, autoimmune diseases, organ failure, animal or tissue cloning, gene therapy, transgenic animals.



   The spheroid-forming cells and ES cell-derived cells and tissues obtained from the invention can be used to provide cells and tissues for transplantation. For instance, ES cells can be grown under conditions to produce hematopoietic cells. Such cells could be used in bone marrow transplants, blood transfusions or infusions. ES cell derived cardiomyocytes can be used in tissue engineering, cell/tissue transplantation, gene therapy, and drug discovery. ES cells derived skin tissue can be used for    reconstructive surgery, ie. , for burn victims, cosmetic surgery to name a few.   



   Further examples of applications for ES derived cells and tissues can be found in Rathjen et al 1998 or Marshal E. 2000.



   In yet another embodiment the invention is directed to a method to identify factors (any substance, media or other conditions, for instance oxygen levels, pH, pressure, cell density) that effect cell proliferation, differentiation and/or spheroid formation or other cellular or spheroid characteristic desired to be monitored, said method comprising culturing the cells as per the method of the invention in the presence of the factor to be tested and then monitoring the effect of the factor on cell proliferation, differentiation and/or spheroid formation or other cell or spheroid characteristic desired. Such monitoring can include comparing the effect of the factor on cells in light of a control culture. In one embodiment, such a control culture can be a negative control which does not contain the factor to be tested.

   Such factors can include factors for screening compounds that may be useful for the treatment or diagnosis of various medical conditions.



   It should be noted that although the discussion above describes the invention in terms of ES cells, the same methods and applications and products of the invention would be applicable to pluripotent cells in general or spheroid-forming cells other than ES cells.



   The present invention also includes the Spheroid-forming and
Embryonic stem cell derived cell culture obtained using the method of the invention, including the initial encapsulated, and/or high density cell culture
In another embodiment the invention provides a method to prevent aggregation between cells comprising encapsulating a cell or group of cells and thus preventing aggregation of said cells with cells not within said capsule. Further the invention also encompassed the use of encapsulating cells, as described herein to control the micro environment of a cell to be cultured. This is not limited to spheroid-forming or ES cells. Such methods can be used to proliferate such cells or to control culturing conditions. To optimize cell culture conditions and to identify factors that effect culture conditions.

   A liquid suspension comprising encapsulated cells, such as spheroid-forming cells and derived cultures there from are also encompassed within the scope of the present invention.



   The following non-limiting examples are illustrative of the present invention:
EXAMPLES
Materials and Methods
ES Cell Culture
Murine embryonic stem cells : CCE,   R1,    D3 (mentioned below) and others (Beddington and Robertson, 1989; Nagy, Rossant et   al.,    1993), were maintained at   37 C    in humidified air with 5% CO2 in Iscoves Modified
Dulbecco's Medium   (IMDM, GIBCO-BRL)    supplemented with 15%   fetal    bovine serum   (Hyclone),    50   U/ml penicillin (GIBCO-BRL), 50, ug    streptomycin  (GIBCO-BRL), 2mM L-glutamine (GIBCO-BRL), 0.1 mM 2-mercaptoethanol, and 500 pM leukemia inhibitory factor   (LIF)    (ES cell media).

   Culture flasks were prepared prior to cell seeding by coating with a solution of 0.   1%    porcine gelatin in phosphate buffered saline. All ES cells were used between passages 15-24.



  ES cell differentiation systems
ES cells were dissociated using 0.25% trypsin-EDTA (Sigma) and cultured in ES cell medium without   LIF    to induce differentiation. Liquid suspension cultures (LSC) were prepared by aliquoting 10 ml of cell solution at the indicated ES cell density into 10 cm petri-dishes (Fisher). For single ES cell analysis (single cell liquid suspension culture scLSC), a single ES cell was placed into each well of a 96 well plate (Nunc) coated with 10% pluronic (F-127, Sigma) solution to prevent cell attachment. This was achieved by preparing a suspension of 20 ES cells/ml and aliquoting   50      ZL    into each well.



  Each well was then visually inspected for the actual number of deposited cells. Empty wells or wells with more than one cell were marked and not included in the analysis. Methylcellulose cultures (MC) were prepared by aliquoting 1 ml of cell suspension with   1% methylcellulose    (M3220, Stem Cell
Technologies) at the indicated ES cell density in 35mm Greiner dishes (Greiner Labortechnik). Hanging drop cultures (HD) were prepared using 10   ptL droplets,    each containing the number of ES cells desired to initiate the EB (as indicated). ES cells were allowed to aggregate in hanging drops for two days before transfer to liquid suspension culture.



   Stirred cultures were performed in spinner cultures   (Bellco    Glass) ranging from 50 to   250ml    of media/vessel. ES cells were added to the media directly (stirred cultures without encapsulation: SC) or encapsulated (ESC) as indicated. A paddle-style impeller was used to agitate the media at 40-60
RPM.



   EBs were enumerated by visual inspection. EBs were dissociated by incubation (1 min,   37 C)    in 0.25% trypsin-EDTA (Sigma), or incubation (30 min,   37 C)    in 0.25%   collagenase    (Sigma) in PBS supplemented with 20% fetal bovine serum for post day 7 EBs. Mechanical shearing was also used to help dissociate cells into a single cell suspension by twice passaging the cell mixture through a 21-gauge needle and syringe (Becton Dickinson).



     Individual cells were    then counted using a hemocytometer and analysed by either myeloid-erythroid colony-forming cell (CFC) assay (Zandstra et   al.   



  1997) or flow cytometry (Zandstra et al. 1997)
Gel Microdrop (GMD) Encapsulation of Individual ES Cells
Individual CCE ES cells were suspended in Hanks Buffered Saline
Solution (HBSS) at a concentration of   107 cells/ml    and added to a 3% agarose solution for a final cell concentration of   2x106 cells/ml.    The agarose solution was dispensed into mineral oil   (nonsolvent)    at   37 C    and subjected to impeller blending using the One Cell System (Ryan et   al.,    1995) to create agarose microdrops containing individual ES cells. The GMDs were washed twice with
HBSS to remove the mineral oil before resuspension in ES cell media without
LIF for differentiation culture.



     Ge/Microdrop (GMD) Encapsulation of Multiple ES Cells   
Three different methods were used to encapsulate multiple ES cells.



   (1) ES cells were gently dissociated from adhered culture using trypsin, creating multi-ES cell clumps or aggregates. Clumps were subsequently encapsulated as described for individual ES cells.



   (2) ES cells were dissociated to a single cell suspension and cultured at high cell density   ( > 105 cells/ml)    in unagitated petri dishes for 12-24 hours to induce cell clump formation via controlled aggregation The average number of
ES cells per clump was controlled by input cell density and by time, and was determined by dividing the input number of ES cells by the number of clumps counted ES cell clumps were then encapsulated as described.



   (3) Individual ES cells were mixed with agarose at a concentration of 1  5x107 cells/ml    and encapsulated as described. Capsules formed in this way were essentially clumps of individual ES cell held together by agarose matrix.



  Capsules themselves were considered ES cell clumps and again encapsulated as described. 



  Hematopoietic Colony Forming Cell Assay
Cells were plated into methylcellulose medium (M3434, Stem Cell
Technologies) containing 50   ng/ml    mrSCF, 10 ng/ml mrlL-3, 10 ng/ml hrlL-6, 3   U/ml      hEpo,    15% fetal calf serum, 0.1 mM 2-mercaptoethanol, and 2mM Lglutamin. Hematopoietic colonies were scored by morphology after 6-7 days.



  Generation of cardiac myocytes from ES cells
CCE and R1 embryonic stem cell were transfected with the MHCneor/PGK-hygror DNA as previously described (Klug et al 1996, WO 95/14079,26 May 1995; U. S. Patent Nos. 5,733, 727 and 6,015, 671). Briefly, 1   lig    of DNA and 25   jig    of salmon testes DNA were mixed in a total volume of 800   1ll    of cells suspended at 4 x 106   cells/mL    on ice. Cells were electroporated (180 volts, 80 mF), incubated on ice for 15 minutes, and plated in 100 mm corning dishes in ES media containing   500pM    LIF. Cells containing the plasmid were selected by culture in ES medium with LIF and   200      ug/mL    hygromycin B for 7 days with daily medium exchange.

   Test ES cells were then encapsulated using the multiple ES cell encapsulation technique as described above in the third technique. After 8-10 days of culture in stirred suspension bioreactors (Bellco spinner flasks or Applikon 1L cell culture bioreactors) in ES cell media without LIF, the G418 was added to the medium to select for the cardiac cells. The composition of the selected cells was analyzed by MF-20 staining using flow cytometry. 5 x 105-106 cells to be stained for MF-20 were washed with PBS and fixed with   100      gi of lntraPrepTM      Permeabilization    Reagent 1 (Immunotech, Westbrook, ME) for 15 minutes at room temperature.

   After washing once with 1   ml    PBS, the cells were permeabilized for 5 minutes with   IntraPrepTM Permeabilization    Reagent 2 and incubated with a 1: 10 dilution of the mouse anti-sarcomeric myosin heavy chain (MF-20) antibody (Transduction Lab, MA) for 15 minutes at room temperature. After washing once with PBS, secondary   FITC-conjugated    goat anti-mouse IgG antibody was added with 1: 100 dilution and the cells were incubated for 15 min at room temperature. In addition, some of the dispersed cells were used for determining the viability of the cells immediately after the procedure of enzymatic dissociation. Test cell populations were washed in ice-cold Hanks   hepes-buffered    salt solution containing 2% FCS (HF) and resuspended at 106 cells/100   FL    in HF.

   Finally, the cells were analyzed on a flow cytometer (XL, Beckman-Coulter, Miami, FL) using the EXpoADCXL4 software (Beckman-Coulter). Positive staining was defined as the emission of a level of fluorescence that exceeded levels obtained by  > 99.9% of cells from the control population stained with only the secondary antibody. Cardiac development was also determined by electron microscopy.



  Generation of Hematopoietic and Endothelial Cells
Hematopoietic and endothelial cells were generated in through ES cell diffierentiation in ES cell media without   LIF    under the conditions described in
Dang et al. 2002. Input ES cells were encapsulated using the multiple ES cell encapsulation technique as described above in the third technique.



  Flow Cytometry for analysis of endothelial and hematopoietic cells
Cells were washed in ice cold Hanks hepes-buffered salt solution containing 2% FCS (HF) and resuspended for 10 min at   4 C    in HF containing a-mouse CD16/CD32 monoclonal antibody (PharMingen) at   1, ug/1001lL    to block non-specific binding. Blocked cells were then incubated at   107      cells/ml    for 40 min at   4 C    with FITC anti-mouse CD34 (PharMingen), PE anti-mouse   Flk1    (PharMingen), and anti-mouse E-Cadherin (Sigma). Anti-mouse E
Cadherin incubation was followed by two washes in ice cold HF and then incubated for 40 min at   4 C    with anti-mouse   FITC-conjugated    IgG (Sigma).



  Stained cells were washed twice in ice cold HF, and 1   Rg/ml    7-aminoactinomycin D (7AAD, Molecular Probes) was added to the final wash. The cells were analyzed on Coulter Epics XL using 4-Colour Expo 32 software for analysis. Positive staining was defined as the emission of a level of fluorescence that exceeded levels obtained by  > 99.9% of the cells from the same starting population stained with a matched   fluorochrome-labelled    irrelevant isotype control antibody. 



  Measurement of Media composition
Oxygen tension, in addition to other parameters, was measured by withdrawing   3ml    of culture media using a gastight syringe (Hamilton), and analyzing using a BioProfile 200   blood/gas    analyzer (Nova Biomedical).



     Neurosphere formation   
R1 murine embryonic stem cells were dissociated into a single cell suspension and transferred at   104    ES cells/ml to media comprised of
Dulbecco's Modified Eagle Medium (DMEM, GIBCO-BRL) supplemented with 20% Knockout serum replacement (Sigma), 50   U/ml    penicillin (GIBCO-BRL), 50   pg    streptomycin (GIBCO-BRL), 2mM L-glutamine   (GIBCO-BRL),    and 0.1 mM 2-mercaptoethanol.



  Example 1-Effects of Cell-Cell Adhesion and EB Aggregation on ES cell culture
To assist in the development of a new bioprocess, the properties of ES cells and EBs that might affect bioprocess efficiency, net cell expansion, and cell fate decisions were investigated. EB formation, growth and development between different culture techniques were compared. Once an understanding of these basic parameters was obtained, guidelines to the design of efficient, scalable bioprocesses were developed.



  (a) Some ES cell lines can form EBs from individual cells.



   Preliminary studies focused on establishing the in vitro developmental potential of individual ES cells. In single cell liquid suspension (scLSC) and methylcellulose culture (MC), EB efficiency (number of EBs divided by input number of ES cells) have been reported anywhere between 2-15%. Because these values were so low, it was uncertain as to whether an individual ES cell could form an EB, or if EB formation occurred only when ES cells aggregated by chance (or by design, as in the hanging drop method). Therefore the first constraint investigated was the number of ES cells required to form an EB.



   Individual murine CCE ES cells were placed by limiting dilution into separate wells of a 96 well plate and differentiated in liquid suspension culture  (ES media without   LIF).    One hour after plating, each well was visually examined to verify the number of cells plated. After seven days of differentiation, the wells were examined for cell aggregates. The cell aggregates were identified as EBs based on their expression of Flk-1 and
CD34, and their ability to form hematopoietic colonies (data not shown). By dividing the total number of EBs per 96 well plate by the number of input ES cells, we determined that individual ES cells were able to form EBs with an overall efficiency of at least   429%    (Table 1).

   In a separate set of experiments, it was determined that   789%    of the starting cell population were undifferentiated ES cells based on E-cadherin and SSEA-1 expression   (data not shown, (Zandstra, et al. 2000) ). Viability, based on exclusion of    dead cell dye 7AAD, of the ES cell population after trypsinization was determined to be   8711%.    This allowed recalculation of the EB formation efficiency from individual, undifferentiated, and viable ES cells to be approximately 60%. These results clearly demonstrated that a majority of individual murine CCE ES cells have the capacity to form an EB, and that these cells could form EBs at a very high efficiency. However, the results also raise the issue of why such high efficiencies were not regularly achieved in standard differentiation cultures.



   The present inventors have shown that murine CCE ES cells have demonstrated the ability to form EBs from single ES cells. In other cell types or under other conditions contact between multiple ES cells to enable EB formation may be preferred. For example, the present study repeated with single murine R1 and D3 ES cells did not form single ES cell derived EBs under the conditions used Instead, aggregation with approximately 5 or more
ES cells was required for survival, proliferation and differentiation (see below).



  Similarly, it has been reported [Thomson JA, Odorico JS. 2000] that current human ES cell lines form EBs only from multi-ES cell aggregates or clumps.



  Regardless, the following aggregation studies reported with CCE ES cells are still illustrative of the extent of EB aggregation and the problem it poses. As such controlling cellular aggregation during expansion and differentiation of said cells is an important factor. 



  (b) EBs aggregate, reducing EB formation efficiency
Further investigation revealed the sensitivity of EB efficiency to initial
ES cell concentration and time (Figure 3). Various initial CCE ES cell concentrations were differentiated in liquid suspension and methylcellulose cultures, and the number of cell aggregates (EBs) per dish was recorded at regular time intervals. In liquid suspension culture, EB formation efficiency increased as ES cell concentration decreased (Figure 3A). Over time, EB formation efficiency decreased for all cell concentrations. However the extent of this decrease was most dramatic in cultures initiated with   105    ES cells/ml, and almost nonexistent at 102 ES   cells/ml.

   In methycellulose cultures    (Figure 3B) initiated with   103    ES cells/ml or less, EB formation efficiency remained constant at approximately 40% over the duration of the experiment. EB formation efficiency gradually decreased in methylcellulose cultures initiated with   104    or more ES cells/ml, however to a lesser extent than their liquid suspension counterparts.



   Based on their similarity, the model of the present invention for EB formation from single ES   cells (429%)    was reconciled with EB efficiency in methylcellulose culture initiated with   103    ES cells/ml or less (40%   6%)    and liquid suspension culture initiated with   102    ES cells/ml or less   (345%).   



  However, an explanation was still required for the discrepancy with the results obtained at higher cell concentrations. It was recognized that the decrease in
EB efficiency over time indicated that the process of EB formation itself was unaffected; rather, another process occurring post-EB formation must be reducing the number of EBs present. Observing the cultures, it was apparent that EB aggregation was the cause of inefficiency. In this process, separate
EBs aggregated and merged into a single, larger EB.



   This phenomenon accounts for decreased EB efficiency with increased cell concentration since likelihood of EB collision and subsequent aggregation would also be greater. Furthermore, aggregation explains the higher EB efficiency in methylcellulose cultures compared to liquid suspension culture: Semi-solid   methycellulose    media impaired cell movement, thereby decreasing likelihood of EB collision and aggregation. 



     (c)    Aggregation reduces net cell expansion
The results demonstrate that CCE ES cells form EBs with an efficiency of   429%,    a value independent of culture method and initial ES cell concentration, and that EB aggregation reduced the apparent efficiency over time. To determine what effect EB aggregation might have on the overall production of cells, the growth rate of various initial ES cell aggregate sizes (1,100, and 1000 ES cells/EB) formed using the single cell liquid suspension culture (scLSC) and hanging drop techniques were compared (Figure 4, Table 2). Regardless of initial aggregate size, all EBs contained a similar number of cells by day 12. Since final EB size is independent of EB aggregation, EB aggregation reduces overall expansion of cells.



  (d) Cell-cell adhesion molecules are responsible for EB aggregation
The process of EB aggregation was studied using the simples scenario: aggregation of two EBs. To visualize cell mixing, one EB generated from wildtyp ES cells was aggregated with one EB generated from ES cells    constitutively expressing GFP (Hadjantonakis et al 1998) ). The extent of    aggregation was quantitatively determined by the"degree of aggregation"    (DOA) -the ratio of the diameter of the interface to the overall length of the    two EB system (Figure 5). Regardless of initial aggregate size (50,100, 400
ES cells/EB, data not shown), complete fusion (DOA  >  90%) was achieved after approximately 16 hours and complete cell mixing (homogenous fluorescence intensity) after 48 hours (Figure 5).



   The mechanism of EB aggregation was investigated using various loss of function treatments (Figure 6). Blocking surface cell adhesion molecules with blocking peptide inhibited EB aggregation. E-cadherin blocking was particularly efficient, also noted by Larue et   al.    (1996) who observed that homozygous E-cadherin null ES cells were unable to aggregate as clumps.



  EBs treated with cytochalasin D also were unable to aggregate. Cytochalasin
D inhibits actin-dependent processes including cell migration and cell division.



  To be sure that impaired cell division was not responsible for this effect, another group was treated with Mitomycin C that inhibited mitosis by preventing DNA synthesis. These EBs aggregated at the same rate as the control (no treatment). Based on these results, a two-step mechanism for EB aggregation was proposed: First, neighboring EBs collide and   homophilic    Ecadherin molecules adhere, holding the EBs together. Cells then migrate and remodel as the EBs fuse into a single spheroid.



   Preventing cell adhesion versus cell migration between neighboring
EBs was the logical strategy for preventing EB aggregation as treatments that could affect cell growth or differentiation (i. e. cytochalasin D) would be undesirable for most applications. E-cadherin, the prominent cell adhesion molecule in this process, is a   Ca2+-dependent homophylic cell-cell    adhesion molecule expressed by ES cells. As ES cells differentiate, E-cadherin expression is downregulated (Zandstra et al. 2000). Using flow cytometry, Ecadherin expression was tracked in differentiating EBs over time (Figure 7. A).



  Expression decreased as cells differentiated-from almost 100% on day 0 (ES cells) to approximately 25% by day 5 of differentiation. As expected, Ecadherin expression correlated with rate of EB aggregation: EBs differentiated for four days or fewer aggregated within 16 hours, while day 6 EBs did not aggregate (Figure7. B). Similarly, while ES cells placed into stirred culture quickly aggregated into large cell clumps that did not grow or differentiate properly, EBs grown in static culture for a minimum of four days could be transferred to stirred culture with little aggregation. These EBs continued to grow and differentiate normally. Therefore, the strategy adopted for controlling cell adhesion need only be implemented while cell adhesion molecule expression remains high. For murine ES cells, this time period is the first four days of differentiation.



   These results suggest that novel approaches to control the aggregation of undifferentiated ES cells and their derivatives may allow them to be cultured in high cell density, stirred suspension bioreactors.



  Example 2-Preventing ES Cell-Cell Adhesion and EB Aggregation
Results in Efficient Generation of Embryonic Stem Cell Derived
Hematopoietic and endothelial cells
The overall schematic of one embodiment of the bioprocess of the invention for the scalable culture of ES cell derived tissues is shown in Figure 2. First, undifferentiated ES cells are expanded under appropriate conditions.



  ES cells are then cultured in a bioreactor conditions that control aggregation and promote cell proliferation and differentiation. More specifically, aggregation of ES cells to induce EB formation is permitted and aggregation between cells within an EB is permitted. However aggregation between ES cells beyond that required to induce EB formation is prevented, and aggregation between separate EBs is prevented.



   After an appropriate period of culture, under defined conditions (including growth factor and medium formulation, oxygen tension, pH, medium supplementation rates), differentiated cells are selected for and used in clinical or pharmaceutical applications. In one aspect, the present invention is primarily concerned the scalable generation of ES cell derived cells and tissues.



  (a) Culture   of EB Under Non-Aggregating Conditions   
Genetically modified ES cells    E-cadherin¯/¯ ES cells    are defective in cell adhesion, unable to grow as clumps or aggregates of cells as they typically do (Larue, Antos et   al.    1996).



  Antibodies to block cell adhesion molecules
As reported in the present study of EB aggregation, treatment of EBs with E-cadherin blocking peptides inhibits EB aggregation. Blocking peptides effectively nullifies the function of E-cadherin, as reported by   Burdsal    et al.



  (1993). Blocking E-cadherin prevents EBs from adhering with neighboring
EBs, thus circumventing subsequent aggregation.



  Reducing inorganic salt concentration or inhibition of ion channels
Cell adhesion can be inhibited by substantially reducing inorganic salt concentration (Boraston et   al.    1992). For example, cell adhesion modulated by calcium-dependent adhesion molecules such as cadherins depends upon calcium influx from both   extracellular calcium    through calcium channels and release from internal calcium stores. Calcium channel inhibitor   LaC13    or thapsigargin was shown to inhibit cell-cell attachment (Ko, Arora et al. 2001). 



     (b)    Encapsulation of ES cells
Preventing EB aggregation was the guiding design principle for new bioprocesses capable of efficient and scalable differentiation of ES cells.



  While it was suspected that E-cadherin was the main molecule involved in cell aggregation, a method was developed to preclude interactions between all cell-cell adhesion molecules on the EB surface by physically blocking their association. In this method, ES cells are encapsulated within a biodegradable matrix and allowed to grow and differentiate in liquid culture, such that each cell-containing capsule gives rise to a single EB. The surrounding matrix prevents separate EBs from contacting one another, thereby preventing aggregation. Formation of EBs in this novel way permits high cell density culture in a homogeneous, controllable, and scalable bioreactor system. The bioprocess of the present invention is termed: encapsulated stirred culture (ESC). The first step in this method involves the encapsulation of individual or multiple ES cells.

   Uniform or"synchronized"EB sizes could be achieved by encapsulating similar numbers of ES cells per capsule. The matrix could be agarose, alginate, or another type of natural or artificial matrix (e. g. polymer) that does not interfere with the growth and differentiation of EBs. Depending upon the matrix used, cells might degrade the matrix as they grow (as is the case with agarose), or they might burst the capsule as cell growth places increasing pressure on the capsule (as is the case with alginate), or they can be be manually released from their capsules by adding degradation enzymes (such as agarase addition to agarose), or changing the culture conditions (such as depleting dissolved calcium ions in the case with alginate).



  Therefore, the size and composition of the capsule can determine the amount of time the EB remains encapsulated. For example, agarose is a simple polysaccharide readily degraded by the cells as they grow. Therefore, the diameter of the agarose capsules determines the amount of time the EB remains encapsulated (Figure 8. A, Table 3). Capsules are designed to retain the EB until cell differentiation results in downregulation of E-cadherin (or other cell adhesion molecule) expression. After this time, EBs that emerge from the capsules will not aggregate. For differentiating EBs from murine ES cells, E-cadherin is sharply down regulated after 4 days (Figure 7. A). It was determined that   70-100, um    agarose capsule diameters provide adequate protection for this period of time, and such capsules were specifically designed (Figure 8. B).

   However, it should be understood that the invention is not intended to be limited to such a capsule size as a person skilled in the art would appreciate that other sizes would work.



     (c)    Encapsulation enables use of stirred culture
ES cells placed directly into stirred culture quickly aggregate into large cell clumps that did not proliferate or differentiate into any of the cell types assayed for (data not shown). Encapsulating ES cells prior to their introduction to stirred culture enabled efficient EB formation and prevented their aggregation. EB efficiency between liquid suspension culture (LSC) [non-stirred, non-encapsulated], stirred culture (SC), and encapsulated stirred culture (ESC) was compared at   103    and   104    input ES cells/ml after 7 days of differentiation culture (Table 4). Whereas EB efficiency in LSC declined as
ES cell density increased, EB efficiency remained high in ESC.



   Fold expansion of total cells in liquid culture (LSC), stirred culture (SC), and encapsulated stirred culture (ESC) is illustrated in Table 5 Cultures were initiated with   104    R1 ES cells/ml and analyzed after 7 days of differentiation.



  Table 5 shows results of fold expansion, after adjusting for cell loss during encapsulation procedure. Yield of encapsulated ES cells was approximately 25% of input. Cells were lost during the various transferring stages between centrifuge tubes, or retained in the mineral oil phase.



   In stirred culture, EB efficiency was significantly improved with encapsulation than without. Encapsulated ES cells were able to form EBs that remained shielded from one another, thus maintaining a high EB efficiency by preventing EB aggregation. EBs emerged from the capsules after approximately four days-coinciding with down regulation of E-cadherin as previously described. Emerged EBs did not aggregate.



   The ability to use stirred or other well-mixed systems ensure that bulk media conditions are homogenous, thus facilitating measurement and control of the system. These culture systems can also accommodate increase demand in cell generation by simply increasing the volume of the tank.



  (d) Bioreactor does not significantly affect cell growth or differentiation
Hematopoietic development within EBs has been extensively studied and well characterized (Keller 1995). Hematopoietic development was therefore selected as the measure by which ES cell differentiation in different culture systems was compared. ES cells become blood cells through a series of developmental steps that can be tracked by cell surface marker expression (Table 6). Murine ES cells differentiate into   Flk-1    expressing mesoderm on day 4 that subsequently give rise to CD34 and CD45 expressing hematopoietic progenitor cells. Hematopoietic progenitors can also be assayed for using the hematopoietic colony forming cell (CFC) assay.



   Ideally, the encapsulating matrix physically prevents EB aggregation but does not impede transport of nutrients and metabolic products to and from the cells, nor influence differentiation or growth of EBs in any manner. To demonstrate this, ES cells were encapsulated in agarose and differentiated in static liquid culture. Cell proliferation and differentiation in this system was compared to standard ES cell differentiation systems: liquid suspension culture, hanging drop, and methylcellulose cultures. Therefore agarose encapsulation of ES cells did not affect EB cell growth in any significant way.



   Cell proliferation and differentiation was also compared between liquid culture (LSC) and the bioprocess enabled by the invention: encapsulated stirred culture (ESC) (Table 7). ESC was able to generate the hematopoietic cell types following the same developmental kinetics as LSC. Cell proliferation and hematopoietic cell frequencies were comparable but not necessarily equivalent between the two systems.



  EXAMPLE 3-Use of Bioprocess to Generate Differentiated Cell Types
Generation of various differentiated cell populations was performed using encapsulated ES cells grown in stirred suspension bioreactors compared to LSC. a) Generation of hematopoietic cells from ES cells.



   Mentioned previously, early mesoderm development was tracked by   Flk-1 cell    surface marker expression on day 4, while hematopoietic progenitors were detected   phenotypically    by co-expression of CD34 and
CD45 cell surface markers, or functionally via methylcellulose colony assays.



  Using our novel invention, various types of hematopoietic progenitor cells were detected, from multipotent myeloid cells (CFC-Mix), to further restricted myeloid and erythroid cells (CFC-Ep,   CFC-Ed,    and CFC-GM) (Table 7). ES cells were differentiated in ES cell media in the absence of LIF. Because the culture was performed in a stirred bioreactor, it is feasibly scalable to any desired volume, and applicable to close   microenvironmental    control   b)    Generation of endothelial cells from ES cells.



   Using the same approach as described above (ES cell differentiated in
ES cell media in the absence of   LIF)    in section a), endothelial cells were detected by coexpression of multiple endothelial cell markers:   Flk-1,    CD34, and CD31. A gate was drawn around   Flk-1    and/or CD34 expressing cells, enabling exclusive analysis of gated cells for expression of CD31 (Figure 9).



  Because the culture was performed in a stirred bioreactor, it is feasibly scalable to any desired volume, and applicable to close   microenvironmental    control. c) Generation of other cell types from ES cells
Other researchers have demonstrated that other cell lineages can also be generated from ES cells in vitro, including hepatocytes,   0-cells,    neurons, glial cells, kidney cells, muscle cells, endothelial cells, hematopoietic cells    [i. e. , Wang et al, 1992 for endothelial cells ; Rohwedel et al., 1994, for cardiac    and muscle cells 1994; Bain et al, 1995 for neuronal cells ; Palacios et al, 1995 for hematopoietic stem cells ; Choi et al, 1998 for hematopoietic and endothelial cells ; Fleishman et   al,    1998 for cardiac cells ;

   Tropepe et al, 1999 for neural cells], and other cell types known to be derived from ES cells in the art. These cell types can be generated in stirred or mixed bioreactor cultures using media and general environment conditions as described in the prior art, only when aggregation is controlled, such as is possible using the bioprocess of this invention. This would be so as it was demonstrated that the bioprocess   of the invention, e. g. , encapsulation, does not adversely affect ES cell    differentiation. The fact that this technology of the invention allows for the normal differentiation of ES cells, while facilitating the large-scale production of ES cell-derivatives makes it useful for a variety of cellular therapy-related applications.



  Example 4-The system facilitates measurement and control of culture environment.



   Current ES cell differentiation processes are not amenable to measurement and control because the static and batch-style nature of these systems result in spatial and temporal gradients. In well-mixed (dynamic) systems, the bulk media conditions are the same everywhere at any given time. That is, any point measurement made within the system would be reflective of any other point. In this way, one can easily and accurately measure bulk conditions and also introduce control elements.



   Using the bioreactor system of Example 2 above, EB development in normoxic ( > 160 mmHg   02    tension) and hypoxic (30-40 mmHg 02 tension) conditions. was compared. Oxygen tension was measured offline using a blood gas analyzer and gas mixture in the incubator headspace was adjusting accordingly. Hematopoietic cell frequency, determined by CD34 and CD45 expression and hematopoietic CFC assay, was higher for EBs generated in hypoxic conditions (Table 8), indicating the positive effect that the ability to control and manipulate the microenvironment, such as allowed by this invention will have on the generation of cells and tissue from pluripotent cells.



   The tendency for ES cells and early EBs to aggregate prevents the use of well-mixed bioreactor systems as well as higher cell density cultures.



  Therefore current differentiation systems are limited to static and batch-style conditions. The bioprocess of the invention enables ES cells to efficiently form EBs in well-mixed culture by controlling aggregation. As mentioned, the use of well-mixed systems automatically permits bulk media measurements and implementation of control strategies. 



  Example 5-The Generation of Embryonic Stem Cell Derived Cardiac cells usina ES cells Transfected with selectable markers
To demonstrate the improved ability to generate large numbers of clinically relevant cell types, the bioreactor culture of encapsulated ES cells was used to generate and select for cardiac myocytes. As for the above hematopoietic and   endothelial development, flow    cytometric analysis was employed to quantitatively evaluate the percentage of ES cell-derived cardiomyocytes generated during the course of differentiation in stirred bioreactors. In this case, ES cells transfected with the selection cassette as outlined above and described in detail in   (Klug    et al 1996, WO   95/14079,    26
May 1995; U. S.

   Patent Nos. 5,733, 727 and 6,015, 671) were cultured for 9 days in 500 mL stirred suspension bioreactors, prior to the addition of G418 to the media. At given time points the EBs were dissociated, the procedure previously described, resulting in a single cell suspension to be stained with a monoclonal antibody specific for sarcromeric myosin heavy chain (MF-20).



  Figure 10 Panel A shows kinetic changes of the percentage of the MF-20 positive cells during the course of differentiation and selection with G418 of transgene-containing ES cells cultured in stirred suspension bioreactors.



  Figure 10 panel B shows morphologic and structural analysis of bioreactorgrown ES cell-derived cardiomyocytes. The cells exhibit a mature sarcomeric organization and Z-banding (arrow). Figure 10 panel C shows a cross section of a"cardic body" (a spheroid of ES cell derived cardiac cells) generated in bioreactors.



   Results showed that only a trace amount of MF-20 staining-positive cells was present in the EBs (0.7%) on day 0 after differentiation, however, the percentage of cells displaying positive staining with MF-20 rose gradually as a function of time after selection by G418, and reached over 70% at day 20 after differentiation. In contrast, the expression level of Oct-4, which is expressed in embryonic stem cells and is one of the first transcription factors differentially regulated during mouse development   (Nichols    and Zevnik, 1998) decreased significantly during the time course of differentiation, from 68.5% at day 0 to undetectable levels (at day 20 after differentiation (data not shown). 



  The observation indicated that at the late state of differentiation and G418 selection, Oct4 positive ES cells were eliminated from the culture system. It is noteworthy that by day 15, the bioreactors typically contained between 2.5 x   107 to    1   x      108    viable cardiac myocytes (as assessed by MF20 expression).



  These observations were consistent in three independent experiments. In some experiments chemical factors such as retinoic acid were added to the media at specific time points to enhance cardiac differentiation and reduce the presence of undifferentiated ES cells (data not shown). The results indicate that differentiation of ES cells with the   G418-selection    system in spinner flask suspension culture system is capable of producing large quantity of cardiomyocytes.



  Example 6: Generation of chimeric spheroids containing predetermined types of cells.



   To show that the encapsulation system was capable of controlling the types of cells present in the spheroids at input, two populations of R1 ES cells, each labeled with a specific flurochrome (green fluorescent protein,
GFP) and (cyan) were encapsulated (see Hadjantonakis et   al.    1998 and
Hadjantonakis AK, Nagy 2001 for descriptions of the cells lines).



  Materials and Methods
Individual R1 ES cells expressing the GFP protein by a constitutively active promoter (Hadjantonakis et al 1998) and individual trophobast stem cells labelled with cyan as previously described (Tanaka et al. 1998) were suspended in Hanks Buffered Saline Solution (HBSS) at a ratio of 1 ES cell to 1 TS cell at a total cell concentration of 2 x   107 cells/ml    and added to a 3% agarose solution for a final cell concentration of   4x106 cells/ml.    The agarose solution was dispensed into mineral oil   (nonsolvent)    at   37 C    and subjected to impeller blending using the One Cell System (Ryan et   al.,    1995).

   Micordrops containing a single ES cell and a single TS cell were identified and isolated by their side scatter characteristics, and by their GFP and cyan fluorescence using a FACSVantage Flow Cytometer (Becton Dickinson). Isolated microdrops were put sorted into ES cell media without   LIF    for analysis of their growth characteristics. 



  Example
To show that the encapsulation system was capable of controlling the types of cells present in the spheroids at input, ES cells and TS cells, each labelled with a specific   flurochrome    (green fluorescent protein, GFP) and cyan were encapsulated as described. Using flow cytometry, the double positive   population, (i. e. , the microcapsules contained a single GFP cell and a single    cyan ^ labelled cells) were sorted into 96 well plates. Microscope based tracking of the cells confirmed that the two cell types survived and proliferated and formed spheroids containing two distinct sources of cells (chimeric spheroids).

   This technology provides the capacity to combine, in spheroids, two or more types of cells, including pluripotent cells and differentiated cells, for the end goal of manipulating the differentiation of the pluripotent cells into specific types of tissue using cell specific signals.



  Example 7: Formation of ES cell derived neurospheres
ES cells were differentiated in serum-free conditions, yielding spheroids that did not stain for mesoderm markers. ES cells cultured in chemically defined serum-free, feeder layer-free, low-density culture conditions readily acquire a neural identity (Tropepe et al. 1999). Under these conditions, ES cells transition to a spheroid-forming primitive neural stem cell population capable of self-renewal and neural and non-neural potentialanalogous to the neurosphere forming neural stem cells reported by Bjornson et al. (1999).



  SUMMARY
While the present invention has been described with reference to what are presently considered to be the preferred examples, it is to be understood that the invention is not limited to the disclosed examples. To the contrary, the invention is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.



  For instance, although ES cells were used in the Examples, the invention can be applied to other types of spheroid-forming or pluripotent cell types, such as embryonic germ cells and early primitive ectoderm-like cells or other type of stem cells such as neuronal stem cells or cells that can differentiate into other cell types and form spheroids in vitro during the differentiation process.



  Controlling aggregation during any type of cell expansion can also be beneficial in improving expansion efficiency of said cell type.



   All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. 



   TABLE 1
EB formation efficiency from CCE muES cells. A single ES cell was deposited into each well of a 96 well plate. Wells were visually inspected for
EBs after 7 days. EB formation efficiency was determined by the ratio of EBs per 96 well plate, to the number of input ES cells per 96 well plate.



  Percentage of viable ES cells in the culture was determined by E-cadherin expression and exclusion of 7AAD dead cell stain.
EMI45.1     





  Experiment <SEP> Input <SEP> # <SEP> # <SEP> of <SEP> Actual <SEP> EB <SEP> % <SEP> E-Cad <SEP> Viability <SEP> % <SEP> Viable <SEP> Calculated
<tb> Number <SEP> of <SEP> ES <SEP> EBs <SEP> Formation <SEP> Positive <SEP> Cells <SEP> ES <SEP> Cells <SEP> EB
<tb> Cells <SEP> Efficiency <SEP> Formation
<tb> Efficiency
<tb> 1 <SEP> 86 <SEP> 38 <SEP> 44.2% <SEP> 68.5% <SEP> 90% <SEP> 61.7%
<tb> 2 <SEP> 97 <SEP> 33 <SEP> 34.0% <SEP> 85% <SEP> 75% <SEP> 63.8%
<tb> 3 <SEP> 68 <SEP> 37 <SEP> 54.4% <SEP> 80% <SEP> 96% <SEP> 76.8%
<tb> 4 <SEP> 70 <SEP> 26 <SEP> 37. <SEP> 1%
<tb> Average <SEP> 80.3 <SEP> 33.5 <SEP> 42% <SEP> 78% <SEP> 87% <SEP> 68% <SEP> 63%
<tb> Standard <SEP> 13.8 <SEP> 5.4 <SEP> 9% <SEP> 9% <SEP> 11 <SEP> % <SEP> 14% <SEP> 17%
<tb> Deviation
<tb>  
TABLE 2
Total cell expansion from EBs initiated with 1,100, or 1000 cells/EB.



  Cell expansion improved significantly as fewer ES cells were used to initiate
EB formation on day 0.
EMI46.1     





  Initial <SEP> Cells/EB <SEP> (day <SEP> 0) <SEP> Maximum <SEP> Cells/EB <SEP> Expansion
<tb> 1 <SEP> 17300 <SEP> 9900 <SEP> 17300
<tb> 100 <SEP> 35100 <SEP> 11000 <SEP> 351
<tb> 1000 <SEP> 35000 <SEP> 3000 <SEP> 35
<tb> 
TABLE 3
Effects of different encapsulation protocols on capsule diameter. 3 different parameters: impeller speed (revolutions per minute, RPM), time (minutes), and ambient temperature, are used to control capsule diameter.
EMI46.2     





  35Nm <SEP> 50m <SEP> 85Nm
<tb> Protocol <SEP> step
<tb> 1 <SEP> 2100 <SEP> RPM <SEP> 1500 <SEP> RPM <SEP> 1200 <SEP> RPM
<tb> 1 <SEP> minute <SEP> 1 <SEP> minute <SEP> 1 <SEP> minute
<tb> Room <SEP> temp <SEP> Room <SEP> temp <SEP> Room <SEP> temp
<tb> 2 <SEP> 2100 <SEP> RPM <SEP> 1500 <SEP> RPM <SEP> 1200 <SEP> RPM
<tb> 1 <SEP> minute <SEP> 1 <SEP> minute <SEP> 1 <SEP> minute
<tb> Ice <SEP> bath <SEP> Ice <SEP> bath <SEP> Ice <SEP> bath
<tb> 3 <SEP> 1100 <SEP> RPM <SEP> 1100 <SEP> RPM <SEP> 700 <SEP> RPM
<tb> 10 <SEP> minutes <SEP> 5 <SEP> minutes <SEP> 5 <SEP> minutes
<tb> Ice <SEP> bath <SEP> Ice <SEP> bath <SEP> Ice <SEP> bath
<tb>  
TABLE 4
EB Efficiency in liquid culture (LSC), stirred culture (SC), and encapsulated stirred culture (ESC).

   Cultures were initiated with   103    or   104   
CCE ES   cells/ml    and analyzed after 7 days of differentiation. EB efficiency in
LSC declined as cell density increased. Aggregation occurred readily in SC regardless of cell concentration. Encapsulation effectively controlled cell aggregation, maintaining a high EB efficiency in stirred culture.
EMI47.1     





  LSC <SEP> SC <SEP> ESC
<tb> 103 <SEP> ES <SEP> cell/ml <SEP> 429% <SEP>  > 0. <SEP> 1% <SEP> 4121%
<tb> 104 <SEP> ES <SEP> cells/ml <SEP> 8. <SEP> 5+2. <SEP> 7% <SEP>  > 0.1% <SEP> 353%
<tb> 
TABLE 5
Fold expansion of total cells in liquid culture (LSC), stirred culture (SC), and encapsulated stirred culture (ESC). Cultures were initiated with   104    R1
ES cells/ml and analyzed after 7 days of differentiation. The table shows results of fold expansion, after adjusting for cell loss during encapsulation procedure. Yield of encapsulated ES cells was approximately 25% of input.



  Cells were lost during the various transferring stages between centrifuge tubes, or retained in the mineral oil phase.
EMI47.2     





  LSC <SEP> SC <SEP> ESC
<tb> 10"ES <SEP> cells/ml <SEP> 2713 <SEP>  >  <SEP> 1 <SEP> 6333
<tb>  
TABLE 6
Hematopoietic and endothelial cell surface markers.
EMI48.1     





  Flk-1 <SEP> Vascular <SEP> endothelial <SEP> growth <SEP> factor <SEP> receptor <SEP> expressed <SEP> by <SEP> early <SEP> mesoderm,
<tb> vascular <SEP> progenitor, <SEP> and <SEP> endothelial <SEP> cells.
<tb>



  CD34 <SEP> Antigen <SEP> expressed <SEP> by <SEP> hematopoietic <SEP> stem, <SEP> hematopoietic <SEP> progenitor, <SEP> and
<tb> endothelial <SEP> cells.
<tb>



  CD45 <SEP> Antigen <SEP> expressed <SEP> by <SEP> hematopoietic <SEP> cells
<tb> CD31 <SEP> Cell <SEP> adhesion <SEP> molecule <SEP> expressed <SEP> by <SEP> platelets, <SEP> endothelial <SEP> cells, <SEP> and <SEP> murine
<tb> ES <SEP> cells.
<tb>  



   TABLE 7
Comparison of cell proliferation and differentiation between standard
ES cell differentiation culture: liquid culture (LSC), and bioprocess of the invention: encapsulated stirred culture (ESC). Proliferation reported as total cells per EB.   Flk-1    expressing mesoderm with hematopoietic potential was assayed on day 4 of differentiation culture. Hematopoietic progenitors coexpressing CD34 and CD45, and/or capable of hematopoietic colony formation (CFC) were assayed on day 7.
EMI49.1     


<tb>



  Analysis <SEP> LSC <SEP> ESC
<tb> Day <SEP> 4-Mesoderm
<tb> Total <SEP> Cells/EB <SEP> 700300 <SEP> 1200450
<tb> Flk-1+ <SEP> cells <SEP> 4412% <SEP> 388%
<tb> Day <SEP> 7-Hematopoietic
<tb> Total <SEP> Cells/EB <SEP> 30001500 <SEP> 3500700
<tb> CD34+CD45+ <SEP> cells <SEP> 1. <SEP> 91. <SEP> 2% <SEP> 1. <SEP> 00. <SEP> 6%
<tb> CFCs <SEP> per <SEP> 105 <SEP> cells <SEP> 14. <SEP> 06. <SEP> 0 <SEP> 5. <SEP> 02. <SEP> 0
<tb> 
TABLE 8
Comparison of hematopoietic cell development (on day 7 of differentiation culture) between normoxic ( > 160 mmHg   02)    and hypoxic (3040 mmHg   02)    encapsulated stirred culture (ESC).
EMI49.2     





  Analysis <SEP> (Day <SEP> 7) <SEP> Normoxic <SEP> ESC <SEP> Hypoxic <SEP> ESC
<tb> Total <SEP> CD34+ <SEP> cells <SEP> 6. <SEP> 1% <SEP> 8.0%
<tb> Total <SEP> CD45+ <SEP> cells <SEP> 0.9% <SEP> 2.6%
<tb> CD34+CD45+ <SEP> cells <SEP> 0.6% <SEP> 1. <SEP> 6%
<tb> CFCs <SEP> per <SEP> 105 <SEP> cells <SEP> 52. <SEP> 0 <SEP> 72. <SEP> 6
<tb>  
TABLE 9
Cell Lineage-Specific Promoters
EMI50.1     


<tb> Promoter <SEP> O <SEP> b <SEP> t <SEP> a <SEP> i <SEP> n <SEP> e <SEP> d <SEP> C <SEP> e <SEP> I <SEP> I <SEP> Literature
<tb> Lineage
<tb> (3-cardiac <SEP> myosin <SEP> heavy <SEP> cardiomyocytes <SEP> Klug <SEP> et <SEP> al. <SEP> (1996) <SEP> J. <SEP> Clin. <SEP> Invest.
<tb> chain <SEP> promoter <SEP> 98: <SEP> 216-24
<tb> MLC-2v <SEP> cardiomyocytes <SEP> Muller <SEP> et <SEP> al. <SEP> (2000) <SEP> FASEB <SEP> J. <SEP> 2000
<tb> Dec; <SEP> 14 <SEP> (15):

   <SEP> 2540-8
<tb> human <SEP> atrial <SEP> natriuretic <SEP> factor <SEP> cardiomyocytes <SEP> Wu <SEP> et <SEP> al. <SEP> (1989) <SEP> J. <SEP> Biol. <SEP> Chem.
<tb>



  *(hANF) <SEP> promoter <SEP> 264: <SEP> 6472-79
<tb> SOX2 <SEP> gene <SEP> neural <SEP> precursors <SEP> Li <SEP> et <SEP> al. <SEP> (1998) <SEP> Curr. <SEP> Biol. <SEP> 8: <SEP> 971-4
<tb> Human <SEP> insulin <SEP> promoter <SEP> Beta <SEP> cells <SEP> Soria <SEP> et <SEP> al. <SEP> (2000) <SEP> Diabetes <SEP> 49: <SEP> 157
<tb> (HIP) <SEP> (insulin-secreting <SEP> cells)
<tb> PDX1 <SEP> promoter <SEP> Beta <SEP> cells <SEP> Gannon <SEP> M. <SEP> et <SEP> al. <SEP> (2001) <SEP> Dev <SEP> Biol.
<tb>



  (insulin-secreting <SEP> cells) <SEP> 2001 <SEP> Oct <SEP> 1; <SEP> 238 <SEP> (1) <SEP> : <SEP> 185-201
<tb> pancreatic <SEP> cells
<tb> Pax4 <SEP> promoter <SEP> Beta <SEP> cells <SEP> Brink <SEP> C. <SEP> et <SEP> al. <SEP> (2001) <SEP> Mech <SEP> Dev. <SEP> 2001
<tb> (insulin-secreting <SEP> cells) <SEP> Jan; <SEP> 100 <SEP> (1): <SEP> 37-43, <SEP> Smith <SEP> SB. <SEP> et <SEP> al.
<tb>



  (2000) <SEP> J <SEP> Biol <SEP> Chem. <SEP> 2000 <SEP> Nov
<tb> 24; <SEP> 275 <SEP> (47): <SEP> 36910-9.
<tb>



  VE-Cadherin <SEP> promoter <SEP> Endothelial <SEP> cells <SEP> Gory <SEP> S. <SEP> et <SEP> al. <SEP> (1999) <SEP> Blood. <SEP> 1999 <SEP> Jan
<tb> 1; <SEP> 93 <SEP> (1) <SEP> : <SEP> 184-92.
<tb> human <SEP> VE-Cadherin-2 <SEP> Endothelial <SEP> cells <SEP> Ludwig <SEP> D. <SEP> et <SEP> al. <SEP> (2000) <SEP> Mamm
<tb> promoter <SEP> Genome. <SEP> 2000 <SEP> Nov; <SEP> 11 <SEP> (11) <SEP> : <SEP> 1030-3
<tb>  
TABLE 10
Examples of cell types derived from the in vitro differentiation of ES cells, as well as examples of additives thought to encourage the differentiation /survival of the specific cell types are indicated. In most cases the additive is added to serum containing media; examples of cells generated in a
Chemically Defined Media (CDM) are also listed.
EMI51.1     





  Cell <SEP> Type <SEP> Generated <SEP> Example <SEP> Additives <SEP> Reference
<tb> Primitive <SEP> ectoderm <SEP> CDM <SEP> Tropepe <SEP> et <SEP> al. <SEP> 2001
<tb> Mesoderm <SEP> precursors <SEP> BMP4, <SEP> Activin <SEP> A <SEP> Wiles <SEP> et <SEP> al. <SEP> 1999
<tb> Endothelial <SEP> cells <SEP> VEGF <SEP> Eichmann <SEP> et <SEP> al. <SEP> 1997
<tb> Hematopoietic <SEP> cells <SEP> IL-11+KL, <SEP> IL-1 <SEP> ICeller <SEP> et <SEP> al. <SEP> 1993
<tb> VEGF+KL <SEP> Kennedy <SEP> et <SEP> al. <SEP> 1997
<tb> VEGF+BMP4 <SEP> Nakayama <SEP> et <SEP> al. <SEP> 2000
<tb> I <SEP> L-2+IL-3+Con <SEP> A <SEP> Chen <SEP> et <SEP> al. <SEP> 1992
<tb> Erythroid <SEP> cells <SEP> Epo <SEP> Wiles <SEP> et <SEP> al. <SEP> 1991
<tb> Myeloid <SEP> cell <SEP> IL-3+1L-1+Epo, <SEP> Wiles <SEP> et <SEP> ai. <SEP> 1991
<tb> IL-3+1L-1+M-CSF/GM-CSF
<tb> IL-3+M-CSF <SEP> Lieschke <SEP> et <SEP> al.

   <SEP> 1995
<tb> Lymphoid <SEP> cells <SEP> VEGF+BMP4 <SEP> Nakayama <SEP> et <SEP> al. <SEP> 2000
<tb> RP. <SEP> 0. <SEP> 010+IL3+IL-6+IL-11 <SEP> Gutierrez-Ramos <SEP> et <SEP> al. <SEP> 1992
<tb> Skeletal <SEP> muscle <SEP> TGFss1/TGFssc2 <SEP> Slager <SEP> et <SEP> al. <SEP> 1993
<tb> DMSO <SEP> Dinsmore <SEP> et <SEP> al. <SEP> 1996
<tb> Cardiacmuscle <SEP> TGss1/TGFss2 <SEP> Slageretal. <SEP> 1993
<tb> RA <SEP> Wobus <SEP> et <SEP> al. <SEP> 1997
<tb> Neuroectoderm <SEP> CDM <SEP> Tropepe <SEP> et <SEP> al. <SEP> 2001
<tb> NGF <SEP> Yamada <SEP> et <SEP> al. <SEP> 1994
<tb> bFGF <SEP> Okabe <SEP> et <SEP> al. <SEP> 1996
<tb> Neurons <SEP> RA <SEP> Bain <SEP> et <SEP> al. <SEP> 1995
<tb> Adipocytes <SEP> RA+insulin+T3 <SEP> Dani <SEP> et <SEP> al. <SEP> 1997
<tb> Beta-cells <SEP> B27 <SEP> Supplement <SEP>  &  <SEP> bFGF <SEP> Lumelsky <SEP> NO. <SEP> et <SEP> al.

   <SEP> 2001
<tb> B27 <SEP> Supplement <SEP>  &  <SEP> Nikotinamid
<tb> Beta-cells <SEP> Nikotinamid <SEP> Soria <SEP> B. <SEP> et <SEP> al. <SEP> 2000
<tb> Beta-cells <SEP> EGF, <SEP> HGF <SEP> and <SEP> Nikotinamid <SEP> Ramiva <SEP> VKM. <SEP> et <SEP> al. <SEP> 2000
<tb>  
TABLE 11
Examples of cell types derived from the in vitro differentiation of human embryonic stem cells, as well as examples of additives thought to encourage the differentiation/survival of the specific cell types are indicated.
EMI52.1     





  Cell <SEP> Type <SEP> Generated <SEP> Example <SEP> Additives <SEP> Reference
<tb> Mesodermal <SEP> cells <SEP> Activin-A <SEP> and <SEP> TGFbeta1 <SEP> Schuldiner <SEP> M, <SEP> et <SEP> al. <SEP> 2000
<tb> Ectodermal, <SEP> Retinoic <SEP> acid <SEP> (RA), <SEP> EGF, <SEP> Schuldiner <SEP> M, <SEP> et <SEP> al. <SEP> 2000
<tb> Mesodermal <SEP> cells <SEP> BMP-4, <SEP> bFGF
<tb> Ectodermal, <SEP> NGF <SEP> and <SEP> HGF <SEP> Schuldiner <SEP> M, <SEP> et <SEP> al. <SEP> 2000
<tb> Mesodermal
<tb> Endodermal <SEP> cells
<tb> Neuronal <SEP> cells <SEP> Retinoic <SEP> acid <SEP> (RA), <SEP> nerve <SEP> Schuldiner <SEP> M, <SEP> et <SEP> al. <SEP> 2001
<tb> growth <SEP> factor <SEP> (betaNGF)
<tb> Hepatocytes <SEP> sodium <SEP> butyrate <SEP> Rambhatla <SEP> L,. <SEP> et <SEP> al. <SEP> 2001
<tb> Beta-cells <SEP> Assady <SEP> S, <SEP> et <SEP> al.

   <SEP> 2001
<tb> Cardiomyocytes <SEP> Kehat <SEP> I, <SEP> et <SEP> al. <SEP> 2001
<tb> Endothelial <SEP> cells <SEP> Levenberg <SEP> S, <SEP> et <SEP> al. <SEP> 2002
<tb>  
FULL CITATIONS FOR REFERENCES REFERRED TO IN THE
SPECIFICATION
1. Assady S, Maor G, Amit M,   Itskovitz-Eldor    J, Skorecki KL,
Tzukerman M. 2001 Diabetes Insulin production by human embryonic stem cells. 50   (8)    : 1691-7.



     2. Bain, G. , D. Kitchens, et al. (1995). "Embryonic stem cells express    neuronal properties in vitro."Dev Biol 168 (2): 342-57.



   3. Bain G, Ray WJ, Yao M, Gottlieb DI : Retinoic acid promotes neural and represses mesodermal gene expression in mouse embryonic stem cells in culture. Biochem Biophys Res Commun 223: 691,
1996.



   4. Beddington RS, Robertson EJ. 1989 An assessment of the developmental potential of embryonic stem cells in the midgestation mouse embryo. Development 105 (4): 733-7.



   5. Bjornson CR, Rietze RL, Reynolds BA,   Magli    MC, Vescovi AL;
Turning brain into blood : a hematopoietic fate adopted by adult neural stem cells in vivo. Science 283: 534-7,1999.



   6. Boraston R, Marshal C, Norman P, Renner G, Warner J;
Elimination of cell aggregation in suspension culture of Chinese hamster ovary (CHO) cells. In : R. E. Spier, J. B. Griffiths, and C.



     MacDonald (eds. ), Animal cell technology : Developments,    processes and products.   Butterworth-Heinemann,    Oxford 1992.



   7. Brink C, Chowdhury K, Gruss P. Pax4 regulator elements mediate beta cell specific expression in the pancreas. Mech Dev. 2001
Jan; 100 (1): 37-43.



     8. Burdsal, C. A. , Damsky, C. H. , and Pedersen, R. A. The role of E-    cadherin and integrins in mesoderm differentiation and migration at the mammalian primitive streak. Development, 118: 829-844  (1993).



   9. Chen U, Kosco M, Staerz U: Establishment and characterization of lymphoid and myeloid mixed-cell populations from mouse late embryoid bodies,"embryonic-stem-cell fetuses". Proc   Natl    Acad Sci
U S A 89: 2541,1992.   



  10. Choi, K. , M. Kennedy, et al. (1998)."A common precursor for    hematopoietic and endothelial   cells."Development    125 (4): 725-32.



  11.   Clarke    DL, Johansson CB,   Wilbertz    J, Veress B,   Nilsson    E,    Karlstrom    H,   Lendahl    U, Frisen J. (2000) Generalized potential of adult neural stem cells. Science 288 (5471): 1660-3.



  12. Dang SM, Kyba   M,    Perlingeiro R,   Daley    GQ, Zandstra PW 2002
Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems. Biotechnol Bioeng   78    (4): 442-53.



  13. Dani C, Smith AG, Dessolin S, Leroy P, Staccini L, Villageois P,
Darimont C, Ailhaud G: Differentiation of embryonic stem cells into adipocytes in vitro. J Cell Sci 110 (Pt 11): 1279,1997.



  14. Dickson MC, Slager HG, Duffie E, Mummery CL, and Akhurst RJ.



   RNA and protein localisations of TGF beta 2 in the early mouse embryo suggest an involvement in cardiac development.



   Development, 117 (2): 625-39 (1993).



  15. Dinsmore J, Ratliff J, Deacon T,   Pakzaban    P, Jacoby D, Galpern    W,    Isacson O : Embryonic stem cells differentiated in vitro as a novel source of cells for transplantation. Cell Transplant 5: 131,
1996.



  16. Doetschman TC, Eistetter H, Katz M, Schmidt W, Kemler R: The in vitro development of   blastocyst-derived    embryonic stem cell lines : formation of visceral yolk sac, blood islands and myocardium. J
Embryol Exp Morphol 87: 27,1985.



  17. Eichmann A, Corbel C, Nataf V, Vaigot P, Breant C, Le Douarin
NM: Ligand-dependent development of the endothelial and hemopoietic lineages from embryonic   mesodermal    cells expressing vascular endothelial growth factor receptor 2. Proc   Natl    Acad Sci U
S A 94: 5141,1997. 



  18. Evans, M. J. and M. H. Kaufman (1981)."Establishment in culture   of pluripotential cells from mouse embryos. "Nature 292 (5819): 154-   
6.   



  19. Fleischmann, M. , W. Bloch, et al. (1998). "Cardiac specific    expression of the green fluorescent protein during early murine embryonic development."FEBS Lett 440 (3): 370-6.



  20. Gannon M, Gamer LW, Wright CV. Regulator regions driving developmental and tissue-specific expression of the essential pancreatic gene   pdx1.    Dev Biol. 2001 Oct 1; 238   (1)    : 185-201.



  21. Lieschke GJ, Dunn AR: Development of functional macrophages from embryonal stem cells in vitro. Exp Hematol 23: 328,1995.



  22. Gory S, Vernet M, Laurent M, Dejana E, Dalmon J, Huber P. The vascular endothelial-cadherin promoter directs endothelial-specific expression in transgenic mice. Blood. 1999 Jan 1; 93   (1)    : 184-92.



  23.   Gutierrez-Ramos    JC, Palacios R : In vitro differentiation of embryonic stem cells into lymphocyte precursors able to generate T and B lymphocytes in vivo. Proc   Natl    Acad Sci U S A   89    : 9171,
1992.



  24. Hadjantonakis AK, Gertsenstein M, Ikawa M, Okabe M, Nagy A.



   1998 Generating green fluorescent mice by germline transmission of green fluorescent ES cells Mech Dev Aug; 76 (1-2): 79-90.



  25. Hadjantonakis AK, Nagy A.. The color of mice: in the light of GFP variant reporters 2001. Histochem Cell Biol 115   (1)    : 49-58.



  26. Irvine DJ, Mayes AM, Satija SK, Barker JG, Sofia-Allgor SJ,   Griffith      LG. , Comparison of tethered star and linear poly (ethylene oxide) for    control of   biomaterials    surface properties. J Biomed Mater Res    1998 Jun 5    ; 40 (3): 498-509.



  27. Jones JM, Thomson JA, Human embryonic stem cell technology,
Semin Reprod Med 2000; 18 (2): 219-23.



  28. Kalyani A, Hobson K, Rao MS. (1997)   Neuroepithelial    stem cells from the embryonic spinal cord: isolation, characterization, and    clonal    analysis. Dev   Biol    186 (2): 202-23. 



  29. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein
A, Livne E, Binah   O,      Itskovitz-Eldor    J, Gepstein L. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001
Aug; 108 (3): 407-14.



  30. Keller, G. M.   (1995)."In    vitro differentiation of embryonic stem cells."Curr Opin Cell Biol 7 (6): 862-9.



  31. Keller G, Kennedy M, Papayannopoulou T, Wiles MV:
Hematopoietic commitment during embryonic stem cell differentiation in culture. Mol   Cell Biol    13: 473,1993.



  32. Kennedy M, Firpo M, Choi   K,    Wall C, Robertson S, Kabrun N, Keller
G: A common precursor for primitive erythropoiesis and definitive haematopoiesis. Nature 386: 488,1997.



     33. Klug MG, Soonpaa MH, Koh GY, Field LJ. , Genetically selected    cardiomyocytes from differentiating embryonic stem cells form stable intracardiac grafts, J Clin   Invest 1996 Jul    1; 98   (1)    : 216-24.



     34. Klug, M. G. , M. H. Soonpaa, et al. (1996)."Genetically selected    cardiomyocytes from differentiating embronic stem cells form stable intracardiac grafts."J Clin Invest 98   (1)    : 216-24.



  35. Ko KS, Arora PD, Bhide V, Chen A,   McCulloch    CA; Cell-cell adhesion in human fibroblasts requires calcium signaling. J Cell Sci
114: 1155-67,2001.   



  36. Larue, L., Antos, C. , Butz, S. , Huber, O., Delmas, V. , Dominis, M.,    and Kemler, R. A role for cadherins in tissue formation.



   Development, 122   :    3185-3194 (1996).



  37. Li M, Pevny L, Lovell-Badge R, Smith A. Generation of purified neural precursors from embryonic stem cells by lineage selection.



   Curr Biol. 1998 Aug 27; 8 (17): 971-4.



  38. Levenberg S, Golub JS, Amit M,   Itskovitz-Eldor    J, Langer R. 2002
Endothelial cells derived from human embryonic stem cells. Proc
Natl Acad Sci U S A 99 (7): 4391-6.



  39. Ludwig D, Lorenz J, Dejana E, Bohlen P,   Hicklin    DJ, Witte L,
Pytowski B.   cDNA    cloning, chromosomal mapping, and expression analysis of human VE-Cadherin-2. Mamm Genome. 2000
Nov; 11   (11)    : 1030-3.



     40. Lumelsky, N. , O. Blondel, P. Laeng, I. Velasco, R. Ravin, and R.   



   McKay. 2001. Differentiation of embryonic stem cells to insulin secreting structures similar to pancreatic islets. Science 292: 1389.



  41. Maltepe E, Schmidt JV, Baunoch D,   Bradfield    CA, Simon MC
1997Nature Abnormal angiogenesis and responses to glucose and oxygen deprivation in mice lacking the protein ARNT
386 (6623): 403-7.



  42. Marshal, E. The business of stem cells. Science, 287 (5457):
1419-1421 (2000).



  43. McKinney-Freeman SL, Jackson KA, Camargo FD, Ferrari G,
Mavilio F, Goodell MA. 2002 Muscle-derived hematopoietic stem cells are hematopoietic in origin. Proc Natl Acad Sci U S A
99 (3): 1341-6.



     44. Muller    M,   Fleischmann    BK, Selbert S, Ji GJ, Endl E, Middeler G,
Muller OJ, Schlenke P, Frese S, Wobus AM, Hescheler J, Katus
HA, Franz WM. Selection of ventricular-like cardiomyocytes from
ES cells in vitro. FASEB J. 2000 Dec; 14 (15): 2540-8.



  45. Mummery C, Ward D, van den Brink CE, Bird SD, Doevendans PA,
Opthof T, Brutel de la Riviere A, Tertoolen L, van der Heyden M,
Pera M. 2002 J Anat Cardiomyocyte differentiation of mouse and human embryonic stem cells. 200 (Pt 3): 233-42.



  46. Nakayama N, Lee J, Chiu L: Vascular endothelial growth factor    synergistically    enhances bone morphogenetic protein-4-dependent    lymphohematopoietic    cell generation from embryonic stem cells in vitro. Blood 95: 2275,2000.



  47. Nagy A, Rossant J, Nagy R, Abramow-Newerly W, Roder JC. 1993
Derivation of completely cell culture-derived mice from early passage embryonic stem cells. Proc Natl Acad Sci U S A
90 (18): 8424-8.



  48. Nichols J,   Zevnik    B, Anastassiadis K, Niwa H, Klewe-Nebenius D,
Chambers   I,    Scholer H, Smith A. 1998 Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell 95 (3): 379-91.



  49.   0kabe    S, Forsberg-Nilsson K, Spiro AC, Segal M, McKay RD:
Development of neuronal precursor cells and functional postmitotic neurons from embryonic stem cells in vitro. Mech Dev 59: 89,1996    50. Palacios, R. , E. Golunski, et al. (1995)."In vitro generation of    hematopoietic stem cells from an embryonic stem cell line."Proc
Natl Acad Sci U S A 92 (16): 7530-4.



  51. Pelton TA, Sharma S, Schulz TC, Rathjen J, Rathjen PD. 2002
Transient pluripotent cell populations during primitive ectoderm formation: correlation of in vivo and in vitro pluripotent cell development. J Cell Sci 115   (Pt 2)    : 329-39.



  52. Perkins AC; Enrichment of blood from embryonic stem cells in vitro. Reprod Fertil Dev 10: 563-72,1998.   



  53. Rambhatla L, . Carpenter MK, Kundu P, Huang L, Chiu CP.   



   Derivation of Hepatocyte like Cells from Human Embryonic Stem
Cells., Abstract Book Keystone Symposia on Pluripotent Stem
Cells : Biology and application. Sheraton Tamarron Resort,
Durango, Colorado. 2001 Feb 6-11; Poster number: 326    54. Ramiya, V. K. , M. Maraist, K. E. Arfors, D. A. Schatz, A. B. Peck,    and J. G. Cornelius. 2000. Reversal of insulin-dependent diabetes using islets generated in vitro from pancreatic stem cells. Nat Med
6: 278.   



  55. Rathjen, P. D. , Lake, J. , Whyatt, L. M. , Bettes, M. D. , and Rathjen, J.   



   Properties and uses of embryonic stem cells : prospects for application to human biology and gene therapy. Reproduction,
Fertility, Development, 10: 31-47 (1998)    56. Rohwedel, J. , V. Maltsev, et al. (1994). "Muscle cell differentiation    of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents."Dev Biol 164   (1)    :
87-101. 



  57. Reubinoff BE, Itsykson P, Turetsky T, Pera MF,   Reinhartz    E,   Itzik A,   
Ben-Hur T. 2001 Neural progenitors from human embryonic stem cells. Nat Biotechnol 19 (12): 1134-40    58. Ryan, C, Nguyen, B. , et al. (1995). "Rapid Assay for Mycobacterial   
Growth and Antibiotic Susceptibility Using Gel Microdrop
Encapsulation."J. Clin. Microbiology 33 (7): 1720-1726.



  59. Schwartz RE, Reyes M, Koodie L, Jiang Y,   Blackstad    M, Lund T,
Lenvik T, Johnson S, Hu WS, Verfaillie CM. (2002)."Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells."J Clin Invest 109 (10): 1291-302.



  60. Schuldiner M, Yanuka O, Itskovitz-Eldor J, Melton DA, Benvenisty
N. Effects of eight growth factors on the differentiation of cells derived from human embryonic stem cells. Proc Nat Acad Sci U S
A. 2000 Oct 10; 97 (21): 11307-12.



  61. Schuldiner M, Eiges R, Eden A, Yanuka O,   Itskovitz-Eldor    J,    Goldstein    RS, Benvenisty N. Induced neuronal differentiation of human embryonic stem cells. Brain Res. 2001 Sep 21; 913 (2): 201
5.



  62. Sefton MV, Hwang JR, Babensee JE. Selected aspects of the microencapsulation of mammalian cells in HEMA-MMA. Ann N Y   Acad Sci. , 831: 260-70 (1997)    63. Slager HG, Van   Inzen    W, Freund E, Van den Eijnden-Van Raaij AJ,
Mummery CL: Transforming growth factor-beta in the early mouse embryo: implications for the regulation of muscle formation and implantation. Dev Genet 14: 212,1993 64. Smith SB, Watada H, Scheel DW, Mrejen C, German MS.



   Autoregulation and maturity onset diabetes of the young transcription factors control the human PAX4 promoter. J Biol
Chem. 2000 Nov 24; 275 (47): 36910-9.



  65. Soria B. 2001 In-vitro differentiation of pancreatic beta-cells.



   Differentiation 68 (4-5): 205-19. 



     66. Soria, B. , A. Skoudy, et al. (2001). "From stem cells to beta cells :    new strategies in cell therapy of diabetes   mellitus."Diabetologia   
44 (4): 407-15    67. Soria, B. , E. Roche, et al. (2000)."Insulin-secreting cells derived    from embryonic stem cells normalize glycemia in streptozotocin   induced diabetic mice. "Diabetes 49 (2): 157-62.



  68. Soria, B. , E. Roche, G. Berna, T. Leon-Quinto, J. A. Reig, and F.   



   Martin. 2000. Insulin-secreting cells derived from embryonic stem cells normalize glycemia in streptozotocin-induced diabetic mice.



   Diabetes 49: 157.



  69. Suzuki A, Zheng YW, Kaneko S, Onodera M, Fukao K, Nakauchi H,
Taniguchi H. 2002   Clonal    identification and characterization of self renewing pluripotent stem cells in the developing liver. J Cell Biol
156   (1)    : 173-84 70. T Tanaka S, Kunath T, Hadjantonakis AK, Nagy A, Rossant J.



   Promotion of   trophoblast    stem cell proliferation by FGF4.1998
Science 282 (5396): 2072-5    71. Thomson, J. A. , J. Itskovitz-Eldor, et al. (1998). "Embryonic stem    cell lines derived from human blastocysts."Science 282 (5391):
1145-7.



  72. Thomson JA, Odorico JS. 2000 Human embryonic stem cell and embryonic germ cell lines. Trends Biotechnol 18 (2): 53-7.



  73. Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F, Sadikot A,    Kaplan    DR, Miller FD. 2001 Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol Sep; 3 (9): 778-84 74. Tropepe V, Hitoshi S, Sirard C, Mak TW, Rossant J, van der Kooy
D: Direct neural fate specification from embryonic stem cells : a primitive mammalian neural stem cell stage acquired through a default mechanism. Neuron 30: 65,2001    75. Tropepe, V. , M. Sibilia, et al. (1999). "Distinct neural stem cells    proliferate in response to EGF and FGF in the developing mouse telencephalon."Dev Biol 208   (1)    : 166-88. 



  76. Turcanu V, Williams NA: Cell identification and isolation on the basis of cytokine secretion: a novel tool for investigating immune responses. Nat Med 7: 373,2001   77. Vallbacka JJ, Nobrega JN, Sefton MV. , Tissue engineering as a    platform for controlled release of therapeutic agents: implantation of    microencapsulated    dopamine producing cells in the brains of rats.,
J Control Release 2001 May 14; 72 (1-3): 93-100   78. Wang, R. , R. Clark, et al. (1992). "Embryonic stem cell-derived    cystic embryoid bodies form vascular channels : an in vitro model of blood vessel development."Development 114 (2): 303-16.



  79.   Wartenberg    M, Gunther J, Hescheler J, Sauer H. Lab Invest 1998
The embryoid body as a novel in vitro assay system for    antiangiogenic agents.    78 (10): 1301-14   80. Weaver, J. C, Williams, G. B., Klibanov, A. , and Demain, A. L. Gel       microdroplets    : rapid detection and enumeration of individual microorganisms by their metabolic activity. Biotechnology, 6: 1084
1089 (1988) 81. Wiles, M. V. and Johansson, B. M. Embryonic stem cell development in chemically defined medium. Experimental Cell
Research, 247: 241-248 (1999) 82. Wiles MV, Keller G: Multiple hematopoietic lineages develop from embryonic stem (ES) cells in culture. Development 111: 259,1991 83.

   Wobus AM, Kaomei G, Shan J, Wellner MC,   Rohwedel    J, Ji G,
Fleischmann B, Katus HA, Hescheler J, Franz WM: Retinoic acid accelerates embryonic stem cell-derived cardiac differentiation and enhances development of   ventricular    cardiomyocytes. J Mol Cell    Cardio 29    : 1525,1997 84. Wu J, LaPointe MC, West BL, Gardner DG. Tissue-specific determinants of human atrial natriuretic factor gene expression in cardiac tissue. J Biol Chem. 1989Apr 15 ; 264 (11): 6472-9.



  85. Yamada G, Kioussi C, Schubert FR, Eto Y, Chowdhury K, Pituello
F, Gruss P: Regulated expression of Brachyury (T),   Nkx1.      1    and Pax genes in embryoid bodies. Biochem Biophys Res Commun
199: 552,1994   86. Zandstra, P. W., Conneally, E. , Petzer, A. L. , Piret, J. M. , and Eaves,   
C. J.,"Cytokine manipulation of primitive human hematopoietic cell self-renewal",. Proc. Natl. Acad. USA, 94: 4698-4703,1997 87. Zandstra PW, Nagy A.   2001Stem cell    bioengineering. Annu Rev
Biomed Eng 3: 275-305.



     88. Zandstra, P. W. , D. A. Lauffenburger, et al. (2000). "A ligand-    receptor signaling threshold model of stem cell differentiation control : a biologically conserved mechanism applicable to hematopoiesis."Blood 96 (4): 1215-22.

Claims

WHAT IS CLAIMED IS : 1. A method of generating cells derived from spheroid-forming cells comprising culturing spheroid-forming cells under controlled cell aggregation conditions that enable spheroid formation in liquid suspension.
2. The method of claim 1 wherein the conditions enable spheroid formation in liquid suspension under high cell density.
3. The method of claim 1 or 2 wherein the cells are kept in liquid suspension by stirring.
4. The method of any one of claims 1-3 wherein controlled cell aggregation conditions means preventing spheroid aggregation.
5. The method of any one of claims 1-4 wherein controlled cell aggregation conditions means permitting cell aggregation sufficient for spheroid formation and preventing aggregation between cells of different spheroids.
6. The method of claim 4 or 5 wherein the preventing aggregation is done by any one or more of the following means: (a) preventing physical association between cell aggregation molecules ; (b) removing inorganic salts from the cell culture; (c) blocking surface aggregation molecules with specific peptides; (d) using spheroid-forming cells that are cell adhesion molecule-null spheroid-forming cells ; and (e) inhibiting cell adhesion molecule expression and/or activity.
Methods that prevent cell aggregation by physically separating one ore more spheroid-forming cells to enable spheroid formation in distinct compartments such as described in the single cell liquid suspension culture (scLSC) method in this patent without limitation to a single cell.
7. The method of any one of claims 1-6 wherein the controlled aggregation is affected by encapsulation of spheroid-forming cells.
8. The method of claim 7 wherein each capsule contains an initial number of spheroid-forming cells sufficient to enable spheroid formation.
9. The method of claim 8 wherein one spheroid is formed per capsule.
10. The method of claim 8 wherein the initial number of cells is 1.
11. The method of claim 7 wherein the capsule initially comprises multiple cells of at least two different cell types.
12. The method of claim 11 wherein a spheroid is formed and the spheroid is a chimeric spheroid.
13. The method of claim 7 wherein the capsules have a capsule body comprising a matrix that permits passage of desired media constituents through the matrix to the spheroid-forming cells and spheroid, but do not permit aggregation with cells outside the capsule.
14. The method of 7 wherein the gel microdrop method is used to encapsulate the spheroid-forming cells.
15. The method of any one of claims 1-14 wherein the cells are cultured under conditions that promote cell expansion but not cell differentiation.
16. The method of any one of claims 1-14 wherein the cells are cultured under conditions that promote cell differentiation.
17. The method of claim 16 wherein the cells are cultured in an environment comprising one or more of the following: growth factors, cytokines, extra cellular matrix molecules, drugs, low molecular weight molecules or compounds, retinoic acid, DMSO.
18. The method of claim 16 or 17 wherein differentiated spheroid-forming cells of interest are selected and harvested.
19. The method of claim 18 wherein the cells of interest are selected on the basis of cell surface markers.
20. The method of claim 18 wherein the differentiated cells of interest are selected from the group consisting of: cardiac myocytes, vascular progenitor cells, neurons, hepatocytes, glial cells, kidney cells, beta cells and derivatives thereof.
21. A method for the selection of specific cell types from spheroid-forming cells comprising the steps of: i. introducing a reporter gene expressing vector into at least one spheroid-forming cell whereby a cell-type specific promoter is combined with an reporter gene, such that the reporter gene is expressed under the control of the cell-type specific promoter; ii. culturing the spheroid-forming cell (s) iii. differentiating spheroid-forming cell (s) in accordance with the method of claim 16 iv. isolating and harvesting the specific cell-type based on the reporter gene expression 22. The method for the selection according to claim 21, wherein the reporter gene is an antibiotic resistance gene and the cell-type of interest is isolated by the addition of an appropriate antibiotic in step (iii) or/and (iv).
23. The method for the selection according to claim 22. wherein the antibiotic resistance gene is selected from but not limited to the group consisting of the Hygro mycin resistence gene (hph), the Zeocin resistence gene (Sh ble), the Puromycin resistance gene (pacA), and the Gentamycin of G418 resistance gene (aph).
24. The method for the selection according to claim 21, wherein the reporter gene is selected from but not limited to the group consisting of luciferase, green fluorescence protein, red fluorescence protein, and yellow fluorescence protein and the cells of interest are selected by fluorencent activated cell sorting (FACS).
25. The method for the selection according to claim 21, wherein the reporter gene is selected from but not limited to the group consisting of luciferase, green fluorescence protein, red fluorescence protein, and yellow fluorescence protein and the cells of interest are selected by fluorencent activated cell sorting (FACS).
26. The method for the selection according to claim 21, wherein the reporter gene is selected from but not limited to the group consisting of luciferase, green fluorescence protein, red fluorescence protein, yellow fluorescence protein, or a his-, myc-, or flag-tag ligated to a heterologous gene, or any heterologous gene which when expressed is inserted into the cell surface and the cells of interest are isolated from the cultured cells by affinity purification.
27. The method for the selection according to claim 21, wherein the cell type specific promoter is selected from but not limited to the group consisting of alpha-cardiac myosin heavy chain, MLC-2v, hANF, SOX2, HIP, PDX1, VE-Cadherin, human VE-Cadherin-2 and the selected cell type of interest is a cardiomyocyte, neural precursor, beta cell, endothelial cell.
28. The method of any one of claims 1-27 wherein the spheroid-forming cells are pluripotent cells.
29. The method of claim 28 wherein the pluripotent cells are selected from the group consisting of embryonic stem cells, embryonic germ layer cells, early primitive ectoderm-like cells, neural stem cells and adult pluripotent cells, adult mesenchymal stem cells, ductal setm cells, muscle derived stem cells, multiple adult progenitor cells.
30. The method of claim 29 wherein the pluripotent cells are embryonic stem cell and the spheroid is an embryoid body.
31. Embryonic stem cell derived cell culture obtained using the method of any one of claims 1-30.
32. The Spheroid-forming cell derived cell culture obtained using the method of anyone of claims 1-30.
33. A method to identify factors that effect cell proliferation, differentiation and/or spheroid formation, said method comprising culturing the cells as per the method of any one of claims 1-30 in the presence of the factor to be tested and then monitoring the effect of the factor on cell proliferation, differentiation and/or spheroid formation.
34. A method of generating cells derived from spheroid-forming cells in accordance with any one of claims 1-30 wherein the spheroid-forming cells are cultured under conditions that enable spheroid formation, said conditions comprising culturing said spheroid-forming cells in liquid suspension under non-aggregating conditions.
35. A culture bioreactor for industrial production of cells derived from spheroid-forming cells comprising: c. culturing spheroid-forming cells in a spheroid-forming cell suitable media under conditions that promote spheroid formation; while d. Inhibiting spheroid aggregation.
36. A method to prevent aggregation between cells comprising encapsulating a cell or group of cells and thus preventing aggregation o said cells with cells not within said capsule.
37. A bioreactor for generating cells from spheroid-forming cells comprising a means for controlling conditions suitable for spheroid formation, such conditions comprising a means for controlling cellular aggregation and means for maintaining said cells and generated cells in suspension.
38. The bioreactor of claim 37 wherein said means for controlling cellular aggregation is means for preventing cellular aggregation.
39. The bioreactor of claim 37 or 38 which is seeded with at least one spheroid-forming cell.
40. The bioreactor of claim 37,38 or 39 wherein at least one spheroid forming cell is encapsulated.
PCT/CA2002/001022 2001-07-04 2002-07-04 A bioprocess for the generation of cells derived from spheroid-forming cells WO2003004626A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP02745012A EP1407002A2 (en) 2001-07-04 2002-07-04 A bioprocess for the generation of cells derived from spheroid-forming cells
AU2002317110A AU2002317110B2 (en) 2001-07-04 2002-07-04 A bioprocess for the generation of cells derived from spheroid-forming cells
CA002488550A CA2488550A1 (en) 2001-07-04 2002-07-04 A bioprocess for producing uniform embryoid bodies from embryonic stem cells in a high density bioreactor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CA002351156A CA2351156A1 (en) 2001-07-04 2001-07-04 A bioprocess for the generation of pluripotent cell derived cells and tissues
US30270601P 2001-07-05 2001-07-05
US10/189,642 US7547547B2 (en) 2001-07-04 2002-07-03 Bioprocess for producing uniform embryoid bodies from embryonic stem cells in a high density bioreactor

Publications (4)

Publication Number Publication Date
WO2003004626A1 true WO2003004626A1 (en) 2003-01-16
WO2003004626A9 WO2003004626A9 (en) 2003-03-06
WO2003004626A3 WO2003004626A3 (en) 2003-05-15
WO2003004626B1 WO2003004626B1 (en) 2003-07-17

Family

ID=27171569

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2002/001022 WO2003004626A1 (en) 2001-07-04 2002-07-04 A bioprocess for the generation of cells derived from spheroid-forming cells

Country Status (5)

Country Link
US (1) US7547547B2 (en)
EP (1) EP1407002A2 (en)
AU (1) AU2002317110B2 (en)
CA (2) CA2351156A1 (en)
WO (1) WO2003004626A1 (en)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004039966A1 (en) * 2002-10-31 2004-05-13 Pfizer Products Inc. Suspension method for producing embryoid bodies, and compositions and methods related thereto
WO2011014740A3 (en) * 2009-07-31 2011-07-07 Chromocell Corporation Methods and compositions for identifying and validating modulators of cell fate
WO2011160128A2 (en) 2010-06-18 2011-12-22 Cellular Dynamics International, Inc. Cardiomyocyte medium with dialyzed serum
US8415155B2 (en) 2009-10-19 2013-04-09 Cellular Dynamics International, Inc. Cardiomyocyte production
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9181529B2 (en) 2010-10-19 2015-11-10 Cellular Dynamics International, Inc. Titration of differentiation medium components
US9487752B2 (en) 2011-03-30 2016-11-08 Cellular Dynamics International, Inc. Priming of pluripotent stem cells for neural differentiation
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
WO2017075389A1 (en) 2015-10-30 2017-05-04 The Regents Of The Universtiy Of California Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US10047345B2 (en) 2012-02-13 2018-08-14 Gamida-Cell Ltd. Culturing of mesenchymal stem cells with FGF4 and nicotinamide
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
US11357799B2 (en) 2014-10-03 2022-06-14 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7160719B2 (en) 2002-06-07 2007-01-09 Mayo Foundation For Medical Education And Research Bioartificial liver system
US8148152B2 (en) * 2003-07-08 2012-04-03 Axiogenesis Ag Method for the preparation of embryoid bodies (EBs) and uses thereof
US20050053586A1 (en) * 2003-09-04 2005-03-10 Bryan Conn Entrapped stem cells and uses thereof
US20050054100A1 (en) * 2003-09-08 2005-03-10 Rennard Stephen I. Methods for fibroblast differentiation
WO2005095578A1 (en) 2004-03-05 2005-10-13 Dsm Ip Assets B.V. Process for cell culturing by continuous perfusion and alternating tangential flow
EP1740945B1 (en) * 2004-04-07 2018-09-19 Ncardia AG Non-invasive, in vitro functional tissue assay systems
EP2270196B1 (en) 2004-05-11 2016-04-20 Axiogenesis Ag Assay for drug discovery based on in vitro differentiated cells
US20050277124A1 (en) * 2004-06-10 2005-12-15 White Steven M Cardiac conduction system cells and uses thereof
EP1784481A1 (en) * 2004-08-25 2007-05-16 Technion Research And Development Foundation, Ltd. Methods of generating embryoid bodies using three dimensional scaffolds
US20080241925A1 (en) * 2004-09-02 2008-10-02 Angela Paz Judith Cruz Three-Dimensional Self Assembly in Suspension of Adherent Cells
CA2583308C (en) * 2004-10-08 2020-01-07 Georgia Tech Research Corporation Microencapsulation of cells in hydrogels using electrostatic potentials
US7225078B2 (en) * 2004-11-03 2007-05-29 Halliburton Energy Services, Inc. Method and system for predicting production of a well
US7803619B2 (en) * 2004-11-24 2010-09-28 Geneprotech, Inc. Embryoid body-based screen
WO2006122147A2 (en) * 2005-05-10 2006-11-16 Rutgers, The State University Of New Jersey Alginate poly-l-lysine encapsulation as a technology for controlled differentiation of embryonic stem cells
US20070042491A1 (en) * 2005-08-18 2007-02-22 Karp Jeffrey M Amplification of cell populations from embryonic stem cells
EP1962719A4 (en) 2005-08-29 2011-05-04 Technion Res And Dev Of Foundation Ltd Media for culturing stem cells
JP5386180B2 (en) 2006-02-28 2014-01-15 ザ トラスティーズ オブ コロンビア ユニヴァーシティ イン ザ シティ オブ ニューヨーク Method for compact aggregation of dermal cells
EP3441459B1 (en) * 2006-08-02 2021-03-17 Technion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
SG187493A1 (en) 2008-01-18 2013-02-28 Univ Minnesota Stem cell aggregates and methods for making and using
WO2009100128A1 (en) * 2008-02-04 2009-08-13 Massachusetts Institute Of Technology Particulate delivery vehicles for embryoid bodies
US20110165128A1 (en) * 2008-03-07 2011-07-07 Columbia University In The City Of New York Homing in mesenchymal stem cells
EP2138571B1 (en) * 2008-06-26 2017-04-12 SpheroTec GmbH Process for the preparation of multicellular spheroids
EP2199383B1 (en) * 2008-12-22 2017-02-15 SpheroTec GmbH Methods for the preparation of fibroblasts
US10130748B2 (en) 2009-03-13 2018-11-20 Mayo Foundation For Medical Education And Research Bioartificial liver
AU2011293440B2 (en) * 2010-08-24 2016-05-05 Katholieke Universiteit Leuven Non-static suspension culture of cell aggregates
AU2013243586A1 (en) * 2012-04-02 2014-11-20 The Children's Mercy Hospital Disposable single use self-contained cyclic pressure and flow bioreactor system
US9790465B2 (en) 2013-04-30 2017-10-17 Corning Incorporated Spheroid cell culture well article and methods thereof
US9550972B2 (en) 2014-09-29 2017-01-24 General Electric Company Devices, systems and methods for automated cell culturing
CN107109339B (en) 2014-10-29 2021-07-02 康宁股份有限公司 Perfusion bioreactor platform
US20180092349A1 (en) * 2015-04-02 2018-04-05 Genea Ip Holdings Pty Limited Handling of biological samples
GB201707143D0 (en) 2017-05-04 2017-06-21 Plasticell Ltd Method for producing cells
US11584906B2 (en) 2017-07-14 2023-02-21 Corning Incorporated Cell culture vessel for 3D culture and methods of culturing 3D cells
US11857970B2 (en) 2017-07-14 2024-01-02 Corning Incorporated Cell culture vessel
PL3652291T3 (en) 2017-07-14 2022-03-28 Corning Incorporated Cell culture vessel
EP3652290B1 (en) 2017-07-14 2022-05-04 Corning Incorporated 3d cell culture vessels for manual or automatic media exchange
WO2020013847A1 (en) 2018-07-13 2020-01-16 Corning Incorporated Microcavity dishes with sidewall including liquid medium delivery surface
JP7353263B2 (en) 2018-07-13 2023-09-29 コーニング インコーポレイテッド Cell culture vessel with stabilization device
WO2020013851A1 (en) 2018-07-13 2020-01-16 Corning Incorporated Fluidic devices including microplates with interconnected wells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5073491A (en) * 1988-12-23 1991-12-17 Hoffman-La Roche Inc. Immobilization of cells in alginate beads containing cavities for growth of cells in airlift bioreactors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7795026B2 (en) * 2000-01-21 2010-09-14 The Johns Hopkins University School Of Medicine Methods for obtaining human embryoid body-derived cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5073491A (en) * 1988-12-23 1991-12-17 Hoffman-La Roche Inc. Immobilization of cells in alginate beads containing cavities for growth of cells in airlift bioreactors

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
COSBY N C ET AL: "MICROENCAPSULATION OF SINGLE MULTIPLE AND ZONA PELLUCIDA-FREE MOUSE PREIMPLANTATION EMBRYOS IN SODIUM ALGINATE AND THEIR DEVELOPMENT IN-VITRO", JOURNAL OF REPRODUCTION AND FERTILITY, vol. 90, no. 1, 1990, pages 19 - 24, XP001118759, ISSN: 0022-4251 *
DANG STEPHEN M ET AL: "Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems.", BIOTECHNOLOGY AND BIOENGINEERING. UNITED STATES 20 MAY 2002, vol. 78, no. 4, 20 May 2002 (2002-05-20), pages 442 - 453, XP002219286, ISSN: 0006-3592 *
MAGYAR JOSEF P ET AL: "Mass production of embryoid bodies in microbeads.", ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, vol. 944, November 2001 (2001-11-01), Conference on Bioartificial Organs, III: Tissue Sourcing, Immunoisolation, and Clinical Trials;Davos, Switzerland; October 07-11, 2000, Tissue sourcing, immunoisolation, and clinical trials. November,, pages 135 - 143, XP001118571, ISBN: 1-57331-342-4 *
PAPAS K K ET AL: "Cultivation of recombinant, insulin-secreting AtT-20 cells as free and entrapped spheroids.", CYTOTECHNOLOGY, vol. 13, no. 1, 1993, pages 1 - 12, XP001118058, ISSN: 0920-9069 *
SAKAI YASUYUKI ET AL: "A new bioartificial liver using porcine hepatocyte spheroids in high-cell-density suspension perfusion culture: In vitro performance in synthesized culture medium and in 100% human plasma.", CELL TRANSPLANTATION, vol. 8, no. 5, 1999, pages 531 - 541, XP001118579, ISSN: 0963-6897 *
WARTENBERG M ET AL: "The embryoid body as a novel in vitro assay system for antiangiogenic agents.", LABORATORY INVESTIGATION;A JOURNAL OF TECHNICAL METHODS AND PATHOLOGY. UNITED STATES OCT 1998, vol. 78, no. 10, October 1998 (1998-10-01), pages 1301 - 1314, XP001107032, ISSN: 0023-6837 *

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004039966A1 (en) * 2002-10-31 2004-05-13 Pfizer Products Inc. Suspension method for producing embryoid bodies, and compositions and methods related thereto
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
WO2011014740A3 (en) * 2009-07-31 2011-07-07 Chromocell Corporation Methods and compositions for identifying and validating modulators of cell fate
US8945848B2 (en) 2009-07-31 2015-02-03 Chromocell Corporation Methods and compositions for identifying and validating modulators of cell fate
US9657357B2 (en) 2009-07-31 2017-05-23 Chromocell Corporation Methods and compositions for identifying and validating modulators of cell fate
US10604739B2 (en) 2009-10-19 2020-03-31 FUJIFILM Cellular Dynamics, Inc. Cardiomyocyte production
US9957482B2 (en) 2009-10-19 2018-05-01 Cellular Dynamics International, Inc. Cardiomyocyte production
US8415155B2 (en) 2009-10-19 2013-04-09 Cellular Dynamics International, Inc. Cardiomyocyte production
US9365827B2 (en) 2009-10-19 2016-06-14 Cellular Dynamics International, Inc. Cardiomyocyte production
EP3255142A1 (en) 2009-10-19 2017-12-13 Cellular Dynamics International, Inc. Cardiomyocyte production
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US9365826B2 (en) 2010-06-18 2016-06-14 Cellular Dynamics International, Inc. Cardiomyocyte medium with dialyzed serum
WO2011160128A2 (en) 2010-06-18 2011-12-22 Cellular Dynamics International, Inc. Cardiomyocyte medium with dialyzed serum
US9181529B2 (en) 2010-10-19 2015-11-10 Cellular Dynamics International, Inc. Titration of differentiation medium components
US9487752B2 (en) 2011-03-30 2016-11-08 Cellular Dynamics International, Inc. Priming of pluripotent stem cells for neural differentiation
US10047345B2 (en) 2012-02-13 2018-08-14 Gamida-Cell Ltd. Culturing of mesenchymal stem cells with FGF4 and nicotinamide
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US10457942B2 (en) 2012-08-13 2019-10-29 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US11220687B2 (en) 2012-08-13 2022-01-11 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US11357799B2 (en) 2014-10-03 2022-06-14 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
WO2017075389A1 (en) 2015-10-30 2017-05-04 The Regents Of The Universtiy Of California Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11872251B2 (en) 2016-01-11 2024-01-16 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery

Also Published As

Publication number Publication date
US7547547B2 (en) 2009-06-16
AU2002317110B2 (en) 2008-06-12
US20030119107A1 (en) 2003-06-26
WO2003004626B1 (en) 2003-07-17
WO2003004626A3 (en) 2003-05-15
EP1407002A2 (en) 2004-04-14
CA2488550A1 (en) 2003-01-16
CA2351156A1 (en) 2003-01-04
WO2003004626A9 (en) 2003-03-06

Similar Documents

Publication Publication Date Title
AU2002317110B2 (en) A bioprocess for the generation of cells derived from spheroid-forming cells
AU2002317110A1 (en) A bioprocess for the generation of cells derived from spheroid-forming cells
Dang et al. Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems
Cameron et al. Improved development of human embryonic stem cell‐derived embryoid bodies by stirred vessel cultivation
Kurosawa Methods for inducing embryoid body formation: in vitro differentiation system of embryonic stem cells
JP5603300B2 (en) Cell culture
Sheridan et al. Analysis of embryoid bodies derived from human induced pluripotent stem cells as a means to assess pluripotency
EP2691512B1 (en) Enriched populations of cardiomyocyte lineage cells from pluripotent stem cells
KR20150067394A (en) Suspension culture of human embryonic stem cells
EP1792979A1 (en) Cell culture system for the enrichment and expansion of stem cells
JP2014039556A (en) Method for identifying modulator of cell signaling
Bahrami et al. Isolation and expansion of endothelial progenitor cells derived from mouse embryonic stem cells
AU2008204566B2 (en) Novel mesenchymal progenitor cells derived from human blastocyst-derived stem cells
WO2023054325A1 (en) Method for separating pituitary cells and hypothalamic cells using cell surface marker
Bauwens et al. Engineering the Pluripotent Stem Cell Niche for Directed Mesoderm Differentiation
AU2012203350B2 (en) Suspension culture of human embryonic stem cells
dos Santos Haematopoietic differentiation of normal and PU. 1 mutated murine embryonic stem cells using hydrogel encapsulation in a HARV bioreactor
Bigdeli To my grandfather
Levenberg et al. Human embryonic stem cell culture for Tissue Engineering
Bigdeli Derivation, Characterization and Differentiation of Feeder-Free Human Embryonic Stem Cells
Dang Scalable bioprocess for controlled differentiation of embryonic stem cells to hematopoietic progenitors
Gerecht-Nir et al. Differentiation of human ES cells
Purpura Controlling the Emergence of Hematopoietic Progenitor Cells from Pluripotent Stem Cells

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

AK Designated states

Kind code of ref document: C2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGE 47, DESCRIPTION, REPLACED BY A NEW PAGE 47; AFTER RECTIFICATION OF OBVIOUS ERRORS AS AUTHORIZED BY THE INTERNATIONAL SEARCHING AUTHORITY

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
B Later publication of amended claims

Free format text: 20030411

WWE Wipo information: entry into national phase

Ref document number: 2002317110

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2002745012

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2002745012

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 2488550

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP