AU2015268664A1 - Methods for purifying cells derived from pluripotent stem cells - Google Patents

Methods for purifying cells derived from pluripotent stem cells Download PDF

Info

Publication number
AU2015268664A1
AU2015268664A1 AU2015268664A AU2015268664A AU2015268664A1 AU 2015268664 A1 AU2015268664 A1 AU 2015268664A1 AU 2015268664 A AU2015268664 A AU 2015268664A AU 2015268664 A AU2015268664 A AU 2015268664A AU 2015268664 A1 AU2015268664 A1 AU 2015268664A1
Authority
AU
Australia
Prior art keywords
cells
cell
markers characteristic
lineage
pancreatic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2015268664A
Other versions
AU2015268664B2 (en
Inventor
Francis Karanu
Alireza Rezania
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Biotech Inc
Original Assignee
Janssen Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2011223900A external-priority patent/AU2011223900A1/en
Application filed by Janssen Biotech Inc filed Critical Janssen Biotech Inc
Priority to AU2015268664A priority Critical patent/AU2015268664B2/en
Publication of AU2015268664A1 publication Critical patent/AU2015268664A1/en
Priority to AU2017276263A priority patent/AU2017276263B2/en
Application granted granted Critical
Publication of AU2015268664B2 publication Critical patent/AU2015268664B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Abstract The present invention is directed to methods to differentiate pluripotent stem cells. In particular, the present invention provides methods of characterization of cells differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage utilizing unique surface markers. The present invention also provides methods to enrich or sort cells expressing markers characteristic of the pancreatic endocrine lineage. The present invention also provides methods to deplete cells that may contaminate populations of cells expressing markers characteristic of the pancreatic endocrine lineage formed by the methods of the present invention, thereby reducing the incidence of tumor formation in vivo following transplantation.

Description

METHODS FOR PURIFYING CELLS DERIVED FROM PLURIPOTENT STEM CELLS [0000] The present application is a divisional application of Australian Application No. 2011223900, which is incorporated in its entirety herein by reference. CROSS REFERENCE TO RELATED APPLICATION [0001] This application claims priority to provisional application serial number 61/309, 193, filed March 1, 2010. FIELD OF THE INVENTION [0002] The present invention is directed to methods to differentiate pluripotent stem cells. In particular, the present invention provides methods of characterization of cells differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage utilizing unique surface markers. The present invention also provides methods to enrich or sort cells expressing markers characteristic of the pancreatic endocrine lineage. The present invention also provides methods to deplete cells that may contaminate populations of cells expressing markers characteristic of the pancreatic endocrine lineage formed by the methods of the present invention, thereby reducing the incidence of tumor formation in vivo following transplantation. BACKGROUND [0003] Pluripotent stem cells have the potential to produce differentiated cell types comprising all somatic tissues and organs. Treatment of diabetes using cell therapy is facilitated by the production of large numbers of cells that are able to function similarly to human islets. Accordingly, there is need for producing these cells derived from pluripotent stem cells, as well as reliable methods for purifying such cells. [0004] Proteins and other cell surface markers found on pluripotent stem cell and cell populations derived from pluripotent stem cells are useful in preparing reagents for the separation and isolation of these populations. Cell surface markers are also useful in the further characterization of these cells. -1 - OO5] In one example, W02009131568 discloses a method of purifying a gut endoderm cell comprising: a) exposing a population of cells derived from pluripotent stem cells comprising a gut endoderm cell to a ligand which binds to a cell surface marker expressed on the gut endoderm cell, wherein said cell surface marker is selected from the group consisting of CD49e, CD99, CD 165, and CD334; and b) separating the gut endoderm cell from cells derived from pluripotent stem cells which do not bind to the ligand, thereby purifying said gut endoderm cell. i006] In another example, WO2010000415 discloses the use of an antibody that binds to the antigen TNAP, or functional fragments of the antibody, alone or in combination with an antibody that binds to CD56, or functional fragments of the antibody, for the isolation of stem cells having adipocytic, chondrocytic and pancreatic differentiation potential. 1007] In another example, U S7371576 discloses the discovery of a selective cell surface marker that permits the selection of a unique subset of pancreatic stems cells having a high propensity to differentiate into insulin producing cells or into insulin producing cell aggregates. 008] In another example, US7585672 discloses a method to enrich a culture derived from human embryonic stem cells for cells of endoderm and pancreatic lineages, the method comprising the steps of (a.) culturing intact colonies of human embryonic stem cells to form whole, intact embryoid bodies surrounded by visceral yolk sac (VYS) cells, wherein the human embryonic stem cells express Oct-4, surface stage-specific embryonic antigen-3/4 (SSEA 3/4) and epithelial cell adhesion molecule (EpCAM); (b) culturing the embryoid bodies of step (a) under conditions that permit the enbryoid body cells to differentiate into a cell population containing cells of the endoderm and pancrea tic lineages; (c) dispersing the cell population of step (b) into single cells; (d) selecting against the expression of SSEA 3/4 positive cells to remove undifferentiated cells from the cells of step (c); (e) selecting against the expression of SSEA-4 positive cells to remove VYS cells front the remaining cells of step (d); and (f) selecting from among the remaining cells of step (e) for the expression of EpCAM positive cells to enrich for cells of endoderm and pancreatic lineages. 2 M09] US7585672 also discloses a method to enrich a culture derived from human embryonic stem cells for cells of endoderm and pancreatic lineages, the method comprising the steps of (a) culturing intact colonies of human embryonic stem cells to form whole, intact embryoid bodies surrounded by visceral yolk sac (VYS) cells, wherein the human embryonic stem cells express Oct-4, surface stage-specific embryonic antigen-3/4 (SSEA 3/4) and, epithelial cell adhesion molecule (EpCAM); (b) culturing the embryoid bodies of step (a) under conditions that permit the embryoid body cells to differentiate into a cell population containing cells of the endoderm and pancreatic lineages; (c) treating the cell population of step (b) with an effective amount of fibroblast growth factor 10 (GFI 0); and (d) dispersing the cell population of step (c) into single cells enriched for cells of endoderm and pancreatic lineages (e) selecting against the expression of SSEA-3/4 positive cells to remove undifferentiated stem cells from the cells of step (d); (f) selecting against the expression of SSEA-1. positive cells to remove VYS cells from the cells of step (e); and (g) selecting from among the remaining cells of step (f) for the expression of EpCAM positive cells to enrich for cells of endoderm and pancreatic lineages, 10010] US7585672 also discloses an enrichment method for the creation of a stem cell derived cell population which does not have tumorigenic capability comprising the steps of (a) culturing intact colonies of human embryonic stern cells to form whole, intact embryoid bodies surrounded by visceral yolk sac (VYS) cells, wherein the human embryonic stem cells express Oct-4, surface stage-specific embryonic antigen 3/4 (SSEA 3/4) and epithelial cell adhesion molecule (EpCAM); (b) culturing the embryoid bodies of step (a) under conditions that permit the embryoid body cells to differentiate into a cell population containing cells of the endoderm and pancreatic lineages; (c) dispersing the cell population of step (b) into single cells; (d) selecting against the expression of SSEA 3/4 positive cells to remove undifferentiated cells from the cells of step (c); (e) selecting against the expression of SSEA-I positive cells to remove VYS cells from the cells of step (d);and (f) selecting from among the remaining cells of step (e) for the expression of EpCAM positive cells, the resulting cells not forming teratomas when injected in immunocompromised mice. 00101 In another example, US20050260749 discloses a method to enrich a culture derived from stem cells for cells of endoderm and pancreatic lineages, the method comprising 3 the steps of culturing stem cells into the formation of embryoid bodies; and selecting among embryoid bodies for the expression of the species appropriate cell surface stage-specific embryonic and culturing only the embryoid bodies which do not express cell surface stage-specific antigen for differentiation into endodermi and pancreatic cells. 1011] hi another example, US120,100003749 discloses an isolated pancreatic stem cell population, wherein the pancreatic stem cell population is enriched for CDI133+CD49f+ pancreatic stem cells. 1012] US20100003749 further discloses the isolation of pancreatic stem cells from primary pancreatic tissue occurs by selecting from a population of pancreatic cells, pancreatic derived cells, or gastrointestinal-derived cells for cells that are C.D133+, CD49f+, or CD133+CD49f;- removing the cells that are CDi5-, wherein the remaining cells are CD15-; introducing the remaining cells to a serum-free culture medium containing one or more growth factors; and proliferating the remaining cells in the culture medium. 013] In another example, Dorrell e cil state: "We have developed a novel panel of cell surface markers for the isolation and study of all major cell types of the human pancreas. Hybridomas were selected after subtractive immunization of Balb/C mice with intact or dissociated human islets and assessed for cell-type specificity and cell surface reactivity by immunohistochemistry and flow cytometry. Antibodies were identified by specific binding of surface antigens on islet (panendocrine or a-specific) and nonislet pancreatic cell subsets (exocrine and duct). These antibodies were used individually or in combination to isolate populations of (, [, exocrine, or duct cells from primary human pancreas by FACS and to characterize the detailed cell composition of human islet preparations. They were also employed to show that human islet expansion cultures originated from nonendocrine cells and that insulin expression levels could be increased to up to 1% of nonal islet cells by subpopulation sorting and overexpression of the transcription factors Pdx-i and ngn3, an improvement over previous results with this culture system. These methods permit the analysis and isolation of functionally distinct pancreatic cell populations with 4 potential for cell therapy" (Stem Cell Research, Volume 1, issue 3, September 2008, Pages 155456). 014] In another example, Sugiyama et al state: "We eventually identified two antigens, called CD133 and CD49f useful for purifying NGN3+ cells from mice. CD133 (also called prominin-1) is a transmembrane protein of unknown function and a known marker of haematopoietic progenitor and neural stem cells. CD49f is also called o6 integrin, and a receptor subunit for laminin, By coining antibodies that recognize CD133 and CD49f, we fractionated four distinct pancreatic cell populations. Immunostaining and RT-PCR revealed that the CD49fthigh CD133-+ cell population (fraction .1 50% of input) comprised mainly differentiated exocrine cells that express CarbA. The CD49flow CD133- fraction (fractionTI, 10% of input) included honnone- cells expressing endocrine products like insulin and glucagon. By contrast, the CD49flow CD133+ fraction (called 'fraction 11% 13% of input) contained NGN3+ cells, but not hormone+ cells, Approximately 8% of fraction II cells produced immunostainable NGN3. In the CD49f-- CD133- fraction (fraction IV% 25% of input), we did not detect cells expressing NGN3, CarbA or ilet hormnnes," (Diabetes. Obesity and Metabolism, Volune 10, Issue s4, Pages 179-185) i015] In another example, Fujikawa et a/ state: "When CD45-TER 119- side-scatterlow GFPhigh cells were sortd, -fetupiotein-positive immature endodem-characterized cells, having high growth potential, were present in this population. Clonal analysis and electron microscopic evaluation revealed that each single cell of this population could differentiate not only into hepatocytes, but also into biliary epitheial Cells, showing their bilineage differentiation activity. When surface markers were analyzed, they were positive for Integrin-a6 and -1, but negative for c-Kit and Thy L ," (Journal qfiepalogy, Vol 39, pages 162-170), 0016] In another example, Zhao e a? state: "In this study, we first identified N-cadherin as a surface marker of hepatic endoderm cells for purification from hES cell-derivates, and generated hepatic progenitor cells from purified hepatic endoderm cells by co culture with marine eibryonic stromal feeders (STO) cells. These hepatic progenitor cells could expand and be passage for more than 100 days. Interestingly, they co expressed the early hepatic marker AFFP and biliary lineage marker KRT7, suggesting 5 that they are a common ancestor of both hepatocytes and cholangiocytes. Moreover, these progenitor cells could be expanded extensively while still maintaining the bipotential of differentiation into hepatocyte-like cells and cholangiocyte-like cells, as verified by both gene expression and functional assays. Therefore, this work offers a new in vitro model for studying liver development, as well as a new source for cell therapy based on hepatic progenitors? (LoS ONE 4(7): e6468. doi: 10. 137 l/journatpone.0006468). 1017] In another example, Cai eta! state: "To further increase the PDX1- cell purity, we sorted the activin A-induced cells using CXCR4 .. a marker for ES cell-derived endodernal cells. Sortina with CXCR4 enriched the endodernal cell population because nearly all the cells in the CXCR4+ population were positive for the endodermal cell marker SOX] 7, and >90% of the cells were positive for FOXA2" (Journal of Molecular Cell Biology Advance Access originally published online on November 12, 2009. Journal of Molecular Cell Biology 2010 2(1):50-60; doi: 10.1 093/jncb/mjp037) 1] In another example, Koblas et a/ state; "We found that population of human CD1 33 positive pancreatic cells contains endocrine progenitors expressing neurogenin-3 and cells expressing human telomerase, ABCG2, Oct-3/4, Nanog, and Rex-], markers of pluripotent stem cells, These cells were able to differentiate into insulin-producing cells in vitro and secreted C-peptide in a glucose-dependent manner. Based on our results, we suppose that the CDI 33 molecule represents another cell surface marker suitable for identification and isolation of pan creatic endocrine progenitors. (Transplant Proc. 2008 Mar;40(2):415-8) 0019j In another example, Sugiyama et al state: "we found CD133 was expressed by NGN3+ cells. CD133 appeared to be localized to the apical membrane of pancreatic ductal epithelial cells." (PNAS 2007 104:175-180; published online before print December 26, 2006, doi:10.1.073/pnas,0609490104), 0020] In another example, Kobayashi el al state: "The embryonic pancreatic epithelium, and later the ductal epithelium, is known to give rise to the endocrine and exoerine cells of the developing pancreas, but no specific surface marker for these cells has 6 been identified. Here, we utilized Dolichos Biflorus Agglutinin (DBA) as a specific marker of these epithelial cells in developing mouse pancreas. From the results of an immunofluorescence study using fluorescein-DBA and pancreatic specific cell markers, we found that DBA detects specifically epithelial, but neither differentiating endocrine cells nor acinar cells. We further applied this marker in an immunomagnetic separation system (Dynabead system) to purify these putative multi potential cells from a mixed developing pancreatic cell population. This procedure could be applied to study differentiation and cell lineage selections in the developing pancreas, and also may be applicable to selecting pancreatic precursor cells for potential cellular engineering." (Biochemical and BiphVysical Research Communications, Volume 293., Issue 2, 3 May 2002, Pages 691-697). 021] Identification of markers expressed by cells derived from pluripotent stem cells would expand the understanding of these cells, aid in their identification in vivo and in vitro, and would enable their positive enrichment in vitro for study and use. Thus, there remains a need for tools that are useful in isolating and. characterizing cells derived from pluripotent stem cells, in particular, cells expressing markers characteristic of the pancreatic endocrine lineage. SUMMARY 1022] In one embodiment, the present invention provides a method to differentiate a population of pluripotent stem cells into a population of cells expressing markers characteristic of the pancreatic endocrine lineage, comprising the steps of a. Culturing a population of phrripotent stem cells, b. Differentiating the population of pluripotent stem cells into a population of cells expressing markers characteristic of the definitive endoderm lineage, c. Differentiating the population of cells expressing markers characteristic of the definitive endoderm lineage into cells expressing markers characteristic of the primitive gut tube lineage, 7 d.. Differentiatig the population of cells expressing markers characteristic of the primitive gut tube lineage into a population of cells expressing markers characteristic of the pancreatic endoderm lineage, and e. Differentiating the population of cells expressing markers characteristic of the pancreatic endoderim lineage into a population cells expressing markers characteristic of the pancreatic endocrine lineage. 1023] In one embodiment, the population of cells expressing markers characteristic of the pancreatic endocrine lineage is transplanted into an animal, wherein the cells expressing markers characteristic of the pancreatic endocrine lineage form insulin producing cells. In one embodim, the fficiency of the formation of insulin producing cells is enhanced by enriching the population .for cells expressing markers characteristic of the pancreatic endocrine lineage prior to transplantation. 024] In one embodiment, the efficiency of the formation. of insulin producing cells is determined by neasur-ing the time taken for the expression of C-peptide to reach detectable levels following transplantation. 1025] In an alternate embodiment, the enrichment decreases the ability of the transplanted cells to form teratonas following transplantation. BRIEF DESCRIPTION OF THE DRAWINGS 1026] Figure I shows the expression of NEUROD (panel a), NGN3 (panel b), PDXI (panel c), NKX6.1 (panel d), NKX2.2 (panel e), and PAX4 (panel f) in populations of CD56 CDl 13, CD56-CDl3~ and CD56~CD13! cells, as detected via real-time PCR. Fold. expression is shown relative to undifferentiated HI embryonic stem cells. 0027] Figure 2 shows the expression of NEUROD (panel a), NGN3 (panel b), PDXIl (panel c), NKX6.1 (panel d), NKX2.2 (panel e), and PAX4 (panel f) as detected via real time PCR, in populations of cells sorted using an antibody to CD 133, Fold expression is shown relative to undifferentiated Hi embryonic stem cells. 0028] Figure 3 shows the expression of NEUROD (panel a), NGN3 (panel b), PDX I (panel c), and NKX6,1 (panel d), as detected via real-time PCR, in populations of cells 8 sorted using an antibody to CD49c. Fold expression is shown relative to undifferentiated 1-1i embryonic stem cells, 029] Figure 4 shows the expression of NEUROD (panel a), NGN3 (panel b), PDX I (panel c), NKX6.1 (panel d). insulin (panel e), and. glucagon (panel ft), as detected via real time PCR, in populations of cells sorted using antibodies to CD56 and CDI 5. Fold expression is shown relative to undifferentiated H I embryonic stem cells. 1030] Figure 5 shows the expression of NEUROD (panel a), NGN3 (panel b), PDXI (panel c), N KX6 1 (panel d), NKX2.2 (panel e), PAX-4 (panel f), glucagon (panel g) and insulin (panel h) as detected via real time PCR, in populations of cells sorted using an antibody to CD15. Fold expression is shown relative to undifferentiated HiI embryonic stem. cells'. '031] Figure 6 shows the expression of NEUROD (panel a), NGN3 (panel b), PDX I (panel c), NKX6.1 (panel d), NKX2.2 (panel e), insulin (panel f), and glucagon (panel g) as detected via real-time PCR, in populations of cells sorted using antibodies to CDS6 and CDS7, Fold expression is shown relative to undifferentiated H1 embryonic stem cells. 032] Figure 7 shows the expression of ZICi (panel a), albumin (panel b), CDX2 (panel c), NGN3 (panel d), PAX4 (panel e), NEUROD (panel f), NKX6.1 (panel g), PTFI alpha u(anel h), and PDXI (panel i), as detected via reaktime PCR, in populations of cells sorted using antibodies to CD56 and CDI84. Fold expression is shown relative to undifferentiated. HI embryonic stem cells. 0033] Figure 8 shows the expression of NEUROD (panel a), NGN3 (panel b), insulin (panel c), and g ucagon (panel d), as detected via real-time PCR., in populations of cells sorted using an antibody to CD98. Fold expression is shown relative to undifferentiated HI embryonic stein cells. 0034] Figure 9 shows the expression of NEUR)OD (panel a), NON3 (panel b), PDX I (panel c), NKX6,1 (panel d), NKX2.2 (panel e), and PAX4 (panel f), as detected via real time PCR, in populations of cells sorted using an antibody to CD47. Fold expression is shown relative to undifferentiated H1 enbryonic stem cells, 9 '035] Figure 10 shows the expression ofPDX-1 (panel a), NKX6.l (panel b), NKX2.2 (panel c), PAX-4 (panel d), PTF I a (panel e), NGN3 (panel ft insulinn (panel g) and glucagon (panel h) as detected via real-time PCR, in populations of cells sorted using an antibody to CD47. Fold expression is shown relative to undifferentia ted H1 embryonic stem cells, 1036] Figure I1 shows the expression of HNF4 alpha (panel a), and LIF receptor (panel b), as detected via real-time PCR, in populations of cells sorted using an antibody to the LIF receptor. Fold expression is shown relative to unsorted cells at DAY 2 of Stage It of the differentiation protocol outlined in Example 1, 10371 Figure 12 shows the expression of OCT4 (panel a), NANOG (panel b), SOX2 (panel c), and goosecoid (panel d), as detected via real-time PCR, in populations of cells depleted of cells expressing SSEA4 using magnetic beads. Fold expression is shown relative to undifferentiated Hi embryonic stem cells. 1038] Figure 13 shows the expression of OCT4 (panel a), NANOG (panel b), SOX2 (panel c), and goosecoid (panel d), as detected via real-time PCR, in populations of cells depleted of cells expressing SSEA4 using FACS. Fold expression is shown relative to undifferentiated HIl embryonic stem cells. DETAIL LED DESCRJ PTION 10391 For clarity of disclosure, and not by way of limitation, the detailed description of the invention is divided into the following subsections that describe or illustrate certain features, embodiments or applications of the present invention. Definitions 0040] "Q-cell lineage" refers to cells with positive gene expression for the transcription factor PDX-1 and at least one of the following transcription factors: NGN3. NKX22, NKX6, 1, NEUROD, ISLI, HNF-3 beta, MAFA, PAX4, and PAX6, Cells expressing markers characteristic of the [i cell lineage include fi cells. 0041] "Cells expressing markers characteristic of the definitive endoderm lineage" as used herein refers to cells expressing at least one of the following markers: SOX 17, 10 GATA4, fHNF-3 beta, GSC, CERI, Nodal, FGF8, Brachyury, Mix-like homeobox protein, FGF4, CD48, comesodermin (EOMES), DKK4, FGF 17, GATA6, CD 184, C Kit, CD99, or 0TX2. Cells expressing markers characteristic of the definitive endodemi lineage include primitive streak precursor cells, primitive streak cells, mesendoderm cells and definitive endodermr cells, 0421 "Cells expressing markers characteristic of the primitive gut tube lineage" refers to cells expressing at least one of the -following markers: HNF-l beta, or H{NF-4 alpha, 043j "Cells expressing mark-ers characteristic of the pancreatic endoderm lineage" as used herein refens to cells expressing at least one of the following markers: IPDX 1, HNF- I beta, PTF-1 alpha, U NF6, or H9. Cells expressing markers characteristic of the pancreatic endodemi lineage include pancreatic endoderm cells. 0441 "Cells expressing markers characteristic of the pancreatic endocrine lineage" as used herein refers to cells expressing at least one of the following markers: NGN3, NEUROD, ISL, PDXI, NKX6.1, PAX4, NGN3, or PTF-l alpha. Cells expressing markers characteristic of the pancreatic endocrine lineage include pancreatic endocrine cells, pancreatic hormone expressing cells, and pancreatic hormone secreting cells, and cells of the f-cell lineage, 0451 "Definitive endoderm" as used herein refers to cells which bear the characteristics of cells arising from the epiblast during gastrulation and which form the gastrointestinal tract and its derivatives. Definitive endodern cells express the following markers: CD184, HNF-3 beta, GATA4, SOX 17, Cerberus, OTX2, goosecoid, c-Kit, (D99, and Mixil. 00461 "Markers" as used herein, are nucleic acid or polypeptide molecules that are differentially expressed in a cell of interest, In this context, differential expression means an increased level for a positive marker and a decreased level for a negative marker. The detectable level of the marker nucleic acid or polypeptide is sufciently h igher or lower in the cells of interest compared to other cells, such that the cell of interest can be identified and distinguished from other cells using any of a variety of methods known in the art. I1 0471 "Pancreatic endocrine cell" or "pancreatic hormone expressing cell" as used herein refers to a cell capable of expressing at least one of the following hormones: insulin, glucagon, somatostatin, and pancreatic polypeptide. 48] "Pancreatic hormone secreting cell" as used herein refers to a cell capable of secreting at least one of the following hormones: insulin, glucagon, sonatostatin, and pancreatic polypeptide. 049J "Pre-primitive streak cell" as used herein refers to a cell expressing at least one of the following markers: Nodal., or FOF8, 050] "Primitive streak cell" as used herein refers to a cell expressing at least one of the following markers: Brachyury, Mix-like homeobox protein, or FGF4, I51] Stem cells are undifferentiated cells defined by their ability at the single cell level to both self-renew and differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terinally differentiated cells. Stem cells are also characterized by their ability to differentiate in tro into functional cells of various cell lineages from multiple germ layers endodermm, mesoderm and ectoderm), as well as to give rise to tissues of muip7le germ layers following transplantation and to contribute substantially to most, if not all, tissues following injection into blastocysts, 0521 Stem cells are classified by their developmental potential as: (i) totipotent, meaning able to give rise to all embryonic and extraembryonic cell types; (ii) pluripotent. meaning able to give rise to all embryonic cell types; (iii) multipotent, meaning able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, henatopoictic stern cells (HSC can produce progeny that include HSC (self- renewal), blood cell restricted oligopotent progenitors and all cell types and elements (e.g. platelets) that are normal components of the blood.); (iv) oligopotent, rneaning able to give rise to a more restricted subset of cell lineages than multipotent stern cells; and (v) unipotent, meaning able to give rise to a single cell lineage (e.g. spermatogen icstem cells), 00531 Differentiation is the process by which an unspecialized ("uncommitted") or less specialized cell acquires the features of a specialized cell such as, for example, a nerve 12 cell or a muscle cell A differentiated or differentiation-induced cell is one that has taken on a more specialized ("committed") position within the lineage ofa cell. The term committed", when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathvay to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type. Dedifferentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell As used herein, the lineage of a cell defines the heredity of the cell, that is, which cells it caie from and what cells it can give rise to. The lineage of a cell places the cell within a hereditary scheme of development and differentiation, A lineage-specific marker refers to a characteristic specifically associated with the phenotype of cells of a lineage of interest and can be used to assess the differentiation of an uncommitted cell to the lineage of interest. 054] Various terms are used to describe cells in culture. "Maintenance" refers generally to cells placed in a growth medium under conditions that facilitate cell growth and/or division that may or may not result in a larger population of the cells. "Passaging" refers to the process of removing the cells from one culture vessel a:nd placing them in a second culture vessel under conditions that faciitate cell growth and/or division. 055] A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a Pl0 culture. The primary culture, that is, the finst culture following the isolation of cells from tissue, is designated PO. Following the first subculture, the cells are described as a secondary culture (P1 or passage 1). After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will he understood by those of skill in the art that there may be many population doubling during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number. The expansion of cells (that is, the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and time between passaging, 13 Eurichment of Cells Expressing Markers Characteristic of the Pancreatic Endocrine Lineage 056] In one embodiment, the present invention provides a method to different iate a population of pluripotent stem cells into a population of cells expressing markers characteristic of the pancreatic endocrine lineage, comprising the steps of: a. Culturing a population of phripotent stem cells, b. Differentiating the population of pluripotent stem cells into a population of cells expressing markers characteristic of the definitive endoderi lineage, c. Differentiating the population of cells expressing markers characteristic of the definitive endodern lineage into cells expressing markers characteristic of the primitive gut tube lineage, d. Differentiating the population of cells expressing markers characteristic of the priitive gut tube lineage into a population of cells expressing markers characteristic of the pancreatic endoderm lineage, and e. Differentiating the population of cells expressing markers characteristic of the pancreatic endoderm lineage into a population cells expressing markers characteristic of the pancreatic endocrine lineage, 1057] In one embodiment. the population of cells expressing markers characteristic of the pancreatic etndocrine lineage is transplanted into an animal, wherein the cells expressing markers characteristic of the pancreatic endocrine lineage form insulin producing cells. In one embodiment, the efficiency of the formation of insulin producing cells is enhanced by enriching the population for cells expressing markers characteristic of the pancreatic endocrine lineage prior to transplantation. 0058] In one embodiment, the efficiency of the formation of insulin producing cells is determined by measuring the time taken for the expression of C-peptide to reach detectable levels following transplantation. 0059] In an altemate embodiment, the enrichment decreases the ability of the transplanted cells to form teratomas following transplantation. 14 0601 Cells expressing markers of the pancreatic endocrine lineage are identified or selected through the binding of antigens, found on the surfaces of the cells, to reagents that specifically bind the cell surface antigen, 10611 In an alternate embodiment, cells expressing markers characteristic of the pancreatic endocrine lineage are furdier differeItiated into insulin producing cells, prior to transplantation into an animal, Insulin producing cells are identified or selected through the binding of antigens, found on the surfaces of the cells, to reagents that specifically bind the cell surface antigen. 1062] In an alternate embodiment, the present invention provides a method to differentiate a population of pluripotent stem cells into a population of cells expressing markers characteristic of the pancreatic endocrine lineage, comprising the steps of: a, Culturing a population of pluripotent stem cells, b. Differentiating the population of pluripotent stem cells into a population of cells expressing markers characteristic of the definitive endoderm lineage, c. Differentiating the population of cells expressing markers characteristic of the definitive endodern lineage into cells expressing markers characteristic of the primitive gut tube lineage, d. Enriching the population of cells that express markers characteristic of the primitive gut tube lineage, e. Differentiating the population of cells expressing markers characteristic of the primitive gut tube lineage into a population of cells expressing markers characteristic of the pancreatic endoderm lineage, and f Differentiating the population of cells expressing markers characteristic of the pancreatic endoderni lineage into a population cells expressing markers characteristic of the pancreatic endocrine lineage. 0063] In one embodiment, the population of Cells expressing markers characteristic of the pancreatic endocrine lineage is transplanted into an animal, wherein the cells expressing markers characteristic of the pancreatic endocrine lineage form insulin 15 producing cells. In one embodiment, the efficiency of the formation of insulin producing cells is enhanced by enriching the population of cells that express markers characteristic of the primitive gut tube lineage prior to transplantation. 1064] Cells expressing markers of the primitive gut tube lineage are identified or selected through the binding of antigens, found on the surfaces of the cells, to reagents that specifically bind the cell surface antigen. Surface Antigens that Faciliawe Enrichment of Cells Expressing Markers Characteristic of the Pancreatic Endocrine Lineagc 1065] In one embodiment, prior to transplantation into an animal, the population of cells expressing markers characteristic of the Pancreatic endocrine lineage is treated with at least one reagent that is capable of binding to a marker selected from the group consisting of CD9, CD13, CDI5, CD47, CD$56,CD73, CDI 17, CDI33, CDI84, CD200, CD3 .8, CD326 and SSEA4. 10661 In one embodiment, treatment with the at least one reagent results in a population of cells expressing markers characteristic of the pancreatic endocrine lineage that are positive for the expression of the marker CD56 and negative for the expression of the marker CD13. 1067] In one embodiment, treatment with the at least one reagent results in a population of cells expressing markers characteristic of the pancreatic endocrine lineage that are positive for the expression of the marker CD56 and negative for the expression of the marker CD 15. 0068] In one embodiment, treatment with the at least one reagent results in a population of cells expressing markers characteristic of the pancreatic endocrine lineage that are negative for the expression of the marker CD1 33. 00691 In one embodiment, treatment with the at least one reagent results in a population of cells expressing markers characteristic of the pancreatic endocrine lineage that are negative for the expression of the marker CD 15. 16 070] In one embodiment, treatment with the at least one reagent results in a population of cells expressing markers characteristic of the pancreatic endocrine lineage that are positive for the expression of the marker CD 184. 1071] In one embodiment, treatment with the at least one reagent results in a population of cells expressing markers characteristic of the pancreatic endocrine lineage that are negative for the expression of the marker SSEA4. Snrface Antigens that Facilitate Enrichment of Inuln Producing Cells 072] In one embodiment, prior to transplantation into an animal, the population of cells expressing markers characteristic of the pancreatic endocrine lineage is further differentiated into a population of insulin producing cells. The population of insulin producing cells is treated with at least one reagent that is capable of binding to a marker selected from the group consisting of CD47, CD56, CD57 CD98 and SSEA4, 1073] In one embodiment, treatment with the at least one reagent resuhs in a population of insulin producing cells that are positive for the expression of the marker CD56 and CD57. Alternatively, the population of insulin producing cells may be positive for the expression of CD98. Alternatively, the population of insulin producing cells may be negative for the expression of CD47. 1074] In one embodiment, treatment with the at least one reagent results in a population of insulin producing cells that are negative for the expression of the marker SSEA4. d075j CD 13 is expressed on the majority of peripheral blood monocytes and granulocytes. It is also expressed by the majority of acute mycloid leukemias, chronic mycloid leukemias in myeloid blast crisis, a smaller percentage of lymphoid leukemias and myeloid cell lines. CD13 is also found in several types of non hematopoietic cells such as fibroblasts and endothelial cells and in a soluble form in blood plasma. CD13 is not expressed on B cells, T cells, platelets or erythrocytes. CD 13 plays a role in biologically active peptide metabolism, in the control of growth and diftfrentiation, in phagocytosis and in bactericidaltmoricidal activities. CD13 also serves as a receptor for human coronaviruses (HCV). 17 076] CD15 is a carbohydrate adhesion molecule that can be expressed on glycoproteins, glycolipids and proteoglycans. CD15 mediates phagocytosis and chemotaxis, fuid on neutrophils; expressed in patients with Hodgkin disease, some B-cell chronic lymphoeyie leukemias, acute lymphoblastic leukemias, and most acute nonlymphocytic leukemias. It is also called Lewis x and SSEA-l (stage specific embryonic antigen I) and represents a marker for marine pluripotent stem cells, in which it plays an important role in. adhesion and migration of the cells in the preimplantation embryo. 1077j CD47 is a membrane protein, which is involved in the increase in intracellular calcium concentration that occurs upon cell adhesion to extracellular matrix. The protein is also a receptor for the C-terminal cell binding domain of thrombospondin, and it may play a role in membrane transport and signal transduction. 1078] CD56, also known as Neural Cell Adhesion Molecule (NCAM) is a homophilic binding glycoprotein expressed on the surface of neurons, glia, skeletal muscle and natural killer cells. NCA]M has been implicated as having a role in cell-cell adhesion, neurite outgrowth, synaptic plasticity, and leading and memory. 079] CD57 also known as HNK- I or Leau 7 , is an antigenic oligosaccharide moiety detected on extracellular proteins of certain cell types. In blood, CD57 is found on 1 5---20% of mononuclear cells, including subsets of NK and T cells, though not on erythrocytes, monocytes, granulocytes, or platelets. Also, CD57 expression can be found on a variety of neural cell types. 0080] CD98 is a glycoprotein that comprises the light subunit of the Large neutral Amino acid Transporter (LAT I). LATI is a heterodimeric membrane transport protein that preferentially transports neutral branched (valine, leucine, isoleucine) and aromatic (tryptophan, tyrosine) amino acids. 0081] CD133 is a glycoprotein also known in humans and rodents as Prorninin I (PROM1). It is a member of pentaspan transmembrane glycoproteins (5-transmembrane, 5-TM), which specifically localizes to cellular protnisions. CD133 is expressed in hematopoictic stem cells, endothelial progenitor cells, glioblastomas, neuronal and 18 glial stem cells. See Corbeil ef ai, Biochem Biophvs Res Counun 285 (4): 939-44, 2001. doi 10.1 006/bbrc.200 1.5271 PM ID 11467842. SwfiAce Atigens that Faciitate Enrichment a/e/s Expressing Markers Characteristic ofthe Prifmtive Gut Tube Lineage 10821 In an alternate embodiment, the present invention provides a method to differentiate a population of pluripotent stern cells into a population of cells expressing markers characteristic of the pancreatic endocrine lineage, comprising the steps of: a. Culuring a population of phuripotent ste cells, b. Differentiating the population of pluripotent stem cells into a population of cells expressing markers characteristic of the definitive endoderm lineage, c. Differentiating the population of cells expressing markers characteristic of the definitive endoderm lineage into cells expressing markers characteristic of the primitive gut tribe lineage, d. Enriching the population of cells that express markers characteristic of the primitive gut tube lineage, c. Differentiating the population of cells expressing markers characteristic of the primitive gut tube lineage into a population of cells expressing markers characteristic of the pancreatic endodern lineage, and E Differentiating the population of cells expressing markers characteristic of the pancreatic endoderm lineage into a population cells expressing markers characteristic of tire pancreatic endocrine lineage. 0083j I one embodiment, the population of cells expressing markers characteristic of the pancreatic endocrine lineage is transplanted into an animal, wherein the cells expressing markers characteristic of the pancreatic endocrine lineage form insulin producing cells. In one embodiment, the efficiency of the formation of insulin producing cells is enhanced by enriching the population of cells that express markers characteristic of the primitive gut tube lineage prior to transplantation, 19 *084] The population of cells that express markers characteristic of the primitive gut tube lineage is treated with at least one reagent that is capable of binding to the LIF receptor, 1085] The cells expressing markers characteristic of the pancreatic endocrine lineage, cells expressing markers characteristic of the primitive gut tube lineage, or insulin producing cells may be enriched., depleted, isolated, separated, sorted and/or purified as further described in the examples, As used herein, the terms "enriched" or "purified" or enriched or purified due to depletion of other known cell populations, indicate that the cells has been subject to some selection process so that the population is enriched. andior purified. Also, the subject cells are also considered relatively enriched and/or purified, i.e. there is significantly more of a particular differentiated cell population as compared to another cell population, or as compared to pluripotent stem cells before "enrichment" or "purification" or as compared to the original or initial cell culture. 1086j Enriching or purifying for a given differentiated cell type may involve "depleting" or "separating" or "sorting" one or more known cell types from another cell type. in one embodiment, a population of cells may be purified by depleting an unwanted differentiated cell type. It may be advantageous to enrich and. purify a cell expressing markers characteristic of the pancreatic endocrine lineage by depleting the culture of known or unknown cell types, In this way, the enriched or purified cell population would not have the bound or attached antibody. Because there is no need to remove the antibody from the purified population, the use of the enriched or purified cells for cell therapies may be improved 00871 Methods -for enriching, depleting, isolating, separating, sorting and/or purifying may include, for example, selective culture conditions, wherein the culture conditions are detrimental to any undesirable cell types. 0088] Methods for enriching, depleting, isolating, separating, sorting and/or purifying may also include, for example, antibody-coated magnetic beads, affinity chromatography and "panning" with antibody attached to a solid matrix or solid phase capture medium, e.g. plate, column or other convenient and available technique, Techniques 20 providing accurate separation include flow cytometry methods which are useful for measuring cell surface and intracellular parameters, as well as shape change and granularity and for analyses of beads used as antibody- or probe-linked reagents. Readouts from flow cytometry assays include, but are not limited to, the mean fluorescence associated with individual fluorescent antibody-detected cell surface molecules or cytokines, or the average fluorescence intensity, the median fluorescence intensity, the variance in fluorescence intensity, or some relationship among these, i089] In some aspects of embodiments with analytical steps involving flow cytometry, minimal parameters or characteristics of the beads are scatter (FS and/or SS) and at least one fluorescent wavelengths. Flow cytometry can be used to quantitare parameters such as the presence of cell surface proteins or conformational or posttranslational modification thereof; intracellular or secreted protein, where permeabilization allows antibody (or probe) access, and. the like. Flow cytometry methods are known in the art, and described in the following: Flow Cytometry and Cell Storing (Springer Lab Manual), Radbruch, Ed., Springer Verlag, 2000; Ormerod, Flow Cytometry, Springer Verlag, 1999; Flow Cytometry Protocols (Methods in Molecular Biology , No 91), Jaroszeski and He]ler, Es., Humana Press, 1998; Current Protocols in Cytometry, Robinson et at, eds, John Wiley & Sons, New York, N.Y, 2000. 1090j The staining intensity of cells may be monitored by flow cytometry, where lasers detect the quantitative levels of fluorochrome (which is proportional to the amount of cell surface marker bound by specific reagents, e.g. antibodies). Flow cytometry, or FACS, may also be used to separate cell populations based on the intensity of binding to a specific reagent, as well as other parameters such as cell size and light scatter. Although the absolute level of staining can differ with a particular fluorochrome and reagent preparation, the data can be normalized to a control, In order to nornnalize the distribution to a control, each cell is recorded as a data point having a particular intensity of staining, 00911 In order to nornalize the distribution to a control, each cell is recorded as a data point having a particular intensity of staining, These data points may be displayed according to a log scale, where the unit of measure is arbitrary staining intensity. In 21 one example, the brightest cells in a population are designated as 4 logs more intense than the cells havingt the lowest level of staining When displayed in this manner, it is clear that the cells falling in the highest log of staining intensity are bright, while those in the lowest intensity are negative, The "low" staining cells, which fall in the 2-3 log of staining intensity, may have properties that are unique from the negative and positive cells. An alternative control may utilize a substrate having a defined density of marker on its surface, for example a fabricated bead or cell line, which provides the positive control for intensity. The "low" designation indicates that the level of staining is above the brightness of an isotope matched control, but is not as intense as the most brightly staining cells normally found in the population. 1092] The readouts of selected parameters are capable of being read simultaneously, or in sequence during a single analysis, as for example through the use of fluorescent antibodies to cell surface molecules, As an example, these can be tagged with different fluorochromes, fluorescent bead, tags, e.g, quanturn dots, etc., allowing analysis of up to 4 or more fluorescent colors simultaneously by flow cytometry. For example, a negative designation indicates that the level of staining is at or below the brightness of an isotype matched negative control; whereas a dim designation indicates that the level of staining can be near the level of a negative stain, but can also be brighter than an isotope matched control. 1093j Identifiers of individual cells, for example different cell types or cell type variants, may be fluorescent, as for example labeling of different unit cell types with different levels of a fluowscent compound, and the like as described herein above. In sone aspects of embodiments where two cell types are to be mixed, one is labeled and the other not. In some aspects of embodiments where three or more cell types are to be included, each cell type may labeled to different levels of fluorescence by incubation with different concentrations of a labeling compound, or for different times, As identifiers of harge numbers of cells, a matrix of fluorescence labeling intensities of two or more different fluorescent colors may be used, such that the number of distinct unit cell types that are identified is a number of fluorescent levels of one color, e.g., carboxyfluorescein succinimidyl ester (CTSE), times the number of fluorescence levels employed of the second color, e.g, tetramethy Ithodaine isothiocyanate (TRITC), or the like, times the number of levels of a third color, etc. Alternatively, 22 intrinsic light scattering properties of the different cell types, or characteristics of the BioMAPs of the test paranwters included in the analysis. may be used in addition to or in place of fluorescent labels as unit cell type identifiers. 1094] In another aspect, cells may be enriched, depleted, separated, sorted and/or purified using conventional affinity or antibody techniques. For example, the 1 igand and/or antibody may be conjugated with labels to allow for ease of separation of the particular cell type, e.g, magnetic beads; biotin, which binds with high affinity to avidin or streptavidin; fluorochromes, which can be used with a fluorescence activated cell sorter; haptens; and the like., 10951 In one embodiment, the ligand, agent, and/or antibodies described herein may be directly or indirectly conjugated to a magnetic reagent, such as a super-paramagnetic microparticle (microparticle), Direct conjugation to a magnetic particle may be achieved by use of various chemical linking groups, as known in the art. In some embodiments,the antibody is coupled to the microparticles through side chain amino or sufhydryl groups and heterofunctional cross-linking reagents. 1096] A large number of heterofunctional compounds are available for linking to entities, For example, at least, 3 (2-pyridyidithio)propionic acid N-hydroxysuccini mide ester (SPDP) or 4-(N-maleimidomethyl)-cyclohexanet--carboxylic acid N hydroxysuccini mide ester (SMC C) with a reactive sulihydryl group on the antibody and a reactive amino group on the magnetic particle can be used, An example of a magnetic separation device is described in WO 90/07380, PCTIUS96/00953, and EP 438,520, incorporated herein by reference in its entirety. 0097] The purified cell population may be collected in any appropriate medium., Suitable media may include, for example., Dulbecco's Modified Eagle Medium (dMEM), Ranks Basic Salt Solution (HBSS), Dulbecco's phosphate buffered saline (dPBS), RPMI, Iscove's modified Dulbecco's medium (IMDM), phosphate buffered saline (PBS) with. 5 mM EDTA., etc., frequently supplemented with fetal calf serum (FCS), bovine serum albumin (BSA), human serum albumin (lHSA), and StemProhESC SFM 23 098] In one embodiment, the cells expressing markers characteristic of the pancreatic endocrine lineage are enriched by treatment with at least one agent that selects cells that do not express markers characteristic of the pancreatic endocrine lineage, In an alternate embodiment, the cells expressing markers characteristic of the pancreatic endocrine lineage are enriched by treatment with at least one agent that selects for insulin-producing cells, 0999] Using the methods described herein, cell populations or cell cultures may be enriched in cell content by at least about 2- to about 1000-fold as compared to untreated cell populations or cell cultures. In some embodiments, cells expressing markers characteristic of the pancreatic endocrine lineage may be enriched by at least about 5 to about 500-fold as compared to untreated cell populations or cell cultures. In other embodiments, cells expressing markers characteristic of the pancreatic endocrine lineage may be enriched from at least about 10- to about 200-fold as compared to untreated cell populations or cell cultures. In still other embodiments, cells expressing markers characteristic of the pancreatic endocrine lineage may be enriched. from at least about 20- to about 100-fold as compared to untreated cell populations or cell cultures. In yet other embodiments, cells expressing markers characteristic of the pancreatic endocrine lineage may be enriched from at least about 40- to about 80-fold as compared to untreated cell populations or cell cultures, In certain embodiments, cells expressing markers characteristic of the pancreatic endocrine lineage may be enriched from at least about 2- to about 20-fold as compared to untreated cell Populations or cell cultures. Characterization of Cells Derived from Pluripotent Stem Cells 01001 The formation of differentiated cells from pluripotent stem cells may be determined by determining the expression of markers characteristic of a given differentiated cell type. In some embodiments, the identification and characterization of a differentiated cell is by expression of a certain marker or different expression levels and patterns of more than one marker, 0101j Specifically, the presence or absence, the high or low expression, of one or more the marker(s) can typify and identify a cell-type. Also, certain markers may have 24 transient expression, whereby the marker is highly expressed during one stage of development and poorly expressed in another stage of development. The expression of certain markers can be determined by measuring the level at which the marker is present in the cells of the cell culture or cell population as compared to a standardized. or normalized control marker. In such processes, the measurement of marker expression can be qualitative or quantitative. One method of quantitating the expression of markers that are produced by marker genes is through the use of quantitative PCR (Q-PCR). Methods of performing Q-PCR are well known in the art. Other methods which are known in the art can also be used to quantitate marker gene expression. For example, the expression of a marker gene product can be detected by using antibodies specific for the marker gene product of interest (e.g. Western blot, flow cytometry analysis, and the like). In certain embodiments, the expression of marker genes characteristic of differentiated cells as well as the lack of significant expression of marker genes characteristic of differentiated cells may be determined. 1102] The expression of tissue-specific gene products can also be detected at the mRNA level by Northem blot analysis, dot-blot hybridization analysis, or by reverse transcriptase initiated polymerase chain reaction (RT-PCR) using sequence-specific primers in standard amplification methods. See U.S. Pat, No. 5,843,780 for further details. Sequence data for particular markers listed in this disclosure can be obtained from public databases such as GenBank. 1103] Pluripotent stem cells may express one or more of the stage-specific embryonic antigens (SSEA) 3 and 4, and markers detectable using antibodies designated Tra-i 60 and Tra-1-81 (Thomson e a, Science 282: 1145, 1998). Differentiation of pluripotent stem cells in vitro results in the loss of SSEA-4, Tra 1-60, and Tra 1-81 expression (if present) and increased expression of SSEA-1. Undifferentiated pluripotent stem cells typically have alkaline phosphatase activity, which can be detected by fixing the cells with 4% paraformaldehyde, and then developing with Vector Red as a substrate, as described by the manufacturer (Vector Laboratories, Butrlingame Calit). Undifferentiated pluripotent stem cells also typically express OCT4 and TERT, as detected by RT-PCR. 25 11041 Markers characteristic of the pancreatic endoderm lineage are selected from the group consisting of PDXMI, HNFI beta, PTF I alpha, IHNF6, HB9 and PROX 1. Suitable for use in the present invention is a cell that expresses at least one of the markers characteristic of the pancreatic endoderm lineage, In one aspect of the present invention, a cell expressing markers characteristic of the pancreatic endoderi lineage is a pancreatic endoderm cell. 105] Markers characteristic of the definitive endodern lineage are selected from the group consisting of SOX17, GATA4 HNF3 beta, GSC, CIER, Nodal, FGF8, Brachyury, Mix-like homeobox protein, FGF4, CD48, comesodermin (EOMES), DKK4, FGF 17, GATA6, CD-184, C-Kit, CD99, and 0TX2, Suitable for use in the present invention is a cell that expresses at least one of the markers characteristic of the definitive endoderm lineage, in one aspect of the present invention, a cell expressing markers characteristic of the definitive endoderm lineage is a primitive streak precursor cell. In an alternate aspect, a cell expressing markers characteristic of the definitive endoderm lineage is a mesendoderm cell. In an alternate aspect, a cell expressing markers characteristic of the defmnitive endodenr lineage is a defintive endoderm cell. i106] Markers characteristic of the pancreatic endocrine lineage are selected from the group consisting of NGN3, NEUROD, ISLI, PDXI, NKX6.1, PAX4, NGN3, and PTF-I alpha. in one embodiment, a pancreatic endocrine cell is capable of expressing at least one of the following hormones: insulin, glucagon, somatostatin, and pancreatic polypeptide. Suitable for use in the present invention is a cell that expresses at least one of the markers characteristic of the pancreatic endocrine lineage. In one aspect of the present invention, a cell expressing markers characteristic of the pancreatic endocrine lineage is a pancreatic endocrine cell. The pancreatic endocrine cell may be a pancreatic hormone-expressing cell. Alternatively, the pancreatic endocrine cell may be a pancreatic hormone-secreting cell 01071 In one aspect of the present invention, the pancreatic endocrine cell is a cell expressing markers characteristic of the p, cell lineage, A cell expressing markers characteristic of the P cell lineage expresses PDX1 and at least one of the following transcription factors: NGN3. NKX2,2, NKX6.1, NEUROD, ISL1, HNF3 beta, 26 MAFA, PAX4, and PAX6. MI one aspect of the present invention, a cell expressing markers characteristic of the P cell lineage is a 0 cell Pluripotent Stern Cells Characterization ofPluripoent Stem Cells $108] Pluripotent stem cells may express one or more of the stage-specific embryonic antigens (SSEA:) 3 and 4, and markers detectabl using antibodies designated Tra- 60 and Tra-1-81 (Thomson ei ed,, Science 282 1145 1998). Differentiation of pluripotent stem cells in vitro results in the loss of SSEA-4, Tra- I-60, and Tra-l-sI expression (if present) and increased expression of SSEA-1. Undifferentiated pluripotent stem cells typically have alkaline phosphatase activity, which can be detected by fixing the cells with 4% parafonnaldehyde and then developing with Vector Red as a substrate, as described by the manufacturer (Vector Laboratories, Burlingame Calif.). Undifferentiated pluripotent stem cells also typically express Oct-4 and TERT, as detected by RT-PCR, 109] Another desirable phenotype of propagated pluripotent stem cells is a potential to differentiate into cells of all three germinal layers: endoderm, mesoderm, and ectodern tissues. Pluripotency of stem cells can be confined, for example, by injecting cells into severe combined immunodeficient (SCID) mice, fixing the teratomas that fom using 4' paraformaldehyde, and then examining them histologically for evidnce of celt types from the three germ layers. Alteatively, pluripotency may be determined by the creation of embryoid bodies and assessing the embryoid bodies for the presence of markers associated with the three germinal layers. 0110] Propagated phuripotent stem cell lines may be karyotyped using a standard G-banding technique and compared to published karyotypes of the corresponding primate species, It is desirable to obtain cells that have a "normal karyoTpe," which means that the cells are euploid, wherein all human chromosomes are present and not noticeably altered. Sources of Pluripotent Stem Cells 27 111] The types of pluripotent sten cells that may be used include established lines of pluripotent cells derived from tissue formed after gestation, including pre-embryonic tissue (such as, for example, a blastocyst), embryonic tissue, or fetal tissue taken any time during gestation, typically but not necessarily before approximately 10-12 weeks gestation. Non-limiting examples are established lines of human embryonic stem cells or human enibryonic germ cells, such as, for example the human embryonic stem cell lines HI, H7, and Ff9 (WiCell). Also contemplated is use of the compositions of this disclosure during the initial establishment or stabilization of such cells, in which case the source cells would be primary pluripotent cells taken directly from the source tissues. Also suitable are cells taken from a pluripotent stem cell population already cultured in the absence of feeder cells, as well as a pluripotent stem cell population already cultured in the presence of feeder cells. Also suitable are mutant human embryonic stem cell lines, such as, for example, BG0lv (BresaGen, Athens, GA). Also suitable are cells derived from adult human somatic cells, such as, for examples, cells disclosed in Takahashi et al, Cell 131: 1-12 (2007). 1112] In one embodiment, human embryonic stem cells are prepared as described by Thomson et al. (U.S& Pat No, 5,843,780; Science 282:1145, 1998; Curr. Top. Dev. Biol. 38:133 ff, 1998: Proc. Natl. Acad. Sci. U.SA. 92:7844, 1995), 1,131 Also contemplated, are pluripotent stem cells that are derived from somatic cells. In one embodiment, pluripotent stem cells suitable fbr use in the present invention may be derived according to the methods described in Takahashi et al (Cell 126: 663-676. 2006). 0114] In an alternate embodiment, pluripotent stem cells suitable for use in the present invention may be derived according to the methods described in Li et al(Cell Stem Cell 4: 16-19, 2009) 0115] In an alternate embodiment, pluripotent stem cells suitable for use in the present invention may be derived according to the methods described in Maherali et a! (Cell Stem Cell 1: 55-70,2007), 28 1116] In an alternate embodiment, pluripotent stem cells suitable for use in the present invention may be derived according to the methods described in Stadtfeld ei al (Cell Stem Cell 2: 230-240). 1117] In an ahernate embodiment, pluripotent stem cells suitable for use in the present invention may be derived according to the methods described in Nakagawa el al (Nature Biotechnology 26: 101-106, 2008). i118] In an alternate embodiment, pluripotent stem cells suitable for use in the present invention may be derived according to the methods described in Takahashi t t l (Cell 131: 861-872, 2007), 1,191 In an alternate embodiment, pluripotent stem cells suitable for use in the present invention may be derived according to the methods described in US patent application Ser. No. 61/256,149, assigned to Centocor R&D, Inc, Culture ofPhripoient Stem Cells 1120] In one embodiment, pluripotent stem cells are cultured on a layer of feeder cells or extracellular matrix protein that support the pluripotent stem cells in various wax's, prior to cahuring according to the methods of the present invention. For example, pluripotent stem cells are cultured on a feeder cell layer that supports proliferation of pluripotent stem cells without undergoing substantial differentiation. The growth of pluripotent stem cells on a feeder cell layer without differentiation is supported using (i) Obtaining a culture vessel containing a feeder cell layer; and (ii) a medium conditioned by culturing previously with another cell type, or a non-conditioned mediumn, for example, free of sermn or even chemically defined. 0121] In another example, pluripotent stem cells are cultured in a culture system that is essentially free of feeder cells, but nonetheless supports proliferation of pluripotent stem cells without undergoing substantial differentiation. The growth of pluripotent stem cells in feeder-cell free culture without differentiation is supported using (i) an adlayer on a solid substrate surface with one or more extracellular matrix proteins; and (ii) a medium conditioned by culturing previously with another cell type, or a non-conditioned medium, for example, free of serum or even chemically defined. 29 11221 In an alternate embodiment, pluripotent stem cells are cultured on a surface modified plate containing from at least about 0.5% N, a sum of 0 and N of greater than or equal to 17,2% and a contact angle of at least about 13,9 degrees in a medium conditioned by culturing previously with another cell type, or a non-conditioned medium, for example, free of serum or even chemically defined, 123] Culture medium: An example of cell culture medium suitable for use in the present invention may be found in US20020072117. Another example of cell culture medium suitable for use in the present invention may be found in US6642048. Another example of cell culture medium suitable for use in the present invention may be found in W02005014799. Another example of cell culture medium suitable for use in the present invention may be found in Xu et al (Stem Cells 22: 972-980, 2004), Another example of cell culture medium suitable for use in the present invention may be found in US20070010011. Another example of cell culture medium suitable for use in the present invention may be found in Cheon et alt (BioReprod D01:10.1 095/biolreprod 105,046870; 19 Oct 2005). Another example of cell culture medium suitable for use in the present Invention may be found in Levenstein et al. (Stem Cells 24: 568-574, 2006). Another example of cell culture medium suitable for use in the present invention may be found in US20050148070. Another example of cell culture medium suitable for use in the present invention may be found in US20050233446. Another example of cell culture medium suitable for use in the present invention may be found in US6800480. Another example of cell culture medium suitable for use in the present invention may be found in US20050244962, Another example of cell culture medium suitable for use in the present invention may be found in W02005065354, Another example of cell culture medium suitable for use in the present invention may be found in W02005086845. 0124] Suitable culture media may also be made from the following components, such as, for example, Dulbecco's modified Eagle's medium (DMEM), G ibco #' 11965-092; Knockout:Dulbecco's modified Eagle's medium (KO DMEM), Gibco # 10829-018; Ham's F1 2/50% DMEM basal medi um; 200 mM L-glutamine, Gibco # 15039-027; non-essential amino acid solution, Gibco 11140-050; fi-mercaptoefhanol, Sigma #t M7522; human recombinant basic fibroblast growth factor (bFGF), Gibco # 13256 029. 30 Dithrentirin olPluripolent Sten Cells 1125] In one embodiment, pluripotent stem cells are propagated in culture and then treated in a manner that promotes their ditTerentiation into another cell type. For example, pluripotent stem cells formed using the methods of the present invention may be differeniated into neural progenitors or cardioniyocytes according to the methods disclosed in W02007030870. i126] In another example, pluripotent stem cells formed using the methods of the present invention may be differentiated into hepatocytes according to the methods disclosed in US patent 6,458,589. .Dilerentiation ofhurpotent Stem Cells Formed UCsing the Akthods ofthe Present invention into C(ells Expressing Markers Charaeristic of the Definitive Endoderm Lineage 1127] Piuripotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage by any method in the at 1128] For example, phripotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in D Amour et al, Nature Biotechnology 23, 1534 - 1541 (2005). 4129] For example,. pluripotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in Shinozaki et a, Development 131, 1651 ~.1662 (2004). 0130] For example, plurinpotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in McLean et al, Stem Cells 25, 29 - 38 (2007).
11311 For example, piuripotent stern cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in D'Amour et ail, Nature Biotechnology 24, 1392 - 1401 (2006), 11321 In another example, pluripotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in US patent application Ser. No. 11/736,908, assigned to LifeScan, Inc. 1133] In another example, pluripotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in US patent application Ser. No. 11 779,311, assigned to LifeScan, Inc, 1134] In another example, phu-ipotent stern cells fonned using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in US patent application Ser. No, 121493,741, assigned to LifeScan. Inc, 1135] Ii another example, pluripotent stem cells formed using the methods of the present invention may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage according to the methods disclosed in US patent application Ser. No, 12/494,789, assigned to LifeScan, Inc. 01361 Formation of cells expressing markers characteristic of the definitive endoderm lineage may be determined by testing for the presence of the markers before and after following a particular protocol. Phripotent stem cells typically do not express such markers, Thus, differentiation of pluripotent cells is detected when cells begin to express them, Diff'rentiation ofPhuripotent Stem Cells Formed Using the eAthods ofthe Present invention into Cells Expressing arkers Characteristic ofthe Panrreatic Endoderm Lineage 3 2 1371 Phiripotent stem cells fbmied using the methods of the present invention may be differentiated into cells expressing markers characteristic of the pancreatic endoderm lineage by any method in the art. 1138] For example, pluripotent stem cells may be differentiated into cells expressing markers characteristic of the pancreatic endoderm lineage according to the methods disclosed in D'Amour eta, Nature Biotechnology 24, 1392 - 1401 (2006). 1139] For example., cells expressing markers characteristic of the definitive endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endodermi lineage, by treating the cells expressing markers characteristic of the definitive endoderm lineage with a fibroblast growth factor and the hedgehog signaling pathway inhibitor KAAD-cyclopainine, then removing the medium containing the tibroblast growth factor and KAAD-cyclopamine and subsequently culturing the cells in medium containing retinoic acid, a fibroblast growth factor and KAAD-cyclopamine. An example of this method is disclosed in Nature Biotechnology 24, 1392 - 1401 (2006). 1140] For example, cells expressing markers characteristic of the definitive endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endoderm lineage, by treating the cells expressing markers characteristic of the definitive endoderm lineage with retinoic acid one fibroblast growth factor for a period of time, according to the methods disclosed in US patent application Ser. No, 11/736,908, assigned to LifeScan, Inc. 01411 For example, cells expressing markers characteristic of the definitive endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endoderm lineage, by treating the cells expressing markers characteristic of the definitive endoderm lineage with retinoic acid (Sigma-Aldrich, MO) and exendin 4, then removing the medium containing DAPT (Sigma-Aldri ch, MO) and exendin 4 and subsequently culturing the cells in medium containing exendin 1, IGF-l and HGF An example of this method is disclosed in Nature Biotechnology 24, 1392 - 1401 (2006). 1142] For example, cells expressing markers characteristic of the pancreatic endoderni lineage obtained according to the mefhods of the present invention are further differentiated into cells expressing markers characteristic of die pancreatic endocrine lineage, by culturing the cells expressing mark-ers characteristic of the pancreatic endoderm lineage in medium containing exendin 4, then removing the medium containing exendin 4 and subsequently culturing the cells in medium containing excndin 1, lGF-1 and HF. An example of this method is disclosed in D' Amour et a', Nature Biotechnology, 2006, 1143] For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by culturing the cells expressing markers characteristic of the pancreatic endoderm lineage in medium containing DAPT (Sigma-Aldrich, MO) and exendin 4. An example of this method is disclosed in D' Amour el al, Nature Biotechnology 2006. 1144] For example, cells expressing markers characteristic of the pancreatic endodermi lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by culturing the cells expressing markers characteristic of the pancreatic endoderm lineage in medium containing exendin 4. An example of this method is disclosed in D' Amour et al, Nature Biotechnology, 2006. 01451 For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser, No, 11.736,908, assigned to LifeScan, Inc. 3 4 11461 For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderni lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No. 11/779,311, assigned to LifeScan, Inc. 1147] For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No. 60/953,178, assigned to LifeScan, Inc, 1,48j For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No. 60/990,529, assigned to LifeScan, Inc. Diiferentiation of Pluripotent Stem Cells Pnned LVing the Aethods ofthe Present Invention into Cel/s Exipressing Markers Characteristic of the Pancreatic Endocrine Lineage 01,491 Pluripotent stem cells formed using the methods of the present invention may be differentiated. into cells expressing markers characteristic of the pancreatic endocrine lineage by any method in the art. 01501 For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine 35S lineage, by culturing the cells expressing markers characteristic of the pancreatic endoderm lineage in medium containing exendin 4, then removing the medium containing exendin 4 and subsequently culturing the cells in medium containing exendin 1, IGF- I and HOF, An example of this method is disclosed in D' Amour et al, Nature Biotechnology 2006, 11511 For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by culturing the cells expressing markers characteristic of the pancreatic endoderm lineagein medium containing DAPT (Sigma-Aldrich, MO) and exendin 4. An example of this method is disclosed in U Amour el al, Nature Biotechnology, 2006. 1152] For example, cells expressing markers characteristic of the pancreatic endodernm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by culturing the cells expressing markers characteristic of the pancreatic endoderm lineage in medium containing exendin 4, An example of this method is disclosed in D' Amour el al, Nature Biotechnology, 2006. 11531 For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated. into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No, 11f736,908, assigned to LifeScan, Inc. 0154] For example, cells expressing markers characteristic of the pancreatic endodermi lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according 36 to the methods disclosed in US patent application Ser. No. 11/779,311, assigned to LifeScan, Inc. 155] For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the mefhods of the present invention are further differentiated into cells expressing markers characteristic of die pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No. 60/953,178, assigned to LifeScan, Inc, 1561 For example, cells expressing markers characteristic of the pancreatic endoderm lineage obtained according to the methods of the present invention are further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No, 60/990,529, assigned to LifeScan, Inc. 157] The present invention is further illustrated, but not limited by, the following examples. EXAM PLES Example I Differentiation of Human Embryonic Stem Cells of the Cell Line HT to Pancreatic Endocrine Cells in the Absence of Fetal Bovine Serum 0158] Cells of the human embryonic stem cells line -il at various passages ( p40 to p5 2 ) were cultured on MATRIGEL (1:30 dilution) coated dishes and differentiated into pancreatic lineages using a multi-step protocol as follows: a. Stage I (Definitive Endoderm): Human embryonic stem cells were cultured in RPMI medium supplemented with 2% fatty acid-free BSA (Catalog# 68700, Proliant, IA), and 100 ng/ml activin A (R&D Systems, MN) plus 20 nug.l WNT 37! 3a (Catalog# 1324-WN-002, R&D Systems, MN) plus 8 ng/ml of bfGF (Catalog# 100-18B, PeproTech, NJ), for one day. Cells were then treated with RPMII medium supplemented with 2% BSA and 100 ng/ml activin A plus 8 ng/mi of bFGF for an additional two days, then b, Stage 11 (Primitive gut tube): Cells were treated with RPM] + 2% fatty acid-free BSA and 50 .ng/ml FGF7 and 0.25 pM SANT-I (#S4572, Sigma, MO), for two to three days, then c, Stage fit (Posterior foregut): Cells were treated with DMEM/High-Giucose supplemented with 1:200 dilution of ITS-X (Invitrogen, CA) and 01% BSA (Lipid Rich) (Invitrogen, Ca No. 11021-045) , 50 ng/mil FGF7, 0.25 pM SANT 1, 2 pM Retinoic acid (RA) (Sigma, MO), 100 ng/l of Noggin (R & D Systems, MN), and Activin A at 20 ng/ml for four days; In certain variations, Nogginr was replaced with the A MPK inhibitor 6-{4-(2-Piperidin I -ylethoxy)phenyl]-3 pyridin-4-ypyrazolo[1 ,5-a]pyrimidine (Sigma, No, PS499) at a concentration of 2 p.M. In yet other variations, a P38 inhibitor (4-[4-(4-Fluorophenyl)-1-(3 phenylpropy)-5-pyridin-4-y-limidazol-2-ylbut-3-yn- 1.-ol) (disclosed in US Patent 6,521655) was added at 2.5 p.M, then d. Stage IV (Pancreatic endocrine precursor): Cells were treated with DMEM/High Glucose supplemented with 1:200 dilution of ITS-X (Invitrogen, CA) and 0.1% BSA (Invitrogcn, Ca), 100 ng/ml Noggin, 1 pM ALK5 inhibitor (SD-208, disclosed in Molecular Pharmnacology 2007 72:152-161) for three days, then e. Stage V (Pancreatic endocrine cells): Cell were treated with DMEM/High Glucose supplemented with 1:200 dilution of1T-X (Invitrogen, CA), 0.1% BSA (invitrogen, Ca), 1 pM ALK5 inhibitor 11 (Catalog# 616452, Calbiochem, Ca) for seven days., then f Stage VI (Mlature Pancreatic endocrine cells): Cells were treated with DMEM/High-Glucose supplemented with. 1:200 dilution of ITS-X (Invitrogen., CA), 0.1% BSA (Inivitrogen, Ca) for seven days, with media changes every other day, 38 Example 2 Flow Cytometric Characterization and Sorting of Enriched Various Pancreatic Cell Lineages 1159] To facilitate the isolation and characterization of novel cell populations form various stages of the differentiation process outlined in Example 1, a detailed characterization of the cells obtained from the various stages was done by flow eytometry. A complete list of antibodies used and the expression levels of surface markers at various stages of differentiation is shown in Table L 1160] Cells of the human embryonic stem cell line HI at various passages (p40 to p52) were cultured on MATRIGEL-coated plates, and differentiated into pancreatic endocrine cells using the protocol described in Example I 11611 Cells at different stages of maturation (posterior foregut (Stage ffI), endocrine precursor cells (Stage IV), pancreatic endocrine cells (Stage V) or mature pancreatic endocrine cells (Stage Vii were gently released by incubation in TrypLE Express (Iavitrogen # 12604, CA) for 2-3 minutes at 37TC and washed twice in BD FACS staining buttr containing 2% BSA (13D 4 554657, CA). Approximately 0.5-1xi10 cells were re-suspended in 1002 00 pl blocking buffer (0,5% human gaimma-globulin diluted 1:4 in staining buffer (BD, CA) for staining. For staining with directly conjugated primary antibodies, the appropriate antibody was added to the cells at a final dilution of .1:20, and cells and incubated for 30 min at 4* C. For unconjugated antibodies, primary antibodies were added to cells at 1:50-1:100 dilution and cells incubated for 30 min at 4"C followed two washes in staining buffer. Cells were then incubated in the appropriate secondary antibodies at 1:500 dilution. Stained cells were re-suspended in 300 pl staining buffer and 5-10 p- of 7AAD added for Ii ve/dead cell discrimination prior to analysis on the BD FACS Canto IL 01621 For cell sorting, approximately 30-40 million cells were similarly processed as for flow cytometric analysis Cells were stained with the appropriate antibodies as shown in Table I. Cells were sorted either into two or three sub-populations as summarized in Table I. Cell sorting gates were established based on the isotype matched controls. An aliquot of sorted cells were analyzed for purity following the sorting followed by 39 PCR analysis for expression of key pancreatic markers. RNA was collected using the Rneasy Mini Kit, Qiagen, CA) was collected from presort sample, and the various fractions. 1163] Cell surface markers used for sorting were selected based on the expression of various markers in populations of cells analyzed at different stages of the differentiation protocol outlined in Example 1. The markers employed in this study are disclosed in Table II Briefly, the surface markers disclosed in Table 11 were used either singly or in combination to sort various populations of cells. Samples of the sorted cells were taken to analyze the expression of markers characteristic of the pancreatic endocrine lineage by real-time PCR, Srnfing x ng markers Characteristic ofthe Pancreatic Endocrine Lineage 164] Antibodies to CDS6 and CD13 were used to sort a population of cells obtained from Stage IV of the differentiation protocol outlined in Example 1 Three populations of cells were identified: a) CD56CD13, b) CD56-CD13 and c) CDS6-CD13" populations of cells. The CD56CD13' population was enriched approximately 1.3 fold following sorting, and the sorted cells were highly enriched for the expression of markers characteristic of the pancreatic endocrine lineage, including NEUROD, NGN3, PDX1, NKX6. 1, NKX2,2 and PAX-4,when compared to unsorted cells at stage IV, or populations of CD56'CDI3" cells, or populations of CDS6-CDI3' cells. See Figure 1, panels a-f. 0165] In a second series of experiments, antibodies to CD133 were used to sort a population of cells obtained from Stage IV of the differentiation protocol outlined in Example L Two populations of cells were identified: a) CD133, and b) CD133" populations of cells. The CD33' population was enriched approximately 1.9 fold following sorting, and the sorted cells were highly enriched for the expression of markers characteristic of the pancreatic endocrine lineage, including NEUROD, NGN3, PDX1, NKX6.1, NKX2.2 and PAX-4;when compared to unsorted cells at stage IV, or populations of CD133" cells. See Figure 2, panels a-f, 40 11661 In a third series of experiments, antibodies to CD49c were used to sort a population of cells obtained from Stage V of the differentiation protocol outlined in Example 1. Two populations of cells were identified: a) CD49ct and b) CD49c"' populations of cells. CD49cA" cells were enriched approximately 3.1 fold following sorting, and the sorted cells were highly enriched fhr de expression of markers characteristic of the pancreatic endocrine lineage, including NEUROD, NGN3, PDXI, and NKX6,1 when compared to unsorted cells or CD49e" cells. See Figure 3, panels a-d. 1167] In a fourth series of experiments, antibodies to CD56 and CD 15 were used to sort a population of cells obtained front Stage IV of the differentiation protocol outlined in Example 1L The following populations of cells were identified: a) CD56*CD15 b) CD56CDI 1 " c) CD15 t and d) CD15 populations of cells. Populations of CD15 cells were enriched approximately I. 1 fold following sorting. Populations of CD56CD15" cells were highly enriched for the expression of markers characteristic of the pancreatic endocrine lineage including NEUROD, NGN3, PDX 1, NKX6,1, hisulin and glucagon compared to unsorted cells, or populations of CD56CDI 5 hi cells. See Figure 4, panels a-fE Similarly, populations of CD cells sorted using a single marker were highly enriched for the expression of markers characteristic of the pancreatic endocrine lineage including NEUROD, NGN3, PDXI, NKX6 1, NKX2.2, PAX-4, gucagon and insulin, when compared to unsorted cells or populations of CDI5" cells. See Figire 5, panels a-h. 11681 In a fifth series of experiments, antibodies to CD56 and CD57 were used to sort a population of cells obtained from Sagne IV of the differentiation protocol outlined in Example 1. Two populations of cells were identified: a) CD56 CD$7 and b) CD56'CD57~populations of cells. Populations of CD56"CD574 cells were enriched approximately 1 9 fold following sorting, CD56 CD57* cells were highly enriched for the expression of markers characteristic of the pancreatic endoderm lineage, including NEUROD, NGN3, PDXI, NKX6. 1, NKX22., as wells as insulin and glucagon, when compared to unsorted cells or populations of CD56.CD57 cells. See Figure 6, panel a-g, Similar results were observed when populations of cells at Stage V of the differentiation protocol outlined in Example I were sorted using antibodies to CD56 and CD57, 41 11691 In a sixth series of experiments, antibodies to CD56 and CD184 were used to sort a population of cells obtained from Stage IV of the differentiation protocol outlined in Example 1, Three populations of cells were identified: a) CD184", b) CD184', and c) CD56 CD184~ populations of cells. Table IV sunmarizes the expression of CD184 in cells before and after the enrichment. Populations of CD184" cells were enriched for the expression of markers characteristic of the pancreatic endocrine lineage, including PAX4, NEUROD, NKX6.1, PDXI and PTF1 alpha, The expression of ZICi, Albumin and CDX2 was decreased. See Figure 7, panels a-i. Sorting ofmInsulin Producing Cells 1.1701 Antibodies to CD98 were used to sort a population of cells obtained from Stage VI of the differentiation protocol outlined in Example I. Two populations of cells were identified: a) CD98 , and b) CD984 1 populations of cells. Populations of CD98 cells were enriched approximately 1.6 fold following sorting. CD98r cells were enriched for the expression of NEUROD, NGN3, insulin, and glucagon. See Figure 8, panels a-d, 1171] In another series of experiments, antibodies to CD47 were used to sort a population of cells obtained from Stage V of the differentiation protocol outlined in Example I Two populations of cells were identified: a) CD474 > and b) CD47' populations of cells. CD 4 7 L,) cells were enriched approximately 3.3 fold following sorting. CD47'"O cells were enriched for the expression of NEURDD, NUN3, PDXI, NKX6.1, NKX2.2 and PAX4. See Figure 9, panels a-f 0172] In another series of experiments, antibodies to CD47 were used to sort a population of cells obtained from Stage VI of the ditfrentiation. protocol outlined in Example 1. Two populations of cells were identified: a) CD47"t and b) CD47'" populations of cells. CD47%o cells were enriched for the expression of PDX- 1, NKX6.1, NKX2:2, PAX-4, PTFia, NQN3, Insulin and Glucagon. See Figure~ 10, panels a-h. Example 3 Sorting of Lif receptor Positive Cells at Primitive GutT ube stage (Stage 2) 42 1731 Cells of the human embryonic stem cell line HI at passage 44 were cultured on MATRIGEL-coated plates, and differentiated into insulin producing cells using the following protocol: a. RPMI medium supplemented with 2% fatty acid-free BSA (Catalog# 68700, Proliant,IA), and 100 ng/ml activin A (R&D Systems, MN) plus 20 ng/ml WNT-3a (Catalog#4 1324-WN-002, R&D Systems, MN) plus 8 ng/ml of bFGF (Catalog# 100-1813, PeproTech, NJ), for one day followed by treatment with RPMI media supplemented with 2% BSA and 100 ng/ml activin A plus 8 ng/mil of bFGI for an additional two days (Stage 1), then b. RPNII + 2% BSA + 50 ng/ml FGF7 + 0.25 pM SANT-1 (#S4572, Sigma, MO), for three days (Stage 2), then c. DMEM/High-Glucose + 1::200 dilution of ITS-X (Invitrogen, CA) + 0.1% BSA (Invitrogen., Ca) 50 ng/nl FGF7 (Peprotech, NJ) + 0.25 pM SANT- I + 2 pM Retinoic acid (RA) (Sigma, MO)+ 100 ng/mi of Noggin (R & D Systems, MN) and 20 ugg/nl of activin A for four days (Stage 3), then dDMEMJHigh-Glucose + 1:200 dilution of ITS-X (invitrogen, CA.)+ 0l% BSA (Invitrogen, Ca) - 100 ng/ml Noggin + 1 pM ALK5 inhibitor (SC10120) + for three days (Stage 4) 11741 Stage 2 cells were dispersed into single cells using TrypLE Express (Invitrogen, Carlsbad, CA) and washed in stage 4 basal media (DM-Hg + lTS-X B USA). Released cells were spun and the resulting cell pellet suspended in a staining buffer consisting of 2% BSA, 0.05% sodium azide in PBS (Sigma, MO), As appropriate, the cells were Fc-receptor blocked for 15 minutes using a 0.1% y-globulin (Sigma) sol ution. Aliquots (approximately 105 cells) were incubated with Lif receptor Phycoerythrin (PE) (R & D Systems, MN) conjugated monoclonal antibodies (5 p t antibody per 10 cells), Controls included appropriate isotype matched antibodies and unstained cells. All incubations with antibodies were performed for 30 mins at 44C after which the cells were washed with the staining buffer. Stained cells were sorted on. a FACS Aria (B1D, Ca). RNA (Rneasy Mini Kit, Qiagen, CA) was collected from 43 presort sample, Lif receptor- fraction and Lif receptor negative fraction. The Lif receptor expression level and pattern is summarized in Table Ill, 175] Table flf summarizes the expression of Lif receptor at days 2 and 3 of stage 2, By day 3 of stage 2, approxinmately70% of the cells expressed Lif receptor. As sunimarized in Table It, high expression of Lif receptor was unique to stage 2 cells, as stage 3 and 4 cells showed minimal expression of Lif receptor. As shown in Figure 11, panels a-b, stage 2 cells enriched for the Lif receptor showed a significant increase in expression of HNF4 alpha as compared to unsorted cells or Lif receptor negative cells. Expression of Lif receptor miRNA as measured by real-time PCR was also enhanced in cell fraction containing Lif-receptor positive cells. Example 4 Magnetic Bead Sorting for Cells for the Depletion of SSEA-4+ Cells to Reduce Tumor Formation in Vivo 11761 Expression of the SSEA4antigen is a key indicator of piuripotency in human embryonic stem cells, and expression of this marker is greatly down regulated during the differentiation process. However, residual SSE A-4 positive cells may be responsible for tumors and/or teratomas that are observed flowing transplantation of partially differentiated cells. To reduce teratoma formation, methods were developed to deplete contaminating SSEA4 cells from differentiated cells prior to transplantation. 01771 Cells of the human embryonic stem cell line H I (passage 40-52) were differentiated to various stages of the differentiation protocol outlined in Example 1. In order to test proof of concept and efficacy of SSEA-4 depletion, this study was first done with cells differentiated only to the primitive gut tube stage (Stage 2 in the differentiation protocol outlined in Example 1) in order to ensure cells still retained higher levels of SSEA-4 expression. In subsequent experiments, cells expressing SSEA-4 were depleted in populations of cells differentiated at Stage 4 of the differentiation protocol outline in Example 1. See Table V for the results observed. Cells were gently released into single cells by incubation in TrypLE Express (Invitrogen # 12604, CA) for 2-3 minutes at 37TC To enhance cell survival and viability during depletion, anti 44 apoptotic agents including 10pNM Y-2763:2 (Cat 4 Y 0503, Sigma, St Louis MO) or 0.5 iM Thiazovivin (Cat # 04-0017, Stemgent, San Diego, CA) were added to the cells prior to collection and in all isolation buffers, 1178] Cells were washed in Isolation Buffer containing Ca and. Mg> free phosphate buffered saline (PBS) supplemented with 0.1 % BSA and 2mM EDTA. Between 10 100 x 104 cells were re-suspended in isolation buffer a final cell density of S x 10i cells per 500 pl Twenty five l SSEA-4 antibody was added per 500 pl of cells and cells incubated for 15-20 minutes at room temperature on a gentle rocker to ensure continuous mixing, Cells were washed in isolation buffer by spinning at 300xg for 8 min. Supernatant was removed and cells re-suspended in original buffer volume and 50 pl of prewashed SSEA-4 Depletion beads (DynaBeadsT SSEA-4, Invitrogen, #11 160D) added for every 500 pi of cell suspension. Cells and beads were mixed and incubated for 15-20 minutes at room temperature with continuous gentle tilting and rotation. Cells were mixed by gentle pipetting and placed on a magnet for 5 min, The supernatant containing SSEA-4 negative cells was transferred to a new tube and the process repeated 2-3 times to remove residual beads. Bead-bound SSEA4* cells were released from magnetic field and both cells populations counted and processed for FACS and PCR analysis. The expression levels of SSEA4 in undifferentiated HI cells, primitive gut cells and Stage IV cells, in both pre-sorted and sorted cell fractions is summarized in Table V. 11791 In populations of cells isolated at stage U of the differentiation protocol outlined in Example 1, 20.5 % of the cells expressed SSEA4 markers in prior to sorting. In. contrast, only 1.8% of the cells expressed the SSEA4 post sort (Table V). The depletion resulted in removal of 91.2 % of the SSEA-4 positive cells. In another experiment using endocrine precursor cells, 253% of cells expressed SSEA-4 prior to depletion, but only 0.9% expressed SSEA-4 after depletion, resulting in 95.5 % removal of SSEA-4 positive cells (Table V). In contrast to differentiated cells, 91.2% of the population of undifferentiated embryonic stem cells expressed SSEA4. 01,80] The sorted SSEA4" cells were highly enriched for the expression of pluripotency markers, including OCT4, NANOG, SOX2 and goosecoid (Figure 12 panels a-d). 45 Example 5 Sorting of SSEA4+"") and SSEA4 4 LO Cells by FACS 181] In order to investigate and confirm the depletion of pluripotent-marker (SSEA-4+) enriched cells from differentiated cells by flow cytometry, cells were differentiated to Stage VI as described in Example L Cells were released from culture using TrypleE Express cell dissociation buffer and cells prepared for sorting as described in Example 2 The SSEA-4 antibody (R&D Systems, Minneapolis, MN, Cat # FAB1435P) was used to isolate two cell fractions identified as SSEA-4(+)Hi and SSEA-4(~)Lo cells. Isolated cell fractions were analyzed for expression of pluripotency markers by RT PCR as described in Example 4. Sitilar to SSEA-4 depleted and enriched fractions obtained using magnetic beads separation, as described in Example 5, the sorted SSEA-4(+-l)Hi cells were highly enriched for the expression of pluripotency markers OCT4, NANOG, SOX2 and goosecoid, unlike the SSEA-4(-)Lo cells, See Figure 13 panels a-d, Example 6 Transplantation of SSEA-4 Depleted Populations of Cells in Vivo 1182] In pilot experiments, SSEA-4 depleted cells weredifferentiated to Stage IV of the differentiation protocol outlined in Example 1, and then transplanted into the kidney capsule of mice to test cell survival and engraftment. The data from the transplanted mice is summarized in Table VIL 01,831 Five to six-week-old male scid-beige mice (.BIg-i b/GbmsVac P LysV< N71 were purchased from Taconic Farms, Mice were housed in microisolator cages with free access to sterilized food and water. In preparation f surgery, mice were identified by ear tagging and their body weight measured and their blood glucose determine by a hand held glucometer (One Touch, LifeScan). Mice were anesthetized with a mixture of isolflurane and oxygen and the surgical site was shaved Aith small animal clippers, Mice were dosed with 0,1 mg/kg Buprenex subcutanuusly pre operatively, The surgical site was prepared with successive washes of 70% isopropyl alcohol, 10% povidone-iodide, and 70% isopropyl alcohol and a left lateral in cision 46 was made through the skin and ascle layers, The left kidney was externalized and kept moist with 0.9% sodium chloride, A 24G x %" LV, catheter was used to penetrate the kidney capsule and the needle was removed. The catheter was then advanced under the kidney capsule to the distal pole of the kidney, 1.841 During the preoperative preparation of the mice, the cells were centrifuged in a 1.5 niL microfige tube and most of the supernatant removed, leaving just enough to collect the pellet of cells. The cells were collected into a Rainin Pos-D positive displacement pipette and the pipette was inverted to allow for the cells to settle by gravity, The excess media was dispensed leaving a packed cell preparation tor transplant. 1185] For transplantation, the Pos-D pipette tip was placed fimly in the hub of the catheter and the cells dispensed from the pipette through the catheter under the kidney capsule and delivered to the distal pole of the kidney. The lumen of the catheter was flushed with a small volume of culture media to deliver the remaining cells and the catheter withdrawn. The kidney capsule was sealed with a low temperature cautery and the kidney was returned its original anatomical position. The muscle was closed with continuous sutures using 5-0 vicryl and the skin closed with wound clips. Mice were dosed with LO. mg/kg Metacam subcutaneously post-operatively. The mouse was removed from the anesthesia and allowed to fully recover. 186J Following transplantation, mice were weighed once per week and blood glucose measured twice a week. At various intervals following transplantation, mice were dosed with 3 g/kg glucose IIP and blood drawn via the retro-orbital sinus 60 minutes following glucose injection into microfuge tubes containing a small amount of heparin. The blood was centrifuged and the plasma placed into a second microfuge tube and frozen on dry ice and then stored at -80'C until human c-peptide assay was performed. Hunan c-peptide levels were determined using the Mercodia/ALPCO Diagnotics Ultrasensitive C-peptide ELISA according to the manufacturer's instructions. 47 187] At the time of sacrifice, blood was collected as described above and nce euthanized The grafts were harvested from the kidney capsule and analyzed by real-time qPCR, immunohistochemistry, and pathology. 1188] Three groups of mice were transplanted with about 3.3 million cells each comprising of i) cell clusters ii) single cells (undepleted) and iii) SSEA4 depleted single cells, Cells differentiated to Stage IV were ei other released with gentle scarping to make small cell clusters, or released with TrypleE into single cells for SSEA-4 depletion, Following SSEA-4 depletion as outlined in Example 5 , both cell clusters and single cell prepations were repeated in low attachment plates (Cos tar, Corning Incorporated, NY Cat # 3471) overnight in precursor (Stage IV) cell differentiation medium prior to transplantation. The rock inhibitor Y-27632 dihydrochrolide monohydrate (Sigma, Cat # Y0503) was added to the culture ovemight at a concentration of 10 pM. Following transplants, mice were monitored as described above for up to 12 weeks post tirnsplants. Graft survival was not visibly demonstrated in the single cells recipients (depleted or undepleted) but was shovn in 2 out of 5 mice receiving cell clusters. One out of 5 mice receiving cell clusters had detectable c-pptide. levels at 12 weeks post transplantation. Poor graft survival was attributed to diminished cell quality and low numbers of cells transplanted in the pilot experiment. 11891 The multi-step differentiation of human embryonic cells into mature, pancreatic endocrine cells through several intermediate steps including definitive endoderm (DE), pancreatic endoderm (PE) and pancreatic precursors is associated. with. dynamic changes in expression of surface markers, AlthouAh the differentiation protocol may produce as yet undefined, heterogeneous cell populations of multiple lineages including ectodermal and mesodermal cell types, tracking the changes in expression of surface markers in pancreatic differentiation medium could identify markers potentially usetil in cell enrichmnt and purification. Table VII shows a summary of surfitee inarkers that either demonstrated an increase or decrease in expression, that may be useful for negative of positive selection of pancreatic endoderm cells, Markers that decreased in expression during the differentiation process include CD1117, CD133, CD181, CD184, CD200, CD221, CD326, CD55, CD57, CD9, and CD98. Markers that increased in exprrssion during the differentiation process include 48 CD13, CD141, CD15, CD318, CD46, CD47, CD49c, CD49e, CD56, and CD73. These markers could singly or in various combinations be used to purify cell populations enriched for pancreatic endoderm and precursors. Example 7 Flow Cytometric Sorting Procedures 1190 Cells at different stages of maturation were gently released by incubation in TrypLE Express (Invitrogen #t 12604, CA) for 2-3 minutes at 37*C and washed twice in BD FACS staying buffer containing 2% BSA (BD # 554657, CA). Based on cell yields, 20-50 x106 single cells were re-suspended in 2-3 ml of blocking buffer (0,5% human gamma-globulin diluted 1:4 in staining buffer (13, CA) for staining. Fluorophore conjugated primary antibodies were added to the cells at a final dilution of 1:20 and cells and incubated for 30 min at 4* C, Following washes, stained cells were re suspended in 2-3 ml staining buffer and 50-60 pl of 7AAD added for liv/dead cell discrimination prior to analysis and cell sorting. Isotype matched control lgG antibodies were used tor negative control staining. For calculating fluorophore compensation values prior to sorting, cell were either left unstained or stained with single fluorphore. of Fluoroscein isothiocyanatc (FITC), Phycoerythrin (PE) or Allophycocyanin (APC) the nuclear dye 7-Aminoactinomucin D (7-AAD). '191] Cell sorting was done using the BD FACSAria cell sorter and the BD FACSDiva soft-ware. Isotype matched control cells were used to establish negative gates for each cell sorting. For each cell sorting experiment, the photonmi itiplier (PMT) voltage Settings werc adjusted using the appropriate fluorophore compensation values to produce a bright population (positive (+) or Hi ) and dim population or cell subset (Negative (~) or Lo). Typically, positive cells populations (4- or Ili) were of the order of third decade or higher (104 while negative population were in the first to second decade (102 10), Using established gates, cells were sorted using a 100 pM nozzle and a flow rate of 1.0, Following sorting, a small aliquot of cells were analyzed to assess the purity of the sorted cell subsets. RNA was collected from the presort and sorted cells using the Rneasy Mini Kit, Qiagen, CA) for RT-PCR analysis, 49 1192] Publications cited throughout this document are hereby incorporated by reference in their entirety, Although the various aspects of the invention have been illustrated above by reference to examples and preferred embodiments, it will be appreciated that the scope of the invention is defined not by the foregoing description but by the following claims properly construed under principles of patent law. 50 Table 1. Flow Cytometric Characterization of Surface Marker Expression at Different Stages of Endodermal/Pancreatic Differentiation Key: ND = Not Determined; +/- = 0-10%; + = 10-50%; ++ = 50-85%; +++ = 85-100% Awfiut rSnwnvm.t It'do (RtS Defidc -e imi' Posknor I dnne tinn ki .eia J~ iiii h fv POVa lEdin { EWai"Vn, Endotker G Tube Fnwgut Ihwursor) e Cels Endcrine n (Sirg2) (Siage 3i (Stage 4) (tke 5) C4l11 ______ ________ ______ _____ (Sraswe (____ _____ _____Stooe 6) BL TR D52BD#2836 ND + CD 105 Endog n~f M~IIIom C 4 ±1 ________ ___________ BL4 18F ____ ____ ____ ____ ___ CD)112 PRR2 BD#51057 CD117 c-kit BD#34109 6 + .J CD118 LIFR, gp190 R&D+FAB2 49P' CD126 1L-6R BD#4551850 + C13 Aminopeptida BD4555394 + _________ seN ______ ____ ____ _____ C)130 L-6R gp130 BD#S55757 + CDM32BD590 C-133 A(33, MILTENYI + + + pro)~mi-kel 130-098 854 CD134 X-40 B1D45154S4 +I" CD135 3k $1F k2 D#89% CD137 90#594464 Ligand CD140a PDGFRa --------0C2 j. , + C.D140b PDGFR3 B D--588 2-- CD142 1550312 + C146 MUCi8 Bl#55i30 5 + + ND C115 BD#45i?43 + + -+ + CD161 B D#340536 1 C D .1 6 4 _ _ _ _ _ _ __...B D # T --- 55 12 9 CD178 Pa CD5 BD55293 + NI) C180 _________BD#551953 +____ ____ CD181 (CRI BD1555939 + + + ND RRA, (CD)83 (XC BD#314550967 +______________ CD184 CR4, ftUs 1D#55976 + + + CD1P85 CXCR5 BD#51959 + + ND CD193 (CR3 #1055865 + . 6 + CD195 21CCR5 1)4555t92 + ND () Bb ---------- 555- - CD200 OX-2 BD#t552475 41 +___ 4+ + 1 +-1 CD2~05 BDYS 58069 + ____ ______ CIY220 Inulin-R B3u559955 ND ) CD22 IGFI R BD-9 + ++++ CD24 BD#555428 ++ + +++ ++ ++ + CD243 MDR-1; P-p BD#557002 +- +- +- +:2 + CD2152 OX-40 LiamdIBD#558164 1- 4 , + CD26 D #555436 + CD271 NGFR BD#557198 + ND ND C"'D275 1D15 #522 + CD28 RBD4# P1 1,555728 + 51 CD29 I t55983 +-*-i-p-I-B-o- 4s4-4-6444+ ++ +.. +++++ +..-4 ------ ------_ _ _ _ -------- --- ---- ------------ -------------------- CD305 IR\RI BDI5508H I TAT_7____ N) CD309 VEGFR2 T D4560494 KDR, CD318 CDCDPI R&D/WAB2 + ________ ____________ 6662P ____ ____ CD32 -CA\M BDs347197 ++ ++ CD33 BD#______ 1 555450 +4 -- 44 _____ - CD33 FGFR2, R&D#FAB66 +i -- 4 4 44i KGF-R-2 -4A CD340 RhB-2 Di3'405 +- HE1R2/ neu (D36 BDi550956 + CD39 BD1#555464 + +- + +- NID C1D42b BD555472 + CD43 BD555475 + -ND (C44 [D59942 + - 1+ CD46 BD555949 + 41 +4 - + ND (D47 BD_+56046 +++ ++ ++ CD49b a2 lItegrin B1)z555669 4 ++- + 4 4K VLA-2 CD49c a3 1ntegri Abormn#ab3 +* 4 + + VLA-3 0489 CD49e 05 Integrin B555617 ++ +++ + 4 CD49f a6 Inteugra BD3555.735 + + + + + + + VLA-6 CD5 B10555696 + + - -+. + + + CD36 INCAM V BDAI 1555518 + + + + +++ + +++ CD57 HD#555619 +++ +-+- -++ + + + + SCD$8 LFA-3 -BD0555920 + - 4;. 4+-6i C D63 I AMP B305 5 NI) +-!- - -) 3 CD66 BD#551480 4 CD1 1 55174 CD73 +I)550257 + ND CD7(14 BD_______ #6)555540 +1f +r- 1 4- - ____ ND C8 C CUR BD0 550494 + + + + CD9 P241 MRP-1 B DU555372 + + - + + - ---- CD91 BD+550496 + + + CD95 Apo- Fas B- D5-674 +--- ND C998BD56076 6 ++ ++ C99 MIC E2 ++ +++ ++ +++ ++ C (1210 IL110 R BD556013 ±: 4+-4 + DLLI R&D4FAB1 ND ND ND + ND EGFR EMB, BD555997 + HER fMILP BD#556016 + NM B BD)58352 + . ... ND NotchD + - ------------ N4 SSEA- R&DFABIf 435P TWFBR3 -Iespan#LS + ND + + + ND TRA1 60 BD__ 560 193 +-+ +++ IRA I41 8 1-0D560 6 *-++ +++ - - + + lTAK B DU 552890 -?+- -6-- 52 . .. .. .. . . .. .. .. . . . . .. .. . . . .. .. .. . . . ... ... .. ... ... .. .. ... ... .. ... ... I ... ... .. ... ... ... ... .. ... ... ..
Table IT Surface Markers used to Enrich for Pancreatic Cell Precursors Ui(d Single Nos Enrk eed Pph ucm Cta miardPopdkAtion s CD6) D13 Ersdrerisc Prcvrsors (S4) B D1) 18 : CDSG6CD 13 641 2.} C 133 Jd tc i ( G) .33- 4s() Embcnne Prmors (44) X091 54 CD49c(w n-doexne Jacxcsors (4) AbCtMcrb04 CD49c"' 31,7 95.9 CDS6 CDI5 n~doene Piccrsors (4 oD655558 CD6 CD1 2640 NID ND 55176 CD CS Endocl nc Prctisors (4) BD#t.1376 CDI 89.6 975 CD35XCD57 MarcciccPrecurs (54) MM538 C5 D 3L.3 59M -9 t n~nc c3; 145 555619 CD984 P nsne Crdh (46 .D65560376 CD9)9C 6L.3 94. - .6 Cn47 xmlcnnecens (4S S6 1>6646 (')4 22. - 3.3 Table HLi Expression levels oft LiF Receptor Stage of Stage 2, Day2 Stage 2, Day3 Stage 3, Day4 Stage 4, Day3 Differentiation _______________ ______________ Expression 47% 70% 5% 1% Level (%~) - - - - -- - - - - -- - - - - ---- ------ ------ ------- ------ ------- ------ ------ ------ ------- ----- ------- ------ ------ ------ ------- ------ 5 4- ---- ------ Table IV. Expression Levels of CD184 Before and After Enrichment ike-ort Enriched C84 CD4 D pi C 54D56 C ( 4D6i 4CDS6-(& CD194- CO184+ CDi4+ CD%4 CD56CD56+ 1% 20% 70% 9% (%) Table V. Expression Levels of SSEA4 Antigen Cells Stage of SSEA-4 Expression %) % Fold Differentiation Depletion P_re-Depletion Post-Depletion Hi Undiffercntiated 91-2 ND ND Hl Primitive Gult 205 I'S 912 Hl Endocrine 20.1 0 9 95 Precursors (Stage RJ55 Table VT. Summary Data of Mice Transplanted with SSEA-4 Depleted Cells Group Cell Type Total Cell No. of Ace Grafts at 12 C-Peptile Io MWeek at 12 0?eks Cell 3.3 million 5 3/5 mice 1/5 mice Clusters visible detectable c~ Single Cells 3.3 million 5 0/5 mice 0/5 mice Undepleted with visible detectable c peptie 3 SSEA-4 3.3 million 2 0.2 mice 0/2 mice Depleted with visible detectable c Single Cells grafts peptide Table V11. Surface Markers Associated with Differentiation of Human Embryonic Stem cells into Pancreatic and Endodernal Lineages, Swface Markers Changes Swrfoce akuers Cell Fmetions Changes During Associated with Used To Enrich Enriched D4iferentiation Surface Aarkers* Pancreatic DE-+E-+~Endocine Endodem/Endocrine CD1 17 Decrease ND CD13 Increase Yes CD 3 CD133 Decrease CD 133 CD142 Increase ND CD15 Increase Yes CD 5 CD181 Decrease ND CD184 Decrease Yes CD184+ CD200 Decrease ND CD221 Decrease N D CD318 Increase ND CD326 Decrease ND CD46 increase ND C147 Increase Yes CD47 CD49c Increase Yes CD49c CD49c Increase ND CD55 Decrease ND CD56 Increase Yes CD56+ CD95 Decrease es DS CDIE3 increase __ ___ND _______ CD9 Decrease ND a Changes associated with Surface Markers Denotes if Expression level of the particular Surface Marker Increased or Decreased as cell were differentiated from Definitive Endoderm (DE, Stage I) to Pancreatic Endoderm (PE, Stage 1I) and finally to Endocrine Cells (Stage V/VI) 56

Claims (3)

  1. 2. The method of claim 1, wherein the population of cells that express markers characteristic of the primitive gut tube lineage is enriched. 3, The method of claim 2, wherein the population of cells that express markers characteristic of the primitive gut tube is enriched by contacting the population of cells expressing markers characteristic of the primitive gut tube lineage with a reagent that is capable of binding the Lif receptor, 57
  2. 4. The method of claim 1, wherein the population of cells expressing markers characteristic of the pancreatic endocrine lineage are brought into contact with at least one reagent that is capable of binding to a marker selected from the group consisting of CD9, CD13, CD 15, CD47, CD56, CD73, CD117, CD133, CD184, CD200, CD318, CD326 and SSEA4.
  3. 5. The method of claim I wherein the mammalian animal is a human. 58
AU2015268664A 2010-03-01 2015-12-11 Methods for purifying cells derived from pluripotent stem cells Active AU2015268664B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2015268664A AU2015268664B2 (en) 2010-03-01 2015-12-11 Methods for purifying cells derived from pluripotent stem cells
AU2017276263A AU2017276263B2 (en) 2010-03-01 2017-12-14 Methods for enriching primitive gut tube

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US30919310P 2010-03-01 2010-03-01
US61/309,193 2010-03-01
AU2011223900A AU2011223900A1 (en) 2010-03-01 2011-02-28 Methods for purifying cells derived from pluripotent stem cells
PCT/US2011/026443 WO2011109279A2 (en) 2010-03-01 2011-02-28 Methods for purifying cells derived from pluripotent stem cells
AU2015268664A AU2015268664B2 (en) 2010-03-01 2015-12-11 Methods for purifying cells derived from pluripotent stem cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2011223900A Division AU2011223900A1 (en) 2010-03-01 2011-02-28 Methods for purifying cells derived from pluripotent stem cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2017276263A Division AU2017276263B2 (en) 2010-03-01 2017-12-14 Methods for enriching primitive gut tube

Publications (2)

Publication Number Publication Date
AU2015268664A1 true AU2015268664A1 (en) 2016-01-07
AU2015268664B2 AU2015268664B2 (en) 2018-01-18

Family

ID=55084444

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2015268664A Active AU2015268664B2 (en) 2010-03-01 2015-12-11 Methods for purifying cells derived from pluripotent stem cells
AU2017276263A Active AU2017276263B2 (en) 2010-03-01 2017-12-14 Methods for enriching primitive gut tube

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2017276263A Active AU2017276263B2 (en) 2010-03-01 2017-12-14 Methods for enriching primitive gut tube

Country Status (1)

Country Link
AU (2) AU2015268664B2 (en)

Also Published As

Publication number Publication date
AU2017276263B2 (en) 2019-07-11
AU2015268664B2 (en) 2018-01-18
AU2017276263A1 (en) 2018-01-18

Similar Documents

Publication Publication Date Title
US10329534B2 (en) Methods for purifying cells derived from pluripotent stem cells
US10266808B2 (en) Method of enriching for pancreatic endocrine cells that express chromogranin A
AU2008355123B2 (en) Methods for purifying endoderm and pancreatic endoderm cells derived from human embryonic stem cells
US11613736B2 (en) Isolation of bona fide pancreatic progenitor cells
AU2015200985B2 (en) Methods for purifying endoderm and pancreatic endoderm cells derived from human embryonic stem cells
AU2017276263B2 (en) Methods for enriching primitive gut tube

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)