US20100015711A1 - Differentiation of Pluripotent Stem Cells - Google Patents

Differentiation of Pluripotent Stem Cells Download PDF

Info

Publication number
US20100015711A1
US20100015711A1 US12/494,789 US49478909A US2010015711A1 US 20100015711 A1 US20100015711 A1 US 20100015711A1 US 49478909 A US49478909 A US 49478909A US 2010015711 A1 US2010015711 A1 US 2010015711A1
Authority
US
United States
Prior art keywords
cells
cat
compound
cell
differentiation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/494,789
Other languages
English (en)
Inventor
Janet Davis
Jiajian Liu
Christine Parmenter
Pascal Ghislain André Bonnet
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
Janet Davis
Jiajian Liu
Christine Parmenter
Bonnet Pascal Ghislain Andre
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janet Davis, Jiajian Liu, Christine Parmenter, Bonnet Pascal Ghislain Andre filed Critical Janet Davis
Priority to US12/494,789 priority Critical patent/US20100015711A1/en
Publication of US20100015711A1 publication Critical patent/US20100015711A1/en
Assigned to CENTOCOR ORTHO BIOTECH INC. reassignment CENTOCOR ORTHO BIOTECH INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIU, JIAJIAN, BONNET, PASCAL GHISLAIN ANDRE, DAVIS, JANET, PARMENTER, CHRISTINE
Priority to US13/434,370 priority patent/US9593305B2/en
Priority to US13/434,392 priority patent/US20120190112A1/en
Priority to US13/434,409 priority patent/US9593306B2/en
Assigned to JANSSEN BIOTECH, INC. reassignment JANSSEN BIOTECH, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CENTOCOR ORTHO BIOTECH INC.
Priority to US15/457,864 priority patent/US10351820B2/en
Priority to US15/457,893 priority patent/US10233421B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0676Pancreatic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0676Pancreatic cells
    • C12N5/0678Stem cells; Progenitor cells; Precursor cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/16Activin; Inhibin; Mullerian inhibiting substance
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/165Vascular endothelial growth factor [VEGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/19Growth and differentiation factors [GDF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/385Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/41Hedgehog proteins; Cyclopamine (inhibitor)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/42Notch; Delta; Jagged; Serrate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes
    • C12N2501/72Transferases (EC 2.)
    • C12N2501/727Kinases (EC 2.7.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/80Neurotransmitters; Neurohormones
    • C12N2501/845Gamma amino butyric acid [GABA]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/999Small molecules not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2531/00Microcarriers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the present invention is directed to methods to differentiate pluripotent stem cells.
  • the present invention is directed to methods and compositions to differentiate pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage comprising culturing the pluripotent stem cells in medium comprising a sufficient amount of GDF-8 to cause the differentiation of the pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage.
  • ⁇ cells insulin-producing cells
  • ⁇ cells appropriate for engraftment.
  • One approach is the generation of functional ⁇ cells from pluripotent stem cells, such as, for example, embryonic stem cells.
  • a pluripotent cell gives rise to a group of cells comprising three germ layers (ectoderm, mesoderm, and endoderm) in a process known as gastrulation.
  • Tissues such as, for example, thyroid, thymus, pancreas, gut, and liver, will develop from the endoderm, via an intermediate stage.
  • the intermediate stage in this process is the formation of definitive endoderm.
  • Definitive endoderm cells express a number of markers, such as, for example, HNF-3beta, GATA4, MIXL1, CXCR4 and SOX17.
  • pancreas arises from the differentiation of definitive endoderm into pancreatic endoderm.
  • Cells of the pancreatic endoderm express the pancreatic-duodenal homeobox gene, PDX1.
  • PDX1 expression marks a critical step in pancreatic organogenesis.
  • the mature pancreas contains, among other cell types, exocrine tissue and endocrine tissue. Exocrine and endocrine tissues arise from the differentiation of pancreatic endoderm.
  • islet cells bearing the features of islet cells have reportedly been derived from embryonic cells of the mouse.
  • Lumelsky et al. (Science 292:1389, 2001) report differentiation of mouse embryonic stem cells to insulin-secreting structures similar to pancreatic islets.
  • Soria et al. (Diabetes 49: 157, 2000) report that insulin-secreting cells derived from mouse embryonic stem cells normalize glycemia in streptozotocin-induced diabetic mice.
  • Hori et al. discloses that treatment of mouse embryonic stem cells with inhibitors of phosphoinositide 3-kinase (LY294002) produced cells that resembled ⁇ cells.
  • Blyszczuk et al. reports the generation of insulin-producing cells from mouse embryonic stem cells constitutively expressing Pax4.
  • retinoic acid can regulate the commitment of embryonic stem cells to form Pdx1 positive pancreatic endoderm. Retinoic acid is most effective at inducing Pdx1 expression when added to cultures at day 4 of embryonic stem cell differentiation during a period corresponding to the end of gastrulation in the embryo (Diabetes 54:301, 2005).
  • Miyazaki et al. reports a mouse embryonic stem cell line over-expressing Pdx1. Their results show that exogenous Pdx1 expression clearly enhanced the expression of insulin, somatostatin, glucokinase, neurogenin3, p48, Pax6, and HNF6 genes in the resulting differentiated cells (Diabetes 53: 1030, 2004).
  • mouse model of embryonic stem cell development may not exactly mimic the developmental program in higher mammals, such as, for example, humans.
  • D'Amour et al. describes the production of enriched cultures of human embryonic stem cell-derived definitive endoderm in the presence of a high concentration of activin and low serum (D'Amour K A et al. 2005). Transplanting these cells under the kidney capsule of mice resulted in differentiation into more mature cells with characteristics of some endodermal organs. Human embryonic stem cell-derived definitive endoderm cells can be further differentiated into PDX1 positive cells after addition of FGF-10 (US 2005/0266554A1).
  • D'Amour et al. states: “We have developed a differentiation process that converts human embryonic stem (hES) cells to endocrine cells capable of synthesizing the pancreatic hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin. This process mimics in vivo pancreatic organogenesis by directing cells through stages resembling definitive endoderm, gut-tube endoderm, pancreatic endoderm and endocrine precursor en route to cells that express endocrine hormones.”
  • hES human embryonic stem
  • Fisk et al. reports a system for producing pancreatic islet cells from human embryonic stem cells (US2006/0040387A1).
  • the differentiation pathway was divided into three stages. Human embryonic stem cells were first differentiated to endoderm using a combination of n-butyrate and activin A. The cells were then cultured with TGF- ⁇ antagonists such as Noggin in combination with EGF or betacellulin to generate PDX1 positive cells. The terminal differentiation was induced by nicotinamide.
  • Benvenistry et al. states: “We conclude that over-expression of PDX1 enhanced expression of pancreatic enriched genes, induction of insulin expression may require additional signals that are only present in vivo” (Benvenistry et al, Stem Cells 2006; 24:1923-1930).
  • Activin A is a TGF-beta family member that exhibits a wide range of biological activities including regulation of cellular proliferation and differentiation, and promotion of neuronal survival. Isolation and purification of activin A is often complex and can often result in poor yields.
  • Pangas, S. A. and Woodruff, T. K states: “Inhibin and activin are protein hormones with diverse physiological roles including the regulation of pituitary FSH secretion. Like other members of the transforming growth factor- ⁇ gene family, they undergo processing from larger precursor molecules as well as assembly into functional dimers. Isolation of inhibin and activin from natural sources can only produce limited quantities of bioactive protein.” (J. Endocrinol. 172 (2002) 199-210).
  • Arai, K. Y. et al states: “Activins are multifunctional growth factors belonging to the transforming growth factor- ⁇ superfamily. Isolation of activins from natural sources requires many steps and only produces limited quantities. Even though recombinant preparations have been used in recent studies, purification of recombinant activins still requires multiple steps.” (Protein Expression and Purification 49 (2006) 78-82).
  • the present invention provides a method to differentiate pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage, comprising culturing the pluripotent stem cells in medium comprising a sufficient amount of GDF-8 to cause the differentiation of the pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage.
  • the medium comprising a sufficient amount of GDF-8 also contains at least one other compound.
  • the at least one other compound is an aniline-pyridinotriazine.
  • the at least one other compound is a cyclic aniline-pyridinotriazine.
  • FIG. 1 shows the differentiation of HI human embryonic stem cells into cells expressing markers characteristic of the definitive endoderm lineage. Differentiation was determined by measuring cell number (Panel A) and SOX17 intensity (Panel B) using an IN Cell Analyzer 1000 (GE Healthcare). Human embryonic stem cells were treated for a total of four days with medium containing 20 ng/ml Wnt3a plus activin A at the concentrations indicated (black bars) or medium lacking Wnt3a but with activin A at the concentrations indicated (white bars).
  • FIG. 2 shows the dose response relationship of activin A and GDF8 used to differentiate cells of the human embryonic stem cell line H1 toward cells expressing markers characteristic of the definitive endoderm lineage.
  • Cells were treated for a total of three days with activin A or GDF8 at the concentrations shown in combination with 20 ng/ml Wnt3a on the first day of assay. Differentiation was determined by measuring SOX17 intensity using a fluorescent antibody probe and high content analysis on a GE Healthcare IN Cell Analyzer.
  • FIG. 3 shows the expression of CXCR4 in cells following the first step of differentiation, according to the methods described in Example 12.
  • H1 cells were treated with 100 ng/ml activin A or 200 ng/ml GDF-8 for a total of three days in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 34 or 2.5 ⁇ M Compound 56 for all three days.
  • CXCR4 expression was measured using a fluorescent antibody probe and flow cytometry, yielding the percentages of positive cells shown.
  • FIG. 4 shows the expression of SOX17 in cells after three days differentiation to definitive endoderm according to the methods described in Example 12.
  • H1 cells were treated for a total of three days with 100 ng/ml activin A or 200 ng/ml GDF-8 in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 34 or 2.5 ⁇ M Compound 56 for all three days.
  • Differentiation was determined by measuring SOX17 intensity (black bars) and resulting cell number (white bars) with fluorescent antibody probes and high content analysis on a GE Healthcare IN Cell Analyzer.
  • FIG. 5 shows the expression of PDX1 and CDX2 protein in cells following the third step of differentiation, according to the methods described in Example 12.
  • H1 cells were treated for a total of three days with 100 ng/ml activin A or 200 ng/ml GDF-8 in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 34 or 2.5 ⁇ M Compound 56 for all three days followed by subsequent differentiation through the second and third steps of differentiation.
  • Protein expression and cell numbers are depicted for each treatment group. For comparative purposes, values are normalized relative to treatment with activin A/Wnt3a.
  • FIG. 6 shows the expression of PDX1 protein (white bars) and cell number (black bars) in cells following the fourth step of differentiation, according to the methods described in Example 12.
  • H1 cells were treated for a total of three days with 100 ng/ml activin A or 200 ng/ml GDF-8 in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 34 or 2.5 ⁇ M Compound 56 for all three days followed by subsequent differentiation through the second, third, and fourth steps of differentiation.
  • Protein expression and cell numbers as determined with fluorescent antibody probes and high content analysis, are depicted for each treatment group. For comparative purposes, values are normalized relative to treatment with activin A/Wnt3a.
  • FIG. 7 shows the protein expression for insulin and glucagon and cell number in cells differentiated according to the methods described in Example 12.
  • H1 cells were treated for a total of three days with 100 ng/ml activin A or 200 ng/ml GDF-8 in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 34 or 2.5 ⁇ M Compound 56 for all three days followed by subsequent differentiation through the second, third, fourth, and fifth steps of differentiation.
  • Protein expression and cell numbers, as determined with fluorescent antibody probes and high content analysis, are depicted for each treatment group. For comparative purposes, values are normalized relative to treatment with activin A/Wnt3a.
  • FIG. 8 shows SOX17 protein expression and cell number in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 13.
  • H1 cells were treated for a total of four days with 100 ng/ml of activin A or 100 ng/ml of a GDF-growth factor in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 34 or 2.5 ⁇ M Compound 56 for the first two days of assay.
  • SOX17 protein expression black bars
  • cell numbers white bars
  • values are normalized relative to treatment with activin A/Wnt3a.
  • Panel 8A shows a series of control conditions for differentiation in the absence of any growth factors (NONE), or with activin A/Wnt3a treatment (AA/Wnt3a) or with individual reagents alone.
  • Panel 8B shows differentiation with GDF-3, alone or in multiple combinations with Wnt3a, Compound 34, or Compound 56.
  • Panel 8C shows differentiation with GDF-5, alone or in multiple combinations with Wnt3a, Compound 34, or Compound 56.
  • Panel 8D shows differentiation with GDF-8, alone or in multiple combinations with Wnt3a, Compound 34, or Compound 56.
  • Panel 8E shows differentiation with GDF-10, alone or in multiple combinations with Wnt3a, Compound 34, or Compound 56.
  • Panel 8F shows differentiation with GDF-11, alone or in multiple combinations with Wnt3a, Compound 34, or Compound 56.
  • Panel 8G shows differentiation with GDF-15, alone or in multiple combinations with Wnt3a, Compound 34, or Compound 56.
  • FIG. 9 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 14.
  • H1 cells were treated for a total of three days with 100 ng/ml of activin A or various growth factors at the concentrations shown in combination with 20 ng/ml Wnt3a or 2.5 ⁇ M Compound 34 for the first day of assay.
  • SOX17 protein expression black bars
  • cell numbers white bars
  • values are normalized relative to treatment with activin A/Wnt3a.
  • Panel 9A shows a series of control conditions for differentiation with Wnt3a alone or in the absence of any growth factors (None) or with activin A/Wnt3a treatment (AA/Wnt3a).
  • Panel 9B shows differentiation with GDF-8 (Vendor PeproTech), at the concentrations shown, in combination with 20 ng/ml Wnt3a.
  • Panel 9C shows differentiation with GDF-8 (Vendor Shenendoah), at the concentrations shown, in combination with 20 ng/ml Wnt3a.
  • Panel 9D shows differentiation with TGF ⁇ 1, at the concentrations shown, in multiple combinations with Wnt3a or Compound 34.
  • Panel 9E shows differentiation with BMP2, at the concentrations shown, in multiple combinations with Wnt3a or Compound 34.
  • Panel 9F shows differentiation with BMP3, at the concentrations shown, in multiple combinations with Wnt3a or Compound 34.
  • Panel 9G shows differentiation with BMP4, at the concentrations shown, in multiple combinations with Wnt3
  • FIG. 10 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15. H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of activin A or 100 ng/ml GDF-8 in combination with 20 ng/ml Wnt3a.
  • SOX17 protein expression as determined with fluorescent antibody probes and high content analysis, is shown as total intensity values for each treatment group, testing control conditions for differentiation with no growth factors added (no treatment), with Wnt3a alone, with activin A or GDF-8 alone, or with activin A/Wnt3a treatment or GDF-8/Wnt3a treatment, where Wnt3a was added only for the first day of assay or for all three days of assay as shown.
  • FIG. 11 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of activin A in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19(Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added only on the first day of assay.
  • Protein expression for SOX17 is depicted by total intensity values.
  • FIG. 12 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of activin A in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added for all three days of assay.
  • Protein expression for SOX17 is depicted by total intensity values.
  • FIG. 13 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of GDF-8 in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added only on the first day of assay.
  • Protein expression for SOX17 is depicted by total intensity values.
  • FIG. 14 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of GDF-8 in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added for all three days of assay.
  • Protein expression for SOX17 is depicted by total intensity values.
  • FIG. 15 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of activin A or 100 ng/ml GDF-8 in combination with 20 ng/ml Wnt3a.
  • Cell numbers, as determined with a fluorescent nuclear probe and high content analysis, are shown for each treatment group, testing control conditions for differentiation with no growth factors added (no treatment), with Wnt3a alone, with activin A or GDF-8 alone, or with activin A/Wnt3a treatment or GDF-8/Wnt3a treatment, where Wnt3a was added only for the first day of assay or for all three days of assay as shown.
  • FIG. 16 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of activin A in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added only on the first day of assay. Cell number yields, as determined with a fluorescent nuclear probe and high content analysis, are shown.
  • FIG. 17 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of activin A in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added for all three days of assay. Cell number yields, as determined with a fluorescent nuclear probe and high content analysis, are shown.
  • FIG. 18 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of GDF-8 in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added only on the first day of assay. Cell number yields, as determined with a fluorescent nuclear probe and high content analysis, are shown.
  • FIG. 19 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15.
  • H1 cells were treated for a total of three days in various timed exposures with 100 ng/ml of GDF-8 in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19 (Panel C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added for all three days of assay. Cell number yields, as determined with a fluorescent nuclear probe and high content analysis, are shown.
  • FIG. 20 shows the expression of various protein markers in cells throughout multiple steps of differentiation according to the methods described in Example 16. H1 cells were treated with 100 ng/ml activin A or 100 ng/ml GDF-8 for a total of three days in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M various compounds (Compound 19, Compound 202, Compound 40, or GSK3 inhibitor BIO) added only on the first day.
  • FIG. 20 panel A shows FACS analysis for the definitive endoderm marker, CXCR4, in cells after the first step of differentiation. CXCR4 expression was measured using a fluorescent antibody probe and flow cytometry, yielding the percentages of positive cells as shown.
  • FIG. 20 shows the expression of various protein markers in cells throughout multiple steps of differentiation according to the methods described in Example 16. H1 cells were treated with 100 ng/ml activin A or 100 ng/ml GDF-8 for a total of three days in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M various
  • panel B shows high content image analysis for normalized SOX17 protein expression (black bars) and recovered cell numbers (white bars) resulting from the first step of differentiation, testing the corresponding treatments shown.
  • panel C shows high content image analysis for relative cell numbers recovered from cultures treated through differentiation step 5.
  • panel D shows high content image analysis for glucagon protein expression from cultures treated through differentiation step 5.
  • panel E shows high content image analysis for insulin protein expression from cultures treated through differentiation step 5.
  • panel F shows the ratio of glucagon to insulin expression in cells from cultures treated through differentiation step 5.
  • expression values in panels B, C, D, E, and F are normalized to the control treatment with activin A and Wnt3a during step 1.
  • FIG. 21 shows the expression of various protein and RT-PCR markers in cells throughout multiple steps of differentiation according to the methods described in Example 17.
  • H1 cells were treated with 100 ng/ml activin A or 100 ng/ml GDF-8 for a total of three days in combination with 20 ng/ml Wnt3a for the first day or various compounds at the following concentrations (Compound 181, Compound 180, Compound 19, Compound 202, Compound 40, Compound 56, or GSK3 inhibitor BIO) added only on the first day.
  • FACS analysis for the definitive endoderm marker, CXCR4 is shown in cells after the first step of differentiation where treatment combined activin A (Panel A) or GDF-8 (Panel B) with Wnt3a or various compounds.
  • CXCR4 expression was measured using a fluorescent antibody probe and flow cytometry, yielding the percentages of positive cells as shown.
  • normalized RT-PCR values for various differentiation markers are shown with respective treatments using activin A or GDF-8 during the first step of differentiation as follows: markers at the end of step one of differentiation for treatments combining activin A (Panel C) or GDF-8 (Panel D); markers at the end of step three of differentiation for treatments combining activin A (Panel E) or GDF-8 (Panel F); markers at the end of step four of differentiation for treatments combining activin A (Panel G) or GDF-8 (Panel H); markers at the end of step five of differentiation for treatments combining activin A (Panel I) or GDF-8 (Panel J).
  • step five of differentiation high content analysis was performed to measure recovered cell numbers for corresponding treatments during the first step of differentiation using activin A (Panel K) or GDF-8 (Panel M). High content analysis was also used to measure glucagon and insulin intensity in recovered cell populations at the end of step five of differentiation, corresponding to treatment with activin A (Panel L) or GDF-8 (Panel N) during the first step of differentiation.
  • FIG. 22 shows the expression of various protein and RT-PCR markers in cells treated according to the methods described in Example 18. H1 cells were treated with 100 ng/ml activin A or 100 ng/ml GDF-8 for a total of three days in combination with 20 ng/ml Wnt3a for the first day or 2.5 ⁇ M Compound 40 or 2.5 ⁇ M Compound 202 only on the first day.
  • panel A shows FACS analysis for the definitive endoderm marker, CXCR4, in cells after the first step of differentiation. CXCR4 expression was measured using a fluorescent antibody probe and flow cytometry, yielding the percentages of positive cells as shown.
  • panel B normalized RT-PCR values for various differentiation markers in cells recovered after the fourth step of differentiation are shown corresponding to respective treatments using activin A/Wnt3a or GDF-8/Compound 40 or GDF-8/Compound 202 during the first step of differentiation.
  • FIG. 23 shows the level of C-peptide detected in SCID-beige mice that received cells at the end of step four of the differentiation protocol as described in Example 18.
  • FIG. 24 panel A shows the expression of CXCR4, as determined by FACS in cells at the end of step one of the differentiation protocol described in Example 19.
  • Panel B shows the expression of various genes, as determined by RT-PCR in cells at the end of step four of the differentiation protocol described in Example 19. Two different experimental replicates are shown (Rep-1 and Rep-2), each subjected to identical treatment protocols.
  • Panel C shows the level of C-peptide detected in SCID-beige mice that received cells at the end of step four of the differentiation protocol as treated with GDF-8 and Wnt3a during the first step of in vitro differentiation.
  • Panel D shows the level of C-peptide detected in SCID-beige mice that received cells at the end of step four of the differentiation protocol as treated with GDF-8 and Compound 28 during the first step of in vitro differentiation.
  • FIG. 25 shows the cell number (panel A) and expression of CXCR4 (panel B) from cells grown on microcarrier beads, treated according to the methods of the present invention as described in Example 22.
  • Cells were grown on Cytodex3 beads without treatment (undifferentiated) or with treatment combining 100 ng/ml activin A with 20 ng/ml Wnt3a (AA/Wnt3a) or with various treatments combining GDF-8 as shown: 50 ng/ml GDF-8with 2.5 ⁇ M Compound 34 (Cmp 34+8); or 50 ng/ml GDF-8 with 2.5 ⁇ M Compound 34 and 50 ng/ml PDGF (Cmp 34+8+D); or 50 ng/ml GDF-8 with 2.5 ⁇ M Compound 34 and 50 ng/ml PDGF and 50 ng/ml VEGF (Cmp 34+8+D+V); or 50 ng/ml GDF-8 with 2.5 ⁇ M Compound 34 and 50 ng/ml PDGF and 50
  • FIG. 26 shows the proliferation of cells following treatment of the compounds of the present invention as described in Example 23.
  • Panels B through I show assay results for treatment using a compound in combination with GDF-8 and measuring MTS OD readings at 1 day, 2 days, and 3 days after initiating the differentiation assay.
  • FIG. 27 shows the expression of various proteins and genes from cells grown on microcarrier beads, treated according to the methods of the present invention.
  • Panel A shows the percent positive expression of CXCR4, CD99, and CD9 as determined by FACS in cells at the end of step one of the differentiation protocol described in Example 24.
  • Panel B shows cells recovered from treatments as shown differentiated through step three of the differentiation protocol.
  • Panel C shows ddCT values for various gene markers expressed in cells treated as shown in step and differentiated through step three of the protocol.
  • 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 terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation and to contribute substantially to most, if not all, tissues following injection into blastocysts.
  • Stem cells are classified by their developmental potential as: (1) totipotent, meaning able to give rise to all embryonic and extraembryonic cell types; (2) pluripotent, meaning able to give rise to all embryonic cell types; (3) multipotent, meaning able to give rise to a subset of cell lineages but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem 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); (4) oligopotent, meaning able to give rise to a more restricted subset of cell lineages than multipotent stem cells; and (5) unipotent, meaning able to give rise to a single cell lineage (e.g., spermatogenic stem cells).
  • HSC hematopoietic stem cells
  • 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 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 of a cell.
  • the term “committed”, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway 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.
  • De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell.
  • the lineage of a cell defines the heredity of the cell, i.e., which cells it came 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.
  • ⁇ -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, NKX2.2, NKX6.1, NEUROD, ISL1, HNF-3 beta, MAFA, PAX4, or PAX6.
  • Cells expressing markers characteristic of the ⁇ cell lineage include ⁇ cells.
  • Cells expressing markers characteristic of the definitive endoderm lineage refers to cells expressing at least one of the following markers: SOX17, GATA4, HNF-3 beta, GSC, CER1, Nodal, FGF8, Brachyury, Mix-like homeobox protein, FGF4 CD48, eomesodermin (EOMES), DKK4, FGF17, GATA6, CXCR4, C-Kit, CD99, or OTX2.
  • Cells expressing markers characteristic of the definitive endoderm lineage include primitive streak precursor cells, primitive streak cells, mesendoderm cells and definitive endoderm cells.
  • Cells expressing markers characteristic of the pancreatic endoderm lineage refers to cells expressing at least one of the following markers: PDX1, HNF-1 beta, PTF1 alpha, HNF6, or HB9.
  • Cells expressing markers characteristic of the pancreatic endoderm lineage include pancreatic endoderm cells, primitive gut tube cells, and posterior foregut cells.
  • Cells expressing markers characteristic of the pancreatic endocrine lineage refers to cells expressing at least one of the following markers: NGN3, NEUROD, ISL1, PDX1, NKX6.1, PAX4, or PTF-1 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 ⁇ -cell lineage.
  • Definitive endoderm 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 endoderm cells express the following markers: HNF-3 beta, GATA4, SOX-17, Cerberus, OTX2, goosecoid, C-Kit, CD99, or MIXL1.
  • Extraembryonic endoderm refers to a population of cells expressing at least one of the following markers: SOX7, AFP, or SPARC.
  • Markers are nucleic acid or polypeptide molecules that are differentially expressed in a cell of interest.
  • 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 sufficiently higher 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.
  • Mesendoderm cell refers to a cell expressing at least one of the following markers: CD48, eomesodermin (EOMES), SOX17, DKK4, HNF-3 beta, GSC, FGF17, or GATA-6.
  • Pantendocrine 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.
  • “Pancreatic endoderm cell”, or “Stage 4 cells”, or “Stage 4”, as used herein, refers to a cell capable of expressing at least one of the following markers: NGN3, NEUROD, ISL1, PDX1, PAX4, or NKX2.2.
  • Pantenatic hormone producing cell refers to a cell capable of producing at least one of the following hormones: insulin, glucagon, somatostatin, and pancreatic polypeptide.
  • Pantix hormone secreting cell refers to a cell capable of secreting at least one of the following hormones: insulin, glucagon, somatostatin, and pancreatic polypeptide.
  • Posterior foregut cell or “Stage 3 cells”, or “Stage 3”, as used herein, refers to a cell capable of secreting at least one of the following markers: PDX1, HNF1, PTF-1 alpha, HNF6, HB-9, or PROX-1.
  • Pre-primitive streak cell refers to a cell expressing at least one of the following markers: Nodal, or FGF8.
  • Primary gut tube cell or “Stage 2 cells”, or “Stage2”, as used herein, refers to a cell capable of secreting at least one of the following markers: HNF1, HNF-4 alpha.
  • Primary streak cell refers to a cell expressing at least one of the following markers: Brachyury, Mix-like homeobox protein, or FGF4.
  • pluripotency of pluripotent stem cells can be confirmed, for example, by injecting cells into severe combined immunodeficient (SCID) mice, fixing the teratomas that form using 4% paraformaldehyde, and then examining them histologically for evidence of cell types from the three germ layers.
  • 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.
  • Propagated pluripotent 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 karyotype,” which means that the cells are euploid, wherein all human chromosomes are present and not noticeably altered.
  • pluripotent stem cells 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 to 12 weeks gestation.
  • 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 to 12 weeks gestation.
  • Non-limiting examples are established lines of human embryonic stem cells or human embryonic germ cells, such as, for example, the human embryonic stem cell lines H1, H7, and H9 (WiCell).
  • 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.
  • cells taken from a pluripotent stem cell population already cultured in the absence of feeder cells are also suitable are mutant human embryonic stem cell lines,
  • 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.S.A. 92:7844, 1995).
  • pluripotent stem cells are prepared as described by Takahashi et al. (Cell 131: 1-12, 2007).
  • pluripotent stem cells are typically cultured on a layer of feeder cells that support the pluripotent stem cells in various ways.
  • 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-free culture without differentiation is supported using a medium conditioned by culturing previously with another cell type. Altematively, the growth of pluripotent stem cells in feeder-free culture without differentiation is supported using a chemically defined medium.
  • the pluripotent stem cells may be plated onto a suitable culture substrate.
  • the suitable culture substrate is an extracellular matrix component, such as, for example, those derived from basement membrane or that may form part of adhesion molecule receptor-ligand couplings.
  • the suitable culture substrate is MATRIGEL® (Becton Dickenson).
  • MATRIGEL® is a soluble preparation from Engelbreth-Holm-Swarm tumor cells that gels at room temperature to form a reconstituted basement membrane.
  • extracellular matrix components and component mixtures are suitable as an alternative. Depending on the cell type being proliferated, this may include laminin, fibronectin, proteoglycan, entactin, heparan sulfate, and the like, alone or in various combinations.
  • the pluripotent stem cells may be plated onto the substrate in a suitable distribution and in the presence of a medium that promotes cell survival, propagation, and retention of the desirable characteristics. All these characteristics benefit from careful attention to the seeding distribution and can readily be determined by one of skill in the art.
  • Suitable culture media may be made from the following components, such as, for example, Dulbecco's modified Eagle's medium (DMEM), Gibco #11965-092; Knockout Dulbecco's modified Eagle's medium (KO DMEM), Gibco #10829-018; Ham's F12/50% DMEM basal medium; 200 mM L-glutamine, Gibco #15039-027; non-essential amino acid solution, Gibco 11140-050; ⁇ -mercaptoethanol, Sigma #M7522; human recombinant basic fibroblast growth factor (bFGF), Gibco #13256-029.
  • DMEM Dulbecco's modified Eagle's medium
  • KO DMEM Knockout Dulbecco's modified Eagle's medium
  • Ham's F12/50% DMEM basal medium 200 mM L-glutamine, Gibco #15039-027; non-essential amino acid solution, Gibco 11140-050; ⁇ -mercaptoethanol, Sigma
  • the present invention provides a method for producing pancreatic hormone producing cells from pluripotent stem cells, comprising the steps of:
  • the pancreatic endocrine cell is a pancreatic hormone producing cell.
  • the pancreatic endocrine cell is a cell expressing markers characteristic of the ⁇ -cell lineage.
  • a cell expressing markers characteristic of the ⁇ -cell lineage expresses PDX1 and at least one of the following transcription factors: NGN3, NKX2.2, NKX6.1, NEUROD, ISL1, HNF-3 beta, MAFA, PAX4, or Pax6.
  • a cell expressing markers characteristic of the ⁇ -cell lineage is a ⁇ -cell.
  • Pluripotent stem cells suitable for use in the present invention include, for example, the human embryonic stem cell line H9 (NIH code: WA09), the human embryonic stem cell line H1 (NIH code: WA01), the human embryonic stem cell line H7 (NIH code: WA07), and the human embryonic stem cell line SA002 (Cellartis, Sweden). Also suitable for use in the present invention are cells that express at least one of the following markers characteristic of pluripotent cells: ABCG2, cripto, CD9, FOXD3, Connexin43, Connexin45, OCT4, SOX2, Nanog, hTERT, UTF-1, ZFP42, SSEA-3, SSEA-4, Tral-60, or Tral-81.
  • markers characteristic of pluripotent cells ABCG2, cripto, CD9, FOXD3, Connexin43, Connexin45, OCT4, SOX2, Nanog, hTERT, UTF-1, ZFP42, SSEA-3, SSEA-4, Tral-60, or Tral
  • the pluripotent stem cells may be cultured on a feeder cell layer.
  • the pluripotent stem cells may be cultured on an extracellular matrix.
  • the extracellular matrix may be a solubilized basement membrane preparation extracted from mouse sarcoma cells (as sold by BD Biosciences under the trade name MATRIGELTM).
  • the extracellular matrix may be growth factor-reduced MATRIGELTM.
  • the extracellular matrix may be fibronectin.
  • the pluripotent stem cells are cultured and differentiated on tissue culture substrate coated with human serum.
  • the extracellular matrix may be diluted prior to coating the tissue culture substrate.
  • suitable methods for diluting the extracellular matrix and for coating the tissue culture substrate may be found in Kleinman, H. K., et al., Biochemistry 25:312 (1986), and Hadley, M. A., et al., J. Cell. Biol. 101:1511 (1985).
  • the extracellular matrix is MATRIGELTM.
  • the tissue culture substrate is coated with MATRIGELTM at a 1:10 dilution. In an alternate embodiment, the tissue culture substrate is coated with MATRIGELTM at a 1:15 dilution. In an alternate embodiment, the tissue culture substrate is coated with MATRIGELTM at a 1:30 dilution. In an alternate embodiment, the tissue culture substrate is coated with MATRIGELTM at a 1:60 dilution.
  • the extracellular matrix is growth factor-reduced MATRIGELTM.
  • the tissue culture substrate is coated with growth factor-reduced MATRIGELTM at a 1:10 dilution. In an alternate embodiment, the tissue culture substrate is coated with growth factor-reduced MATRIGELTM at a 1:15 dilution. In an alternate embodiment, the tissue culture substrate is coated with growth factor-reduced MATRIGELTM at a 1:30 dilution. In an alternate embodiment, the tissue culture substrate is coated with growth factor-reduced MATRIGELTM at a 1:60 dilution.
  • Markers characteristic of the definitive endoderm lineage are selected from the group consisting of SOX17, GATA4, HNF-3 beta, GSC, CER1, Nodal, FGF8, Brachyury, Mix-like homeobox protein, FGF4 CD48, eomesodermin (EOMES), DKK4, FGF17, GATA6, CXCR4, C-Kit, CD99, and OTX2.
  • Suitable for use in the present invention is a cell that expresses at least one of the markers characteristic of the definitive endoderm lineage.
  • a cell expressing markers characteristic of the definitive endoderm lineage is a primitive streak precursor cell.
  • a cell expressing markers characteristic of the definitive endoderm lineage is a mesendoderm cell.
  • a cell expressing markers characteristic of the definitive endoderm lineage is a definitive endoderm cell.
  • Markers characteristic of the pancreatic endoderm lineage are selected from the group consisting of PDX1, HNF-1 beta, PTF1 alpha, HNF6, HB9 and PROX1.
  • Suitable for use in the present invention is a cell that expresses at least one of the markers characteristic of the pancreatic endoderm lineage.
  • a cell expressing markers characteristic of the pancreatic endoderm lineage is a pancreatic endoderm cell.
  • 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.
  • 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.
  • the pancreatic endocrine cell may be a pancreatic hormone secreting cell.
  • pluripotent stem cells may be differentiated into cells expressing markers characteristic of the definitive endoderm lineage by culturing the pluripotent stem cells in medium comprising a sufficient amount of GDF-8 to cause the differentiation of the pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day to about seven days.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day to about six days.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day to about five days.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day to about four days.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day to about three days.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day to about two days.
  • the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day.
  • the GDF-8 is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the GDF-8 is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the GDF-8 is used at a concentration from about 5 ng/ml to about 25 ng/ml. In an alternate embodiment, the GDF-8 is used at a concentration of about 25 ng/ml.
  • the medium comprising a sufficient amount of GDF-8 also contains at least one other factor.
  • the at least one other factor is selected from the group consisting of: EGF, FGF4, PDGF-A, PDGF-B, PDGF-C, PDGF-D, VEGF, muscimol, PD98059, LY294002, U0124, U0126, and sodium butyrate.
  • the EGF is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the EGF is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the EGF is used at a concentration of about 50 ng/ml.
  • the FGF4 is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the FGF4 is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the FGF4 is used at a concentration of about 50 ng/ml.
  • the PDGF-A is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the PDGF-A is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the PDGF-A is used at a concentration of about 50 ng/ml.
  • the PDGF-B is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the PDGF-B is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the PDGF-B is used at a concentration of about 50 ng/ml.
  • the PDGF-C is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the PDGF-C is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the PDGF-C is used at a concentration of about 50 ng/ml.
  • the PDGF-D is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the PDGF-D is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the PDGF-D is used at a concentration of about 50 ng/ml.
  • the VEGF is used at a concentration from about 5 ng/ml to about 500 ng/ml. In an alternate embodiment, the VEGF is used at a concentration from about 5 ng/ml to about 50 ng/ml. In an alternate embodiment, the VEGF is used at a concentration of about 50 ng/ml.
  • the muscimol is used at a concentration from about 1 ⁇ M to about 200 ⁇ M. In an alternate embodiment, the muscimol is used at a concentration from about 1 ⁇ M to about 20 ⁇ M. In an alternate embodiment, the muscimol is used at a concentration of about 20 ⁇ M.
  • the PD98059 is used at a concentration from about 0.1 ⁇ M to about 10 ⁇ M. In an alternate embodiment, the PD98059 is used at a concentration from about 0.1 ⁇ M to about 1 ⁇ M. In an alternate embodiment, the PD98059 is used at a concentration of about 1 ⁇ M.
  • the LY294002 is used at a concentration from about 0.25 ⁇ M to about 25 ⁇ M. In an alternate embodiment, the LY294002 is used at a concentration from about 0.25 ⁇ M to about 2.5 ⁇ M. In an alternate embodiment, the LY294002 is used at a concentration of about 2.5 ⁇ M.
  • the U0124 is used at a concentration from about 0.1 ⁇ M to about 10 ⁇ M. In an alternate embodiment, the U0124 is used at a concentration from about 0.1 ⁇ M to about 1 ⁇ M. In an alternate embodiment, the U0124 is used at a concentration of about 1 ⁇ M.
  • the U0126 is used at a concentration from about 0.1 ⁇ M to about 10 ⁇ M. In an alternate embodiment, the U0126 is used at a concentration from about 0.1 ⁇ M to about 1 ⁇ M. In an alternate embodiment, the U0126 is used at a concentration of about 1 ⁇ M.
  • the sodium butyrate is used at a concentration from about 0.05 ⁇ M to about 5 ⁇ M. In an alternate embodiment, the sodium butyrate is used at a concentration from about 0.05 ⁇ M to about 0.5 ⁇ M. In an alternate embodiment, the sodium butyrate is used at a concentration of about 0.5 ⁇ M.
  • the at least one other factor is selected from the group consisting of: an aniline-pyridinotriazine, a cyclic aniline-pyridinotriazine, N- ⁇ [1-(Phenylmethyl)azepan-4-yl]methyl ⁇ -2-pyridin-3-ylacetamide, 4- ⁇ [4-(4- ⁇ [2-(Pyridin-2-ylamino)ethyl]amino ⁇ -1,3,5-triazin-2-yl)pyridin-2-yl]oxy ⁇ butan-1-ol 3-( ⁇ 3-[4-( ⁇ 2-[Methyl(pyridin-2-yl)amino]ethyl ⁇ amino)-1,3,5-triazin-2-yl]pyridin-2-yl ⁇ amino)propan-1-ol, N ⁇ 4 ⁇ -[2-(3-Fluorophenyl)ethyl]-N ⁇ 2 ⁇ -[3-(4
  • the present invention provides compounds that are capable of differentiating pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage.
  • the compound that is capable of differentiating pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage is an aniline-pyridinotriazine of the Formula (1):
  • n represents an integer from 1 to 4
  • Z represents N or C
  • R 1 and R 8 each independently represent hydrogen, Het 14 , cyano, halo, hydroxy, C 1-6 alkoxy-, C 1-6 alkyl-, mono- or di(C 1-4 alkyl)amino-carbonyl-, mono- or di(C 1-4 alkyl)amino-sulfonyl, C 1-6 alkoxy-substituted with halo or R 1 represents C 1-6 alkyl substituted with one or where possible two or more substituents selected from hydroxy or halo;
  • R 2 and R 9 each independently represents hydrogen, C 1-4 alkyl, C 2-4 alkenyl, Het 3 , Het 4 -C 1-4 alkyl-, Het 5 -C 1-4 alkylcarbonyl-, mono- or di(C 1-4 alkyl)amino-C 1-4 alkyl-carbonyl- or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or C 1-4 alkyloxy-;
  • R 3 and R 7 each independently represent hydrogen, C 1-4 alkyl, Het 6 , Het 7 -C 1-4 alkyl-, C 2-4 alkenylcarbonyl-optionally substituted with Het 8 -C 1-4 alkylaminocarbonyl-, C 2-4 alkenylsulfonyl-, C 1-4 alkyloxyC 1-4 alkyl- or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or C 1-4 alkyloxy-;
  • R4, R5, R6 and R 10 each independently represent hydrogen or C 1-4 alkyl optionally substituted with hydroxy, Het 9 or C 1-4 alkyloxy;
  • Het 1 and Het 2 each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het 1 and Het 2 are optionally substituted with amino, hydroxy, C 1-4 alkyl, hydroxy-C 1-4 allcyl-, phenyl, phenyl-C 1-4 alkyl-, C 1-4 alkyl-oxy-C 1-4 alkyl-mono- or di(C 1-4 alkyl) amino- or amino-carbonyl-;
  • Het 3 and Het 6 each independently represent, heterocycle selected from pyrrolidinyl or piperidinyl wherein said Het 3 and Het 6 are optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-;
  • Het 4 , Het 7 and Het 9 each independently represent a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or piperidinyl wherein said Het 4 , Het 7 and Het 9 are optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-;
  • Het 5 represents a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or pipendinyl wherein said Het 5 is optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-;
  • Het 10 , Het 11 and Het 13 each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het 10 , Het 11 and Het 13 are optionally substituted with amino, hydroxy, C 1-4 alkyl, hydroxy-C 1-4 alkyl-, phenyl, phenyl-C 1-4 alkyl-, C 1-4 alkyl-oxy-C 1-4 alkyl-, amino-carbonyl- or mono- or di(C 1-4 alkyl)amino-;
  • Het 12 represents a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het 12 is optionally substituted with amino, hydroxy, C 1-4 alkyl, hydroxy-C 1-4 alkyl-, phenyl, phenyl-C 1-4 alkyl-, C 1-4 alkyl-oxy-C 1-4 alkyl-; mono- or di(C 1-4 alkyl)amino- or amino-carbonyl-;
  • Het 14 represents a heterocycle selected from morpholinyl; pyrrolidinyl; piperazinyl; imidazolyl; pyrrolyl; 2,3,4-triazapyrrolyl; 1,2,3-triazolyl; pyrazolyl; or piperidinyl wherein said Het 14 is optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-; in particular Het 14 represents a heterocycle selected from morpholinyl; pyrrolidinyl; pyrrolyl; 2,3,4-triazapyrrolyl; piperazinyl or piperidinyl wherein said Het 14 is optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alky
  • the aniline-pyridinotriazine is a compound of the Formula (1).
  • the aniline-pyridinotriazine is a compound of the Formula (2).
  • the aniline-pyridinotriazine is a compound of the Formula (3).
  • the aniline-pyridinotriazine is a compound of the Formula (4).
  • the aniline-pyridinotriazine is a compound of the Formula (5).
  • the aniline-pyridinotriazine is a compound of the Formula (6).
  • the aniline-pyridinotriazine is a compound of the Formula (7).
  • the aniline-pyridinotriazine is a compound of the Formula (8).
  • the aniline-pyridinotriazine is a compound of the Formula (9).
  • the aniline-pyridinotriazine is a compound of the Formula (10).
  • the aniline-pyridinotriazine is a compound of the Formula (11).
  • the aniline-pyridinotriazine is a compound of the Formula (12).
  • the aniline-pyridinotriazine is a compound of the Formula (13).
  • the compound that is capable of differentiating pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage is a cyclic aniline-pyridinotriazine of the Formula (14):
  • n represents an integer from 1 to 4
  • Z represents N or C
  • Y represents —NR 2 —C 1-6 alkyl-CO—NR 4 —, —C 1-4 alkyl-NR 9 —C 1-4 alkyl-, C 1-6 alkyl-CO-Het 10 -, -Het 11 -CO—C 1-6 alkyl-, -Het 12 -C 1-6 alkyl-, —CO-Het 13 -C 1-6 alkyl-, —CO—NR 10 —C 1-6 alkyl-, -Het 1 -C 1-6 alkyl-CO—NR 5 —, or -Het 2 -CO—NR 6 — wherein the-C 1-6 alkyl-linker in —NR 2 —C 1-6 alkyl-CO—NR 4 — or -Het 1 -C 1-6 alkyl-CO—NR 5 — is optionally substituted with one or where possible two or more substituents selected from hydroxy, methoxy, aminocarbonyl, halo, phenyl, ind
  • X 1 represents a direct bond, C 1-4 alkyl, C 1-4 alkyloxy-, C 1-4 alkyl-CO—, C 2-4 alkenyl, C 2-4 alkynyl, or C 1-4 alkyl-NR 3 —, wherein said C 1-4 alkyl or C 2-4 alkenyl is optionally substituted with one or where possible two or more halo substituents;
  • X 2 represents a direct bond, C 1-4 alkyl, C 1-4 alkyloxy-, C 1-4 alkyl-CO—, C 2-4 alkenyl, C 2-4 alkynyl, or C 1-4 alkyl-NR 7 —, wherein said C 1-4 alkyl or C 2-4 alkenyl is optionally substituted with one or where possible two or more halo substituents;
  • R 1 and R 8 each independently represent hydrogen, Het 14 , cyano, halo, hydroxy, C 1-6 alkoxy-, C 1-6 alkyl-, mono- or di(C 1-4 alkyl)amino-carbonyl-, mono- or di(C 1-4 alkyl)amino-sulfonyl, C 1-6 alkoxy-substituted with halo or R 1 represents C 1-6 alkyl substituted with one or where possible two or more substituents selected from hydroxy or halo;
  • R 2 and R 9 each independently represents hydrogen, C 1-4 alkyl, C 2-4 alkenyl, Het 3 , Het 4 -C 1-4 alkyl-, Het 5 -C 1-4 alkylcarbonyl-, mono- or di(C 1-4 alkyl)amino-C 1-4 alkyl-carbonyl- or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or C 1-4 alkyloxy-;
  • R 3 and R 7 each independently represent hydrogen, C 1-4 alkyl, Het 6 , Het 7 -C 1-4 alkyl-, C 2-4 alkenylcarbonyl-optionally substituted with Het 8 -C 1-4 alkylaminocarbonyl-, C 2-4 alkenylsulfonyl-, C 1-4 alkyloxyC 1-4 alkyl- or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or C 1-4 alkyloxy-;
  • R4, R5, R6 and R 10 each independently represent hydrogen or C 1-4 alkyl optionally substituted with hydroxy, Het 9 or C 1-4 alkyloxy;
  • Het 1 and Het 2 each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het 1 and Het 2 are optionally substituted with amino, hydroxy, C 1-4 alkyl, hydroxy-C 1-4 allcyl-, phenyl, phenyl-C 1-4 alkyl-, C 1-4 alkyl-oxy-C 1-4 alkyl-mono- or di(C 1-4 alkyl) amino- or amino-carbonyl-;
  • Het 3 and Het 6 each independently represent, heterocycle selected from pyrrolidinyl or piperidinyl wherein said Het 3 and Het 6 are optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-;
  • Het 4 , Het 7 and Het 9 each independently represent a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or piperidinyl wherein said Het 4 , Het 7 and Het 9 are optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-;
  • Het 5 represents a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or pipendinyl wherein said Het 5 is optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-;
  • Het 10 , Het 11 and Het 13 each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het 10 , Het 11 and Het 13 are optionally substituted with amino, hydroxy, C 1-4 alkyl, hydroxy-C 1-4 alkyl-, phenyl, phenyl-C 1-4 alkyl-, C 1-4 alkyl-oxy-C 1-4 alkyl-, amino-carbonyl- or mono- or di(C 1-4 alkyl)amino-;
  • Het 12 represents a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het 12 is optionally substituted with amino, hydroxy, C 1-4 alkyl, hydroxy-C 1-4 alkyl-, phenyl, phenyl-C 1-4 alkyl-, C 1-4 alkyl-oxy-C 1-4 alkyl-; mono- or di(C 1-4 alkyl)amino- or amino-carbonyl-;
  • Het 14 represents a heterocycle selected from morpholinyl; pyrrolidinyl; piperazinyl; imidazolyl; pyrrolyl; 2,3,4-triazapyrrolyl; 1,2,3-triazolyl; pyrazolyl; or piperidinyl wherein said Het 14 is optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alkyl-, C 1-4 alkyloxyC 1-4 alkyl or polyhydroxy-C 1-4 alkyl-; in particular Het 14 represents a heterocycle selected from morpholinyl; pyrrolidinyl; pyrrolyl; 2,3,4-triazapyrrolyl; piperazinyl or piperidinyl wherein said Het 14 is optionally substituted with one or where possible two or more substituents selected from C 1-4 alkyl, C 3-6 cycloalkyl, hydroxy-C 1-4 alky
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (14).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (15).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (16).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (17).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (18).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (19).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (20).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (21).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (22).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (23).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (24).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (25).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (26).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (27).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (28).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (29).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (30).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (31).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (32).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (33).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (34).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (35).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (36).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (37).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (38).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (39).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (40).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (41).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (42).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (43).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (44).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (45).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (46).
  • the cyclic aniline-pyridinotriazine is a compound of the Formula (47).
  • the at least one other factor is a compound of the Formula (48):
  • the at least one other factor is a compound of the Formula (49):
  • the at least one other factor is a compound of the Formula (50):
  • the at least one other factor is a compound of the Formula (51):
  • the at least one other factor is a compound of the Formula (52):
  • the at least one other factor is a compound of the Formula (53):
  • the at least one other factor is a compound of the Formula (54):
  • the at least one other factor is a compound of the Formula (55):
  • the at least one other factor is a compound of the Formula (56):
  • the at least one other factor is a compound of the Formula (57):
  • the at least one other factor is a compound of the Formula (58):
  • the at least one other factor is a compound of the Formula (59):
  • 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. Pluripotent stem cells typically do not express such markers. Thus, differentiation of pluripotent cells is detected when cells begin to express them.
  • the efficiency of differentiation may be determined by exposing a treated cell population to an agent (such as an antibody) that specifically recognizes a protein marker expressed by cells expressing markers characteristic of the definitive endoderm lineage.
  • an agent such as an antibody
  • RT-PCR quantitative reverse transcriptase polymerase chain reaction
  • Northern blots in situ hybridization
  • immunoassays such as immunohistochemical analysis of sectioned material, Western blotting, and for markers that are accessible in intact cells, flow cytometry analysis (FACS) (see, e.g., Harlow and Lane, Using Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Laboratory Press (1998)).
  • pluripotent stem cell markers include, for example, the expression of one or more of the following: ABCG2, cripto, FOXD3, Connexin43, Connexin45, OCT4, SOX2, Nanog, hTERT, UTF-1, ZFP42, SSEA-3, SSEA-4, Tral-60, or Tral-81.
  • the differentiated cells may be purified by exposing a treated cell population to an agent (such as an antibody) that specifically recognizes a protein marker, such as CXCR4, expressed by cells expressing markers characteristic of the definitive endoderm lineage.
  • an agent such as an antibody
  • a protein marker such as CXCR4
  • Cells expressing markers characteristic of the definitive endoderm lineage may be differentiated into cells expressing markers characteristic of the pancreatic endoderm lineage by any method in the art or by any method proposed in this invention.
  • cells expressing markers characteristic of the definitive endoderm lineage 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 a fibroblast growth factor and the hedgehog signaling pathway inhibitor KAAD-cyclopamine, then removing the medium containing the fibroblast growth factor and KAAD-cyclopamine and subsequently culturing the cells in medium containing retinoic acid, a fibroblast growth factor and KAAD-cyclopamine.
  • a fibroblast growth factor and the hedgehog signaling pathway inhibitor KAAD-cyclopamine an example of this method is disclosed in Nature Biotechnology 24, 1392-1401 (2006).
  • cells expressing markers characteristic of the definitive endoderm lineage 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 and at least one fibroblast growth factor for a period of time, according to the methods disclosed in U.S. patent application Ser. No. 11/736,908, assigned to LifeScan, Inc.
  • cells expressing markers characteristic of the definitive endoderm lineage 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 and at least one fibroblast growth factor for a period of time, according to the methods disclosed in U.S. patent application Ser. No. 11/779,311, assigned to LifeScan, Inc.
  • cells expressing markers characteristic of the definitive endoderm lineage 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 according to the methods disclosed in U.S. patent application Ser. No. 60/990,529.
  • Cells expressing markers characteristic of the definitive endoderm lineage may be treated with at least one other additional factor that may enhance the formation of cells expressing markers characteristic of the pancreatic endoderm lineage.
  • the at least one other additional factor may enhance the proliferation of the cells expressing markers characteristic of the pancreatic endoderm lineage formed by the methods of the present invention.
  • the at least one other additional factor may enhance the ability of the cells expressing markers characteristic of the pancreatic endoderm lineage formed by the methods of the present invention to form other cell types, or improve the efficiency of any other additional differentiation steps.
  • the at least one additional factor may be, for example, nicotinamide, members of TGF- ⁇ family, including TGF- ⁇ 1, 2, and 3, serum albumin, members of the fibroblast growth factor family, platelet-derived growth factor-AA, and -BB, platelet rich plasma, insulin growth factor (IGF-I, II), growth differentiation factor (such as, for example, GDF-5, -6, -8, -10, -11), glucagon like peptide-I and II (GLP-I and II), GLP-1 and GLP-2 mimetobody, Exendin-4, retinoic acid, parathyroid hormone, insulin, progesterone, aprotinin, hydrocortisone, ethanolamine, beta mercaptoethanol, epidermal growth factor (EGF), gastrin I and II, copper chelators such as, for example, triethylene pentamine, forskolin, Na-Butyrate, activin, betacellulin, ITS, noggin, neurite growth factor, nodal, val
  • the at least one other additional factor may be supplied by conditioned media obtained from pancreatic cells lines such as, for example, PANC-1 (ATCC No: CRL-1469), CAPAN-1 (ATCC No: HTB-79), BxPC-3 (ATCC No: CRL-1687), HPAF-II (ATCC No: CRL-1997), hepatic cell lines such as, for example, HepG2 (ATCC No: HTB-8065), and intestinal cell lines such as, for example, FHs 74 (ATCC No: CCL-241).
  • pancreatic cells lines such as, for example, PANC-1 (ATCC No: CRL-1469), CAPAN-1 (ATCC No: HTB-79), BxPC-3 (ATCC No: CRL-1687), HPAF-II (ATCC No: CRL-1997), hepatic cell lines such as, for example, HepG2 (ATCC No: HTB-8065), and intestinal cell lines such as, for example, FHs 74 (ATCC No: CCL-241).
  • pancreatic endoderm lineage specific markers include the expression of one or more transcription factors such as, for example, Hlxb9, PTF-1a, PDX-1, HNF-6, HNF-1beta.
  • the efficiency of differentiation may be determined by exposing a treated cell population to an agent (such as an antibody) that specifically recognizes a protein marker expressed by cells expressing markers characteristic of the pancreatic endoderm lineage.
  • an agent such as an antibody
  • RT-PCR quantitative reverse transcriptase polymerase chain reaction
  • Northern blots in situ hybridization
  • immunoassays such as immunohistochemical analysis of sectioned material, Western blotting, and for markers that are accessible in intact cells, flow cytometry analysis (FACS) (see, e.g., Harlow and Lane, Using Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Laboratory Press (1998)).
  • Cells expressing markers characteristic of the pancreatic endoderm lineage may be differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage by any method in the art or by any method disclosed in this invention.
  • cells expressing markers characteristic of the pancreatic endoderm lineage may be differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage according to the methods disclosed in D'Amour et al, Nature Biotechnology 24, 1392-1401 (2006).
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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 and exendin 4, then removing the medium containing DAPT and exendin 4 and subsequently culturing the cells in medium containing exendin 1, IGF-1 and HGF.
  • An example of this method is disclosed in Nature Biotechnology 24, 1392-1401 (2006).
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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, then removing the medium containing exendin 4 and subsequently culturing the cells in medium containing exendin 1, IGF-1 and HGF.
  • An example of this method is disclosed in D'Amour et al, Nature Biotechnology, 2006.
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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 and exendin 4.
  • An example of this method is disclosed in D'Amour et al, Nature Biotechnology, 2006.
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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.
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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 U.S. patent application Ser. No. 11/736,908, assigned to LifeScan, Inc.
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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 U.S. patent application Ser. No. 11/779,311, assigned to LifeScan, Inc.
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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 U.S. patent application Ser. No. 60/953,178, assigned to LifeScan, Inc.
  • cells expressing markers characteristic of the pancreatic endoderm lineage 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 according to the methods disclosed in U.S. patent application Ser. No. 60/990,529.
  • Cells expressing markers characteristic of the pancreatic endoderm lineage may be treated with at least one other additional factor that may enhance the formation of cells expressing markers characteristic of the pancreatic endocrine lineage.
  • the at least one other additional factor may enhance the proliferation of the cells expressing markers characteristic of the pancreatic endocrine lineage formed by the methods of the present invention. Further, the at least one other additional factor may enhance the ability of the cells expressing markers characteristic of the pancreatic endocrine lineage formed by the methods of the present invention to form other cell types or improve the efficiency of any other additional differentiation steps.
  • the at least one additional factor may be, for example, nicotinamide, members of TGF- ⁇ family, including TGF- ⁇ 1, 2, and 3, serum albumin, members of the fibroblast growth factor family, platelet-derived growth factor-AA, and -BB, platelet rich plasma, insulin growth factor (IGF-I, II), growth differentiation factor (such as, for example, GDF-5, -6, -8, -10, -11), glucagon like peptide-I and II (GLP-I and II), GLP-1 and GLP-2 mimetobody, Exendin-4, retinoic acid, parathyroid hormone, insulin, progesterone, aprotinin, hydrocortisone, ethanolamine, beta mercaptoethanol, epidermal growth factor (EGF), gastrin I and II, copper chelators such as, for example, triethylene pentamine, forskolin, Na-Butyrate, activin, betacellulin, ITS, noggin, neurite growth factor, nodal, val
  • the at least one other additional factor may be supplied by conditioned media obtained from pancreatic cells lines such as, for example, PANC-1 (ATCC No: CRL-1469), CAPAN-1 (ATCC No: HTB-79), BxPC-3 (ATCC No: CRL-1687), HPAF-II (ATCC No: CRL-1997), hepatic cell lines such as, for example, HepG2 (ATCC No: HTB-8065), and intestinal cell lines such as, for example, FHs 74 (ATCC No: CCL-241).
  • pancreatic cells lines such as, for example, PANC-1 (ATCC No: CRL-1469), CAPAN-1 (ATCC No: HTB-79), BxPC-3 (ATCC No: CRL-1687), HPAF-II (ATCC No: CRL-1997), hepatic cell lines such as, for example, HepG2 (ATCC No: HTB-8065), and intestinal cell lines such as, for example, FHs 74 (ATCC No: CCL-241).
  • Markers characteristic of cells of the pancreatic endocrine lineage are well known to those skilled in the art, and additional markers characteristic of the pancreatic endocrine lineage continue to be identified. These markers can be used to confirm that the cells treated in accordance with the present invention have differentiated to acquire the properties characteristic of the pancreatic endocrine lineage.
  • Pancreatic endocrine lineage specific markers include the expression of one or more transcription factors such as, for example, NGN3, NEURO, or ISL1.
  • ⁇ cell lineage specific characteristics include the expression of one or more transcription factors such as, for example, PDX1, NKX2.2, NKX6.1, ISL1, PAX6, PAX4, NEUROD, HNF1 beta, HNF6, HNF3 beta, or MAFA, among others. These transcription factors are well established in the art for identification of endocrine cells. See, e.g., Edlund (Nature Reviews Genetics 3: 524-632 (2002)).
  • the efficiency of differentiation may be determined by exposing a treated cell population to an agent (such as an antibody) that specifically recognizes a protein marker expressed by cells expressing markers characteristic of the pancreatic endocrine lineage.
  • an agent such as an antibody
  • the efficiency of differentiation may be determined by exposing a treated cell population to an agent (such as an antibody) that specifically recognizes a protein marker expressed by cells expressing markers characteristic of the ⁇ cell lineage.
  • RT-PCR quantitative reverse transcriptase polymerase chain reaction
  • Northern blots in situ hybridization
  • immunoassays such as immunohistochemical analysis of sectioned material, Western blotting, and for markers that are accessible in intact cells, flow cytometry analysis (FACS) (see, e.g., Harlow and Lane, Using Antibodies: A Laboratory Manual, New York: Cold Spring Harbor Laboratory Press (1998)).
  • the efficiency of differentiation is determined by measuring the percentage of insulin positive cells in a given cell culture following treatment.
  • the methods of the present invention produce about 100% insulin positive cells in a given culture.
  • the methods of the present invention produce about 90% insulin positive cells in a given culture.
  • the methods of the present invention produce about 80% insulin positive cells in a given culture.
  • the methods of the present invention produce about 70% insulin positive cells in a given culture.
  • the methods of the present invention produce about 60% insulin positive cells in a given culture.
  • the methods of the present invention produce about 50% insulin positive cells in a given culture.
  • the methods of the present invention produce about 40% insulin positive cells in a given culture.
  • the methods of the present invention produce about 30% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 20% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 10% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 5% insulin positive cells in a given culture.
  • the efficiency of differentiation is determined by measuring glucose-stimulated insulin secretion, as detected by measuring the amount of C-peptide released by the cells.
  • cells produced by the methods of the present invention produce about 1000 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 900 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 800 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 700 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 600 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 500 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 400 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 500 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 400 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 300 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 200 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 100 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 90 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 80 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 70 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 60 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 50 ng C-peptide/pg DNA.
  • cells produced by the methods of the present invention produce about 40 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 30 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 20 ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 10 ng C-peptide/pg DNA.
  • the present invention provides a method for treating a patient suffering from, or at risk of developing, Type1 diabetes. This method involves culturing pluripotent stem cells, differentiating the pluripotent stem cells in vitro into a ⁇ -cell lineage, and implanting the cells of a ⁇ -cell lineage into a patient.
  • this invention provides a method for treating a patient suffering from, or at risk of developing, Type 2 diabetes. This method involves culturing pluripotent stem cells, differentiating the cultured cells in vitro into a ⁇ -cell lineage, and implanting the cells of a ⁇ -cell lineage into the patient.
  • the patient can be further treated with pharmaceutical agents or bioactives that facilitate the survival and function of the transplanted cells.
  • agents may include, for example, insulin, members of the TGF- ⁇ family, including TGF- ⁇ 1, 2, and 3, bone morphogenic proteins (BMP-2, -3, -4, -5, -6, -7, -11, -12, and -13), fibroblast growth factors-1 and -2, platelet-derived growth factor-AA, and -BB, platelet rich plasma, insulin growth factor (IGF-I, II) growth differentiation factor (such as, for example, GDF-5, -6, -7, -8, -10, -15), vascular endothelial cell-derived growth factor (VEGF), pleiotrophin, endothelin, among others.
  • IGF-I, II insulin growth factor
  • VEGF vascular endothelial cell-derived growth factor
  • Other pharmaceutical compounds can include, for example, nicotinamide, glucagon like peptide-I (GLP-1) and II, GLP-1 and -2 mimetibody, Exendin-4, retinoic acid, parathyroid hormone, MAPK inhibitors, such as, for example, compounds disclosed in U.S. Published Application 2004/0209901 and U.S. Published Application 2004/0132729.
  • GLP-1 and II glucagon like peptide-I
  • GLP-1 and -2 mimetibody GLP-1 and -2 mimetibody
  • Exendin-4 retinoic acid
  • parathyroid hormone retinoic acid
  • MAPK inhibitors such as, for example, compounds disclosed in U.S. Published Application 2004/0209901 and U.S. Published Application 2004/0132729.
  • the pluripotent stem cells may be differentiated into an insulin-producing cell prior to transplantation into a recipient.
  • the pluripotent stem cells are fully differentiated into ⁇ -cells prior to transplantation into a recipient.
  • the pluripotent stem cells may be transplanted into a recipient in an undifferentiated or partially differentiated state. Further differentiation may take place in the recipient.
  • Definitive endoderm cells or, alternatively, pancreatic endoderm cells, or, alternatively, ⁇ cells may be implanted as dispersed cells or formed into clusters that may be infused into the hepatic portal vein.
  • cells may be provided in biocompatible degradable polymeric supports, porous non-degradable devices or encapsulated to protect from host immune response.
  • Cells may be implanted into an appropriate site in a recipient. The implantation sites include, for example, the liver, natural pancreas, renal subcapsular space, omentum, peritoneum, subserosal space, intestine, stomach, or a subcutaneous pocket.
  • additional factors such as growth factors, antioxidants or anti-inflammatory agents, can be administered before, simultaneously with, or after the administration of the cells.
  • growth factors are utilized to differentiate the administered cells in vivo. These factors can be secreted by endogenous cells and exposed to the administered cells in situ. Implanted cells can be induced to differentiate by any combination of endogenous and exogenously administered growth factors known in the art.
  • the amount of cells used in implantation depends on a number of various factors including the patient's condition and response to the therapy, and can be determined by one skilled in the art.
  • this invention provides a method for treating a patient suffering from, or at risk of developing diabetes.
  • This method involves culturing pluripotent stem cells, differentiating the cultured cells in vitro into a ⁇ -cell lineage, and incorporating the cells into a three-dimensional support.
  • the cells can be maintained in vitro on this support prior to implantation into the patient.
  • the support containing the cells can be directly implanted in the patient without additional in vitro culturing.
  • the support can optionally be incorporated with at least one pharmaceutical agent that facilitates the survival and function of the transplanted cells.
  • Support materials suitable for use for purposes of the present invention include tissue templates, conduits, barriers, and reservoirs useful for tissue repair.
  • synthetic and natural materials in the form of foams, sponges, gels, hydrogels, textiles, and nonwoven structures which have been used in vitro and in vivo to reconstruct or regenerate biological tissue, as well as to deliver chemotactic agents for inducing tissue growth, are suitable for use in practicing the methods of the present invention. See, for example, the materials disclosed in U.S. Pat. No. 5,770,417, U.S. Pat. No. 6,022,743, U.S. Pat. No. 5,567,612, U.S. Pat. No. 5,759,830, U.S. Pat. No.
  • the pharmaceutical agent can be mixed with the polymer solution prior to forming the support.
  • a pharmaceutical agent could be coated onto a fabricated support, preferably in the presence of a pharmaceutical carrier.
  • the pharmaceutical agent may be present as a liquid, a finely divided solid, or any other appropriate physical form.
  • excipients may be added to the support to alter the release rate of the pharmaceutical agent.
  • the support is incorporated with at least one pharmaceutical compound that is an anti-inflammatory compound, such as, for example, compounds disclosed in U.S. Pat. No. 6,509,369.
  • the support may be incorporated with at least one pharmaceutical compound that is an anti-apoptotic compound, such as, for example, compounds disclosed in U.S. Pat. No. 6,793,945.
  • the support may also be incorporated with at least one pharmaceutical compound that is an inhibitor of fibrosis, such as, for example, compounds disclosed in U.S. Pat. No. 6,331,298.
  • the support may also be incorporated with at least one pharmaceutical compound that is capable of enhancing angiogenesis, such as, for example, compounds disclosed in U.S. Published Application 2004/0220393 and U.S. Published Application 2004/0209901.
  • the support may also be incorporated with at least one pharmaceutical compound that is an immunosuppressive compound, such as, for example, compounds disclosed in U.S. Published Application 2004/0171623.
  • an immunosuppressive compound such as, for example, compounds disclosed in U.S. Published Application 2004/0171623.
  • the support may also be incorporated with at least one pharmaceutical compound that is a growth factor, such as, for example, members of the TGF- ⁇ family, including TGF- ⁇ 1, 2, and 3, bone morphogenic proteins (BMP-2, -3, -4, -5, -6, -7, -11, -12, and -13), fibroblast growth factors-1 and -2, platelet-derived growth factor-AA, and -BB, platelet rich plasma, insulin growth factor (IGF-I, II) growth differentiation factor (such as, for example, GDF-5, -6, -8, -10, -15), vascular endothelial cell-derived growth factor (VEGF), pleiotrophin, endothelin, among others.
  • a growth factor such as, for example, members of the TGF- ⁇ family, including TGF- ⁇ 1, 2, and 3, bone morphogenic proteins (BMP-2, -3, -4, -5, -6, -7, -11, -12, and -13), fibroblast growth factors-1 and -2, platelet
  • Other pharmaceutical compounds can include, for example, nicotinamide, hypoxia inducible factor 1-alpha, glucagon like peptide-I (GLP-1), GLP-1 and GLP-2 mimetibody, and II, Exendin-4, nodal, noggin, NGF, retinoic acid, parathyroid hormone, tenascin-C, tropoelastin, thrombin-derived peptides, cathelicidins, defensins, laminin, biological peptides containing cell- and heparin-binding domains of adhesive extracellular matrix proteins such as fibronectin and vitronectin, MAPK inhibitors, such as, for example, compounds disclosed in U.S. Published Application 2004/0209901 and U.S. Published Application 2004/0132729.
  • MAPK inhibitors such as, for example, compounds disclosed in U.S. Published Application 2004/0209901 and U.S. Published Application 2004/0132729.
  • the incorporation of the cells of the present invention into a scaffold can be achieved by the simple depositing of cells onto the scaffold.
  • Cells can enter into the scaffold by simple diffusion (J. Pediatr. Surg. 23 (1 Pt 2): 3-9 (1988)).
  • Several other approaches have been developed to enhance the efficiency of cell seeding.
  • spinner flasks have been used in seeding of chondrocytes onto polyglycolic acid scaffolds (Biotechnol. Prog. 14(2): 193-202 (1998)).
  • Another approach for seeding cells is the use of centrifugation, which yields minimum stress to the seeded cells and enhances seeding efficiency.
  • Yang et al. developed a cell seeding method (J. Biomed. Mater. Res. 55(3): 379-86 (2001)), referred to as Centrifugational Cell Immobilization (CCI).
  • CCI Centrifugational Cell Immobilization
  • the human embryonic stem cell lines H1, H7, and H9 were obtained from WiCell Research Institute, Inc., (Madison, Wis.) and cultured according to instructions provided by the source institute.
  • the human embryonic stem cells were also seeded on plates coated with a 1:30 dilution of reduced growth factor MATRIGELTM (BD Biosciences; Cat #356231) and cultured in MEF-conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB).
  • the cells cultured on MATRIGELTM were routinely passaged as clusters using collagenase IV (Invitrogen/GIBCO; Cat #17104-019), Dispase (Invitrogen; Cat #17105-041), or Liberase CI enzyme (Roche; Cat #11814435001). In some instances, the cells were passaged as single cells using ACCUTASE (Sigma; Cat #A6964).
  • Human embryonic stem cells used in these examples were maintained in an undifferentiated, pluripotent state with passage on average every four-days. Passage was performed by exposing cell cultures to a solution of collagenase (1 or 10 mg/ml; Sigma-Aldrich) for 10 to 30 minutes at 37° C. followed by gentle scraping with a pipette tip to recover cell clusters. Clusters were allowed to sediment by gravity, followed by washing to remove residual collagenase. Cell clusters were split at a 1:3 ratio for routine maintenance culture or a 1:1 ratio for later assay. All human ES cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotypic phenotype and absence of mycoplasma contamination.
  • Activin A is an important mediator of differentiation in a broad range of cell types, including differentiation of embryonic stem cells to definitive endoderm.
  • human embryonic stem cells are treated with a combination of activin A and Wnt3a, various genes representative of definitive endoderm are up-regulated.
  • a bioassay that measures this differentiation in human embryonic stem cells was adapted in miniaturized format to 96-well plates for screening purposes. Validation was completed using treatment with commercial sources of activin A and Wnt3a recombinant proteins and measuring protein expression of the transcription factor SOX17, considered to be a representative marker of definitive endoderm.
  • Live Cell Assay Briefly, clusters of H1 human embryonic stem cells were grown on reduced growth factor MATRIGELTM (Invitrogen; Cat #356231)-coated tissue culture plastic. Cells were passaged using collagenase (Invitrogen; Cat #17104-019) treatment and gentle scraping, washed to remove residual enzyme, and plated in a ratio of 1:1 (surface area) on reduced growth factor MATRIGELTM-coated 96-well black plates (Packard ViewPlates; Perkin Elmer; Cat #6005182).
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with 100 ⁇ l per well mouse embryonic fibroblast (MEF) conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB).
  • MEF mouse embryonic fibroblast
  • test samples added to the assay wells were diluted in DMEM:F12 with 0.5% FCS (HyClone; Cat #SH30070.03) and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN).
  • test samples added to the assay wells were diluted in DMEM:F12 with 2% FCS, without any Wnt3a.
  • Negative control samples omitted treatment with both activin A and Wnt3a.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • FIG. 1 shows validation of the screening assay, testing a two-fold dilution curve of a commercial source of activin A (PeproTech) and measuring both cell number ( FIG. 1A ) and SOX17 intensity ( FIG. 1B ).
  • Optimal activin A effects for induction of SOX17 expression were generally observed in the 100-200 ng/ml range with an EC 50 of 30-50 ng/ml.
  • Omitting Wnt3a from treatment on days 1 and 2 of assay failed to produce measurable SOX17 expression ( FIG. 1B , white bars). Absence of activin A also failed to yield SOX17 expression ( FIG. 1B ).
  • Differentiation of pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage is mediated through a series of receptor-ligand interactions that in turn activate receptor kinases leading to phosphorylation and nuclear translocation of downstream substrates, eventually regulating expression of specific target genes.
  • Optimal activation of these signaling cascades in some cell types may require inhibition of opposing default pathways.
  • redundant pathways involving alternative members of a larger kinase family may substitute in part for one or more signaling molecules.
  • canonical and non-canonical pathways may diverge with different initiating stimuli but may lead to a similar functional outcome.
  • Cell-based functional screens are one approach to identify novel targets and methods that can impact specific cellular responses.
  • One very powerful approach involves a series of iterative screens whereby leads or hits from one screen are integrated into a subsequent screen.
  • a series of different variables are integrated in a combinatorial fashion (for example, growth factors with kinase inhibitors) to identify novel effects on cellular differentiation.
  • a library of small molecules comprising aniline-pyridinotriazines, cyclic aniline-pyridinotriazines and intermediate structures in their synthesis was tested for properties important during definitive endoderm differentiation of human embryonic stem cells, specifically for effects to retain or enhance cell number at the conclusion of a ‘first’ differentiation step in low serum and in the absence of the growth factor activin A.
  • Cell assay seeding Briefly, clusters of H1 human embryonic stem cells were grown on reduced growth factor MATRIGELTM (Invitrogen; Cat #356231)-coated tissue culture plastic. Cells were passaged using collagenase (Invitrogen; Cat #17104-019) treatment and gentle scraping, washed to remove residual enzyme, and plated with even dispersal at a ratio of 1:1 (surface area) on reduced growth factor MATRIGELTM-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of 100 ⁇ l/well.
  • MATRIGELTM Invitrogen; Cat #3562311
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • the compounds tested were made available as 5 mM stocks in 96-well plate format, solubilized in 100% DMSO (Sigma; Cat #D2650) and stored at ⁇ 80° C.
  • the library compounds were further diluted to an intermediate concentration of 0.2 mM in 50 mM HEPES (Invitrogen; Cat #15630-080), 20% DMSO and stored at 4° C. Test conditions were performed in triplicate, feeding on alternate days over a four-day assay period.
  • Primary screening assays were initiated by aspirating culture medium from each well followed by three washes in PBS (Invitrogen; Cat #14190) to remove residual growth factors and serum.
  • test volumes of 200 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.5% FCS (HyClone; Cat #SH30070.03) and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN) plus 2.5 ⁇ M test compound.
  • test volumes of 200 ⁇ l per well were added back containing DMEM:F12 base medium supplemented with 2% FCS plus 2.5 ⁇ M test compound, without Wnt3a.
  • Positive control samples contained the same base medium supplemented with FCS, substituting 100 ng/ml recombinant human activin A (PeproTech; Cat #120-14) for the test compound throughout the four-day assay along with Wnt3a (20 ng/ml) added only on days 1 and 2.
  • Negative control samples contained DMEM:F12 base medium supplemented with FCS, adding Wnt3a on days 1 and 2 but omitting activin A.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 1 shows results of primary screening for the compounds tested, showing their effects on the differentiation of human embryonic stem cells to cells expressing markers characteristic of the definitive endoderm lineage in the absence of activin A.
  • the results include quantitative measures of both cell number and SOX17 intensity, where respective data points were averaged from triplicate wells and analyzed for each parameter using identical fields in each well. Expression of the transcription factor SOX17 is considered indicative of definitive endoderm differentiation.
  • Primary screening results were captured from eight 96-well screening plates. Plate to plate variability was reduced with inclusion of individual positive and negative controls on each plate. Results are normalized and expressed as a percentage of the positive control. Emphasis was placed on retention or amplification of cell number at the conclusion of assay.
  • Table 2 lists a subset of 27 compounds and their analyzed results from the primary screening, where these hits appeared to retain cell number at a level equivalent to or better than the positive control despite the absence of activin A in the screening assay.
  • SOX17 expression was induced in the absence of activin A (for example, the cyclic aniline-pyridinotriazines Compound 35 and Compound 22.
  • the compounds shown in Table 2 were selected for further evaluation for effects on the differentiation of human embryonic stem cells to cells expressing markers characteristic of the definitive endoderm lineage in the absence of activin A.
  • a titration curve for activin A with a constant amount of Wnt3a showed at least two effects during DE differentiation: 1) maintaining cell numbers or preventing cell loss; and 2) inducing a marker of DE, for example, SOX17 expression (Example 2).
  • Primary screening from Example 3 identified compounds that could maintain similar or improved cell numbers in assay relative to addition of activin A/Wnt3a alone.
  • a secondary screening assay was conducted to evaluate the effect of combinations of the identified compounds with other growth factors, specifically EGF and FGF4, on the generation of definitive endoderm.
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • a secondary screening assay was conducted, testing in triplicate and feeding on alternate days over the four-day assay timeframe. Assays were initiated by aspirating culture medium from each well followed by three washes in PBS to remove residual growth factors and serum. Test volumes of 80 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.625% FCS (HyClone; Cat #SH30070.03), 25 ng/ml Wnt3a (R&D Systems), and 3.125 ⁇ M compound plus 20 ⁇ l 5 ⁇ stock of growth factors to yield a final concentration of 0.5% FCS, 20 ng/ml Wnt3a, and 2.5 ⁇ M compound plus 50 ng/ml EGF and 50 ng/ml FGF4 in the assay.
  • DMEM:F12 base medium Invitrogen; Cat #11330-032
  • FCS HyClone
  • Wnt3a R&D Systems
  • Positive control wells (100 ⁇ l/well) contained the same base medium supplemented with 0.5% FCS, 20 ng/ml Wnt3a and 100 ng/ml activin A.
  • Negative control wells (100 ⁇ l/well) contained the same base medium with 0.5% FCS and 20 ng/ml Wnt3a, omitting activin A.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat #AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS.
  • 4 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 3A shows the results for two growth factors, EGF and FGF 4 (50 ng/ml each) tested in combination with the aniline-pyridinotriazine compounds shown in Table 2 for their effects on the differentiation of human embryonic stem cells into cells expressing markers characteristic of the definitive endoderm lineage in the absence of activin A. Results are ranked in descending order for best effects on SOX17 expression. Although the effects of these compounds on SOX17 expression were considered weak relative to the activin A/Wnt3a positive control, the responses for some of these compounds were considered significant. For example a selection of the compounds appear to have unique properties with respect to retaining high cell numbers per well during assay, presumably either by preventing apoptosis or by modulating cell cycle.
  • a secondary assay was conducted to evaluate the effect of the compounds of the present invention with combinations of other individual growth factors or compounds known from the literature to regulate definitive endoderm differentiation.
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • EGF EGF
  • FGF4 Cat #235-F4
  • PDGF-A Cat #221-AA
  • PDGF-B Cat #220-BB
  • PDGF-C Cat #1687-CC
  • PDGF-D Cat #1159-SB
  • PDGF-A/B Cat #222-AB
  • VEGF VEGF
  • BMP-1 Cat #1927-ZN
  • BMP-2 Cat #355-BM
  • BMP-4 Cat #314-BP
  • BMP-6 Cat #507-BP
  • BMP-7 Cat #222-AB
  • BMP-2/7 Cat #3229-BM
  • BMP-7 Sigma; Cat #B1434
  • LY294002 (Cayman; Cat 70920), PD98059, U0126, U0124 (EMD Biosciences; Cat #453710), muscimol (Tocris; Cat #0289), biuculline (Tocris; Cat #0130), sodium butyrate (Sigma; Cat #B5887). All growth factors were solubilized in PBS with 0.1% BSA (Sigma; Cat #A7888) and stored frozen at ⁇ 80° C. Small molecules were solubilized in 100% DMSO (Sigma; Cat #D2650) and stored frozen at ⁇ 80° C.
  • the compounds were available as 5 mM stocks in 96-well plate format, solubilized in 100% DMSO and stored at ⁇ 80° C.
  • the compounds of the present invention were further diluted to an intermediate concentration of 0.2 mM in 50 mM HEPES (Invitrogen; Cat #15630-080), 20% DMSO and stored at 4° C. All growth factors and inhibitors were prepared in a deep well, 96-well polypropylene plate, diluted to 5 ⁇ intermediate stocks in DMEM:F12 base medium at the beginning of assay and stored at 4° C.
  • a secondary screening assay was conducted, testing in triplicate and feeding on alternate days over the four-day assay timeframe. Assays were initiated by aspirating culture medium from each well followed by three washes in PBS to remove residual growth factors and serum. Test volumes of 80 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.625% FCS (HyClone; Cat #SH30070.03), 25 ng/ml Wnt3a (R&D Systems), and 3.125 ⁇ M compound plus 20 ⁇ l 5 ⁇ stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20 ng/ml Wnt3a, and 2.5 ⁇ M compound.
  • All remaining growth factors were tested at a final assay concentration of 50 ng/ml (EGF, FGF4, PDGF-A, PDGF-B, PDGF-C, PDGF-D, PDGF-A/B, VEGF, BMP-1, BMP-2, BMP-4, BMP-6, BMP-7, BMP-2/7).
  • Final assay concentrations of small molecules tested were as follows: muscimol (20 ⁇ M), PD98059 (1 ⁇ M), LY294002 (2.5 ⁇ M), U0124 (1 ⁇ M), U0126 (1 ⁇ M), sodium butyrate (0.5 mM).
  • Positive control wells (100 ⁇ l/well) contained the same base medium supplemented with 0.5% FCS, 20 ng/ml Wnt3a and 100 ng/ml activin A.
  • Negative control wells (100 ⁇ l/well) contained the same base medium with 0.5% FCS and 20 ng/ml Wnt3a, omitting activin A.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat #AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS.
  • 4 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 4 shows the results for the differentiation of human embryonic stem cells into cells expressing markers characteristic of the definitive endoderm lineage following treatment with the compounds of the present invention in combination with individual growth factors or other small molecules.
  • members of the BMP family BMP-1, BMP-2, BMP-4, BMP-6, BMP-7, BMP-2/7) inhibited or had negligible effects on SOX17 expression.
  • PDGF-A, -AB, -C, and -D some members of the PDGF family (PDGF-A, -AB, -C, and -D) provided an increase in SOX17 expression (10-25% of the activin A/Wnt3a control).
  • Other growth factors showing similar increases in SOX17 expression included EGF (34%), VEGF (18%), and FGF4 (17%), although FGF4 was not able to support retention of cell numbers.
  • the small molecule muscimol (GABA A receptor agonist) tested in combination with Compound 35 also provided a modest increase in SOX17 expression; the GABA A antagonist bicuculline had no effect on SOX17 expression.
  • EGF, FGF4, PDGF-A, PDGF-B, PDGF-AB, PDGF-C, and PDGF-D and muscimol were selected for additional evaluation during definitive endoderm differentiation.
  • a secondary assay was conducted to evaluate the effects of combinations of different compounds with other individual agents on definitive endoderm differentiation.
  • the other agents selected for this screen had previously shown a modest increase in definitive endoderm formation, as tested with Compound 17 and as denoted in Table 5.
  • a broader panel of compounds was evaluated in with these agents, either in single pair-wise comparisons or pooled combinations.
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF-conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of the assay.
  • EGF EGF
  • FGF4 Cat #235-F4
  • PDGF-A Cat #221-AA
  • PDGF-D Cat #1159-SB
  • PDGF-A/B Cat #222-AB
  • VEGF VEGF
  • Muscimol was purchased from Tocris (Cat #0289). All growth factors were solubilized in PBS with 0.1% BSA (Sigma; Cat #A7888) and stored frozen at ⁇ 80° C. Muscimol was solubilized in 100% DMSO (Sigma; Cat #D2650) and stored frozen at ⁇ 80° C.
  • a secondary screening assay was conducted, testing in triplicate and feeding on alternate days over the four-day assay timeframe. Assays were initiated by aspirating culture medium from each well followed by three washes in PBS to remove residual growth factors and serum. Test volumes of 80 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.625% FCS (HyClone; Cat #SH30070.03), 25 ng/ml Wnt3a (R&D Systems), and 3.125 ⁇ M compound plus 20 ⁇ l 5 ⁇ stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20 ng/ml Wnt3a, and 2.5 ⁇ M.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat #AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS.
  • 4 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on grayscale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 5 shows compounds previously identified as hits (Table 2) tested in a definitive endoderm bioassay in various combinations with growth factors and muscimol, without activin A. Some compounds had minimal or weak effects on SOX17 expression with all growth factor combinations tested. However, some compounds were able to induce significant SOX17 expression with some but not all growth factor combinations.
  • GDF-8 also known as myostatin, is a member of the TGF- ⁇ family and has been shown to use the activin type II and TGF- ⁇ type I receptors (ALK4/5) to induce SMAD 2/3 phosphorylation.
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • EGF EGF
  • FGF4 Cat #235-F4
  • PDGF-A Cat #221-AA
  • PDGF-D Cat #1159-SB
  • PDGF-A/B Cat #222-AB
  • VEGF Cat #293-VE
  • GDF-8 GDF-8
  • Muscimol was purchased from Tocris (Cat #0289). All growth factors were solubilized in PBS with 0.1% BSA (Sigma; Cat #A7888) and stored frozen at ⁇ 80° C. Muscimol was solubilized in 100% DMSO (Sigma; Cat #D2650) and stored frozen at ⁇ 80° C.
  • Cyclic aniline-pyridinotriazine compounds were available as 5 mM stocks in 96-well plate format, solubilized in 100% DMSO and stored at ⁇ 80° C. Compound 34 was further diluted to an intermediate concentration of 0.2 mM in 50 mM HEPES (Invitrogen; Cat #15630-080), 20% DMSO and stored at 4° C. All growth factors and inhibitors were prepared in a deep well, 96-well polypropylene plate, diluted to 5 ⁇ intermediate stocks in DMEM:F12 base medium at the beginning of assay and stored at 4° C.
  • a secondary screening assay was conducted, testing in triplicate and feeding on alternate days over the four-day assay timeframe. Assays were initiated by aspirating culture medium from each well followed by three washes in PBS to remove residual growth factors and serum. Test volumes of 80 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.625% FCS (HyClone; Cat #SH30070.03), 25 ng/ml Wnt3a (R&D Systems), and 3.125 ⁇ M Compound 27 plus 20 ⁇ l 5 ⁇ stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20 ng/ml Wnt3a, and 2.5 ⁇ M Compound 34.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat #AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS.
  • 4 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 6 shows results of this assay. Where GDF-8 was present in any combination with the Compound 34, a substantial increase in SOX17 expression was observed. Furthermore, GDF-8 and Wnt3a with Compound 34 were sufficient to yield SOX17 expression (88% of control) in a range similar to that seen with 100 ng/ml activin A/Wnt3a treatment. It appears that the growth factor GDF-8 can serve as a replacement for activin A during definitive endoderm differentiation of human embryonic stem cells.
  • Cell assay seeding Briefly, clusters of H1 human embryonic stem cells were grown on reduced growth factor MATRIGELTM (Invitrogen; Cat #356231)-coated tissue culture plastic. Cells were passaged using collagenase (Invitrogen; Cat #17104-019) treatment and gentle scraping, washed to remove residual enzyme, and plated with even dispersal at a ratio of 1:1 (surface area) on reduced growth factor MATRIGELTM-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of 100 ⁇ l/well.
  • MATRIGELTM Invitrogen; Cat #3562311
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • EGF EGF
  • FGF4 Cat #235-F4
  • PDGF-A Cat #221-AA
  • PDGF-D Cat #1159-SB
  • PDGF-A/B Cat #222-AB
  • VEGF Cat #293-VE
  • GDF-8 GDF-8
  • the compounds were further diluted to an intermediate concentration of 0.2 mM in 50 mM HEPES (Invitrogen; Cat #15630-080), 20% DMSO and stored at 4° C. Test conditions were performed in single wells, feeding on alternate days over a four-day assay period. Primary screening assays were initiated by aspirating culture medium from each well followed by three washes in PBS (Invitrogen; Cat #14190) to remove residual growth factors and serum.
  • test volumes of 200 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.5% FCS (HyClone; Cat #SH30070.03) and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN) plus 2.5 ⁇ M compound. All remaining growth factors were tested at a final assay concentration of 50 ng/ml (EGF, FGF4, PDGF-A, PDGF-A/B, VEGF) with the exception of GDF-8 tested at 25 ng/ml. Final assay concentration of muscimol was 20 ⁇ M.
  • Positive control samples contained the same base medium supplemented with 0.5% FCS plus 20 ng/ml Wnt3a and 100 ng/ml recombinant human activin A (PeproTech; Cat #120-14).
  • Negative control samples contained DMEM:F12 base medium supplemented with 0.5% FCS and 20 ng/ml Wnt3a.
  • test volumes of 200 ⁇ l per well were added back containing DMEM:F12 base medium supplemented with 2% FCS plus 2.5 ⁇ M compound, without Wnt3a.
  • Positive control samples contained the same base medium supplemented with FCS, substituting 100 ng/ml recombinant human activin A (PeproTech; Cat #120-14) for the aniline-pyridinotriazine compound throughout the four-day assay along with Wnt3a (20 ng/ml) on days 1 and 2.
  • Negative control samples contained DMEM:F 12 base medium supplemented with FCS, adding Wnt3a on days 1 and 2 but omitting treatment with activin A.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell times area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • GDF-8 and a combination of growth factors/agonists were tested with a new set of aniline-pyridinotriazine compounds.
  • Results from two assay plates in this single experiment are ranked with respect to SOX17 responses (as a percentage of the positive control treatment with activin A and Wnt3a). Additional compounds were identified that show significant synergistic activity with the growth factor/agonist pool. These compounds were effective in both retaining assay cell number and yielding SOX17 expression during human embryonic stem cell differentiation in the absence of activin A. A list of these hits with greater than 25% activity of the positive control is shown in Table 8.
  • Cells were allowed to attach as clusters and then recover log phase growth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • EGF EGF
  • FGF4 Cat #235-F4
  • PDGF-A Cat #221-AA
  • PDGF-D Cat #1159-SB
  • PDGF-A/B Cat #222-AB
  • VEGF Cat #293-VE
  • GDF-8 GDF-8
  • Activin A was purchased from PeproTech (Cat #).
  • Muscimol was purchased from Tocris (Cat #0289). All growth factors were solubilized in PBS with 0.1% BSA (Sigma; Cat #A7888) and stored frozen at ⁇ 80° C.
  • Muscimol was solubilized in 100% DMSO (Sigma; Cat #D2650) and stored frozen at ⁇ 80° C.
  • the compounds were available as 5 mM stocks in 96-well plate format, solubilized in 100% DMSO and stored at ⁇ 80° C.
  • the compounds were further diluted to an intermediate concentration of 0.2 mM in 50 mM HEPES (Invitrogen; Cat #15630-080), 20% DMSO and stored at 4° C. All growth factors and inhibitors were prepared in a deep well, 96-well polypropylene plate, diluted to 5 ⁇ intermediate stocks in DMEM:F12 base medium at the beginning of assay and stored at 4° C.
  • a secondary screening assay was conducted, testing in triplicate and feeding on alternate days over the four-day assay timeframe. Assays were initiated by aspirating culture medium from each well followed by three washes in PBS to remove residual growth factors and serum. Test volumes of 80 ⁇ l per well were added back containing DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.625% FCS (HyClone; Cat #SH30070.03), 25 ng/ml Wnt3a (R&D Systems), 12.5 ng/ml activin A, and 3.125 ⁇ M compound plus 20 ⁇ l 5 ⁇ stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20 ng/ml Wnt3a, 10 ng/ml activin A, and 2.5 ⁇ M compound.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat #AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS.
  • 4 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 9 shows results from assay of various compounds and different combinations of growth factors with low doses of activin A. Some compounds showed robust synergistic responses with various growth factors. In other cases, the synergistic effects were more modest but significant relative to a low dose activin A control. Other compounds had no activity relative to the low dose activin A control.
  • Cyclic aniline-pyridinotriazine compounds were also tested in a screening format using cells dispersed through enzymatic treatment to single cells and plated in monolayer for assay.
  • the assay also made changes to eliminate serum that can provide growth factors even at low doses. To that end the basal medium was changed and serum was replaced with fatty acid free BSA.
  • the assay was shortened from four days to three days to provide a more narrow timeframe to measure results.
  • the assay included two growth factors, EGF and FGF4 that had previously shown significant but sub-optimal effects on definitive endoderm differentiation in the absence of activin A.
  • Cell assay seeding Briefly, clusters of H1 human embryonic stem cells were grown on reduced growth factor MATRIGELTM (Invitrogen; Cat #356231)-coated tissue culture plastic. Cultures were treated with Accutase (Sigma; Cat #A6964), using equivalent volumes of 10 ml per 10 cm 2 surface area for 5 minutes at 37° C., then gently resuspended, pelleted by centrifugation, and resuspended in MEF conditioned medium for counting. For assay seeding, cells were plated at 50,000 cells/cm 2 on TM reduced growth factor MATRIGELTM-coated 96-well black plates (Packard ViewPlates; Cat #6005182) using volumes of 100 ⁇ l/well.
  • MATRIGELTM Invitrogen; Cat #3562311
  • Cells were allowed to attach and recover log phase growth over a 3 to 5 day period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • Test volumes of 80 ⁇ l per well were added back containing RPMI 1640 base medium (Invitrogen; Cat #22400-089) supplemented with 2.5% fatty acid free BSA (MP Biomedicals LLC; Cat #152401), 10 ng/ml bFGF (PeproTech Inc; Cat #100-18B), 25 ng/ml Wnt3a (R&D Systems; Cat #1324-WN) and 3.125 ⁇ M Compound 22 plus 20 ⁇ l 5 ⁇ stock of growth factors to yield a final concentration of 2% fatty acid free BSA, 8 ng/ml bFGF (PeproTech Inc; Cat #100-18B), 20 ng/ml Wnt3a, and 2.5 ⁇ M Compound 22 in assay.
  • RPMI 1640 base medium Invitrogen; Cat #22400-089
  • Positive control wells contained the same base medium supplemented with 2% fatty acid free BSA, 8 ng/ml bFGF, 20 ng/ml Wnt3a, and 100 ng/ml recombinant human activin A (PeproTech; Cat #120-14).
  • Negative control wells contained the same base medium supplemented with 2% fatty acid free BSA, 8 ng/ml bFGF, 20 ng/ml Wnt3a but omitted treatment with activin A.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat #AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS.
  • 4 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • Table 10 shows results of this assay with Compound 34. Control samples with EGF and/or FGF4 alone without the Compound 34 had low SOX17 expression. Addition of Compound 34 added significant enhancement of SOX17 expression.
  • GDF-8 is able to replace activin A to differentiate human embryonic stem cells to cells expressing markers characteristic of the definitive endoderm lineage. It was important to know the relative potencies of GDF-8GDF-8 and activin A with respect their ability to differentiate human embryonic stem cells to cells expressing markers characteristic of the definitive endoderm lineage.
  • a dose response assay was conducted using equivalent concentrations of each growth factor to compare results during embryonic stem cell differentiation.
  • Preparation of cells for assay Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGEL-coated dishes in MEF conditioned medium with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen, Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the monolayer with MEF conditioned culture medium and gentle scraping to recover cell clusters. Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase. Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human embryonic stem cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotypic phenotype and for absence of mycoplasma contamination.
  • Cell clusters used in the assay were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and seeded onto reduced growth factor MATRIGELTM-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat #6005182) in volumes of 100 ⁇ l/well.
  • MEF conditioned medium supplemented with 8 ng/ml bFGF was used for initial plating and expansion.
  • Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back an aliquot (100 ⁇ l) of test medium. Test conditions were performed in quadruplicate over a total three-day assay period, feeding on day 1 and day 2 by aspirating and replacing the medium from each well with fresh test medium. Two 12-channel polypropylene basins (Argos technologies, Inc, Cat #B3135) were used to make the test media containing different concentrations of Activin A (PeproTech; Cat #120-14) or GDF-8 (R&D Systems, Cat #788-G8).
  • Channels numbered 2 through 12 of each basin contained 1 ml assay medium composed of RPMI-1640 medium (Invitrogen; Cat #22400) supplemented with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat #152401) and 8 ng/ml bFGF (PeproTech Inc.; Cat #100-18B), and with 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF) added on day 1, omitted on day 2 and 3.
  • Channel number 1 of each basin contained 1600 ng/ml Activin A or 1600 ng/ml GDF-8, diluted into the same assay medium.
  • One ml of medium was transferred from channel number 1 to channel number 2 and mixed well.
  • a fresh pipette tip was used to transfer one ml of medium from channel number 2 to channel number 3, followed by thorough mixing.
  • the same procedure was repeated in sequence through channel number 11 for each respective basin.
  • Channel number 12 of each basin contained medium without Activin A or GDF-8. By doing this, a series of two-fold test dilutions was created, containing Activin A or GDF-8 at concentrations ranging from 1.6 ng/ml to 1600 ng/ml, for addition to the respective assay wells.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total SOX17 intensity in each well were obtained using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each quadruplicate data set. Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 to 4500.
  • Total SOX17 intensity data were calculated using GraphPad Prism 4.02 (GraphPad Software, Inc., Lo Jolla, Calif.). Data were normalized to define the smallest and largest values in each data set as 0% and 100%, respectively. Table 11 shows the normalized values for each of the activin A and GDF-8 data sets. Two sigmoidal dose-response curves are shown in FIG. 2 as generated using the normalized values shown in Table 11. The R 2 values, indicating curve fit, were calculated using GraphPad Prism and determined to be 0.9944 for activin A and 0.9964 for GDF-8. Using GraphPad Prism, EC 50 values for each growth factor were calculated and determined to be 13.9 ng/ml for activin A and 184.8 ng/ml for GDF-8.
  • GDF-8 is less potent than activin A with respect to inducing human embryonic stem cells to differentiate to cells expressing markers characteristic of the definitive endoderm lineage. Nonetheless, GDF-8 can substitute for activin A and at specific concentrations, can induce an equivalent population of definitive endoderm cells, as denoted by SOX17 expression.
  • Parallel populations of human embryonic stem cells were differentiated to cells expressing markers characteristic of the definitive endoderm lineage using GDF-8 in combination with either Compound 34 or Compound 56. Thereafter, a step-wise differentiation protocol was applied to treated cells to promote differentiation toward pancreatic endoderm and endocrine lineages. A parallel control consisting of cells treated with Activin A and Wnt3a was maintained for comparison purposes throughout the step-wise differentiation process. Samples were taken at every stage of the differentiation to determine the appearance of proteins and mRNA biomarkers representative of the various stages of differentiation.
  • Preparation of cells for assay Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM-coated dishes in MEF conditioned medium with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen; Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the monolayer with MEF conditioned culture medium and gentle scraping to recover cell clusters. Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase. Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human ES cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma.
  • Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and seeded onto reduced growth factor MATRIGELTM-coated 24-well, black wall culture plates (Arctic White; Cat #AWLS-303012) in volumes of 0.5 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout the duration of assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back an aliquot (0.5 ml) of test medium. Test conditions for the first step of differentiation were conducted over a three-day period, feeding daily by aspirating and replacing the medium from each well with fresh test medium.
  • Step 2 of the differentiation protocol was carried out over two days.
  • Cells were fed daily by aspirating the medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM:F12 medium (Invitrogen; Cat #11330-032) containing 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat #152401), 50 ng/ml FGF7 (PeproTech; Cat #100-19), and 250 nM cyclopamine (Calbiochem; Cat #239804).
  • DMEM:F12 medium Invitrogen; Cat #11330-032
  • Albumin Bovine Fraction V Fatty Acid Free
  • FGF7 Fatty Acid Free
  • Step 3 of the differentiation protocol was carried out over four days.
  • Cells were fed daily by aspirating medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM-high glucose (Invitrogen; Cat #10569) supplemented with 1% B27 (Invitrogen; Cat #17504-044), 50 ng/ml FGF7, 100 ng/ml Noggin (R&D Systems; Cat #3344-NG), 250 nM KAAD-cyclopamine (Calbiochem; Cat #239804), and 2 ⁇ M all-trans retinoic acid (RA) (Sigma-Aldrich; Cat #R2625).
  • Step 4 of the differentiation protocol was carried out over three days.
  • Cells were fed daily by aspirating the medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin, 100 ng/ml Netrin-4, 1 ⁇ M DAPT (EMD Biosciences; Cat #565770), and 1 ⁇ M Alk 5 inhibitor (Axxora; Cat #ALX-270-445).
  • DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin, 100 ng/ml Netrin-4, 1 ⁇ M DAPT (EMD Biosciences; Cat #565770), and 1 ⁇ M Alk 5 inhibitor (Axxora; Cat #ALX-270-445).
  • Axxora Cat #ALX-270-445
  • Step 5 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 1% B27, and 1 ⁇ M Alk 5 inhibitor. Medium in each well was aspirated and replaced with a fresh aliquot (0.5 ml) on all days.
  • cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation.
  • Other culture wells were subjected to high content image analysis for protein expression levels of insulin and glucagon.
  • Step 6 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 1% B27. Medium in each well was aspirated and replaced with a fresh aliquot (0.5 ml) on alternating days. At the conclusion of the sixth step of differentiation, cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation.
  • FACS Analysis Cells for FACS analysis were blocked in a 1:5 solution of 0.5% human gamma-globulin (Sigma; Cat #G-4386) in PBS (Invitrogen; Cat #14040-133): BD FACS staining buffer—BSA (BD; Cat #554657) for 15 minutes at 4° C. Cells were then stained with antibodies for CD9 PE (BD; Cat #555372), CD99 PE (Caltag; Cat #MHCD9904) and CXCR4 APC (R&D Systems; Cat #FAB173A) for 30 minutes at 4° C.
  • BSA BD FACS staining buffer—BSA (BD; Cat #554657) for 15 minutes at 4° C. Cells were then stained with antibodies for CD9 PE (BD; Cat #555372), CD99 PE (Caltag; Cat #MHCD9904) and CXCR4 APC (R&D Systems; Cat #FAB173A) for 30 minutes at 4° C.
  • BD FACS staining buffer After a series of washes in BD FACS staining buffer, the cells were stained for viability with 7-AAD (BD; Cat #559925) and run on a BD FACSArray. A mouse IgG1K Isotype control antibody for both PE and APC was used to gate percent positive cells.
  • RNA samples were purified by binding to a silica-gel membrane (Rneasy Mini Kit, Qiagen, Calif.) in the presence of an ethanol-containing, high-salt buffer followed by washing to remove contaminants.
  • the RNA was further purified using a TURBO DNA-free kit (Ambion, INC), and high-quality RNA was then eluted in water. Yield and purity were assessed by A260 and A280 readings on a spectrophotometer.
  • CDNA copies were made from purified RNA using an ABI (ABI, CA) high capacity cDNA archive kit.
  • reagents were purchased from Applied Biosystems. Real-time PCR reactions were performed using the ABI PRISM® 7900 Sequence Detection System. TAQMAN® UNIVERSAL PCR MASTER MIX® (ABI, CA) was used with 20 ng of reverse transcribed RNA in a total reaction volume of 20 ⁇ l. Each cDNA sample was run in duplicate to correct for pipetting errors. Primers and FAM-labeled TAQMAN® probes were used at concentrations of 200 nM. The level of expression for each target gene was normalized using a human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) endogenous control previously developed by Applied Biosystems.
  • GPDH human glyceraldehyde-3-phosphate dehydrogenase
  • Primer and probe sets are listed in Table 12. After an initial incubation at 50° C. for 2 min followed by 95° C. for 10 min, samples were cycled 40 times in two stages—a denaturation step at 95° C. for 15 sec followed by an annealing/extension step at 60° C. for 1 min. Data analysis was carried out using GENEAMP®7000 Sequence Detection System software. For each primer/probe set, a Ct value was determined as the cycle number at which the fluorescence intensity reached a specific value in the middle of the exponential region of amplification. Relative gene expression levels were calculated using the comparative Ct method.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • 5 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Secondary antibodies used for analysis included 1:400 dilution Alexa Fluor 647 chicken anti-mouse IgG (Invitrogen; Cat #A-21463), 1:200 dilution Alexa Fluor 488 donkey anti-goat IgG (Invitrogen; Cat #A11055), 1:1000 dilution Alexa Fluor 647 chicken anti-rabbit IgG (Invitrogen; Cat #A21443), and 1:1000 dilution Alexa Fluor 488 chicken anti-mouse IgG (Invitrogen; Cat #A21200).
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 and 4500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control.
  • PCR results for representative differentiation markers are shown in Table 13 for cells harvested from each step of differentiation. Samples treated with GDF-8 and Wnt3a or with GDF-8 and either Compound 34 or Compound 56 showed similar, or in some instances, improved expression levels of expression markers associated with endodermal and endocrine differentiation.
  • FIG. 3 shows the results of the FACS analysis, showing the expression of the definitive endoderm marker, CXCR4, after the first step of differentiation.
  • Treatment of human embryonic stem cells with GDF-8 and Wnt3a yielded an equivalent percentage of CXCR4 positive cells compared to treatment with activin A and Wnt3a.
  • treatment of human embryonic stem cells with GDF-8 and a small molecule also yielded an equivalent or slightly higher percentage of CXC4 positive cells.
  • FIG. 4 shows high content image analysis for normalized SOX17 protein expression in human embryonic stem cells after three days differentiation to definitive endoderm. Levels of expression for treatment groups using GDF-8 with Wnt3a or GDF-8 with a small molecule are similar to treatment with Activin A and Wnt3a.
  • FIG. 5 shows high content image analysis for normalized Pdx1 and Cdx2 protein expression in human embryonic stem cells after the third step of differentiation to pancreatic endoderm.
  • Levels of expression for treatment groups using GDF-8 with Wnt3a or GDF-8 with Wnt3a or GDF-8 with a compound of the present invention show equivalent levels of PDX1 and CDX2.
  • the cell number retained after differentiation decreased thereby increasing the ratio of PDX1 expressing cells.
  • Similar results were obtained showing equivalent normalized PDX1 expression in all treatment groups after the fourth step of differentiation as shown in FIG. 6 .
  • FIG. 7 normalized protein levels of insulin and glucagon are shown, demonstrating equivalent expression between the Activin A and GDF-8 treatment groups.
  • GDF-8 in combination with Wnt3a or Compound 34 or Compound 56, can substitute for activin A during definitive endoderm differentiation and subsequent pancreatic endoderm and endocrine differentiation.
  • H1 human embryonic stem cell line Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM (BD Biosciences; Cat #356231)-coated dishes in MEF conditioned medium with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen; Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the monolayer with MEF conditioned culture medium and gentle scraping to recover cell clusters. Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase. Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human ES cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma.
  • Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and seeded onto reduced growth factor MATRIGELTM-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat #6005182) in volumes of 0.1 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout the duration of assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back aliquots (100 ⁇ l) of test medium. Test conditions were performed in triplicate over a total four-day assay period, feeding on day 1 and day 3 by aspirating and replacing the medium from each well with fresh test medium.
  • GDF-3 PeproTech; Cat #120-22
  • GDF-5 DePuy Orthopaedics, Inc., a Johnson & Johnson company
  • GDF-8 R&D Systems; Cat #788-G8
  • GDF-10 R&D Systems; Cat #1543-BP
  • GDF11 PeproTech; Cat #120-11
  • GDF-15 R&D Systems; Cat #957-GD
  • basal medium DMEM:F12 medium Invitrogen; Cat #11330-032
  • fetal bovine serum Hyclone; Cat #SH30070.03
  • test samples were created to evaluate activin A or various GDFs in combination with Wnt3a or Compound 34 or Compound 56. These test samples were added in 20 ⁇ l aliquots (5 ⁇ concentrated) to appropriately matched assay wells to yield a final assay volume of 100 ⁇ l in each well at the final assay conditions indicated. In the first set of control samples, the following conditions were tested: 1) no additive (i.e.
  • each of the six conditions was combined with 100 ng/ml GDF-5.
  • each of the six conditions was combined with 100 ng/ml GDF-8.
  • each of the six conditions was combined with 100 ng/ml GDF-10.
  • each of the six conditions was combined with 100 ng/ml GDF-11.
  • each of the six conditions was combined with 100 ng/ml GDF-15.
  • all wells for all test samples received 100 ng/ml Activin A or 100 ng/ml respective GDF growth factor, without Wnt3a or Compound 34 or Compound 56, diluted into DMEM:F12 medium supplemented with 2% FBS.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • 5 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 and 4500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control.
  • FIG. 8 shows high content image analysis for SOX17 protein expression in human embryonic stem cells after four days differentiation to definitive endoderm. In each case, results are normalized to the positive control treatment with activin A and Wnt3a. In FIG. 8A , only the positive control treatment yielded significant expression of SOX17; treatment with Wnt3a alone or either Compound 34 or Compound 56 alone failed to induce SOX17 expression. In FIG. 8 , panels B through G, normalized SOX17 expression levels are shown for each GDF growth factor substituting for activin A in the respective treatments. GDF-3 ( FIG. 8B ) and GDF-5 ( FIG. 8C ) induced weak expression of SOX17 and only in test samples where one of the compounds of the present invention was present. GDF10 ( FIG. 8A ).
  • FIG. 8D shows results for treatment groups using GDF-8 where GDF-8 in combination with either Compound 34 or Compound 56 caused a robust induction of SOX17, exceeding results seen with the activin A/Wnt3a positive control.
  • the presence of Compound 34 or Compound 56 combined with a GDF growth factor also caused an increase in cell number during differentiation.
  • Preparation of cells for assay Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM-(BD Biosciences; Cat #356231)-coated dishes in MEF conditioned medium with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen; Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the monolayer with MEF conditioned culture medium and gentle scraping to recover cell clusters. Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase.
  • H1 human embryonic stem cell line Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM-(BD Biosciences; Cat #356231)-coated dishes in MEF conditioned medium with passage on average every four days.
  • Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human embryonic stem cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma.
  • Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and seeded onto reduced growth factor MATRIGELTM-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat #6005182) in volumes of 0.1 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back aliquots (100 ⁇ l) of test medium. Test conditions were performed in triplicate over a total three day assay period, feeding on day 1 and day 2 by aspirating and replacing the medium from each well with fresh test medium.
  • BMP-2 R&D Systems; Cat #355-BM
  • BMP-3 R&D Systems; Cat #113-BP
  • BMP-4 R&D Systems; Cat #314-BP
  • TGF ⁇ -1 R&D Systems; Cat #240-B
  • GDF-8 PeproTech; Cat #120-00
  • GDF-8 GDF-8 (Shenandoah; Cat #100-22); and activin A (PeproTech; Cat #120-14).
  • each well was treated with 80 ⁇ l of growth medium [RPMI-1640 (Invitrogen; Cat #22400) containing 2.5% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat #152401), and 10 ng/ml bFGF].
  • growth medium RPMI-1640 (Invitrogen; Cat #22400) containing 2.5% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat #152401), and 10 ng/ml bFGF].
  • 25 ng/ml Wnt3a R&D Systems; Cat #1324-WN/CF
  • activin A was added to the growth medium to yield a final assay concentration of 100 ng/ml.
  • Comparative controls for this assay included: 1) no added growth factors; 2) Wnt3a alone; and 3) activin A with Wnt3a.
  • Each commercial source of GDF-8 was tested in combination with Wnt3a.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • 5 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 and 4500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control.
  • FIG. 9 shows high content image analysis for SOX17 protein expression in human embryonic stem cells after three days differentiation to definitive endoderm. In each case, results are normalized to the positive control treatment for activin A with Wnt3a.
  • the results in FIG. 9A show that treatment with growth medium alone, or Wnt3a alone failed to induce SOX17 expression; only the addition of activin A caused a robust expression of SOX17.
  • panels B and C results for each of the commercial sources of GDF-8 are depicted, showing differences in potency between the two vendors. Although less potent than activin A, there was significant induction of SOX17 expression in cells treated with GDF-8 in combination with Wnt3a.
  • FIG. 9 shows high content image analysis for SOX17 protein expression in human embryonic stem cells after three days differentiation to definitive endoderm. In each case, results are normalized to the positive control treatment for activin A with Wnt3a.
  • the results in FIG. 9A show that treatment with growth medium alone, or Wnt3a alone failed to induce SO
  • H1 human embryonic stem cell line Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM (BD Biosciences; Cat #356231)-coated dishes in MEF conditioned medium supplemented with 8 ng/ml bFGF (PeproTech Inc.; Cat #100-18B) with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen; Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the monolayer with MEF conditioned culture medium and gentle scraping to recover cell clusters.
  • MATRIGELTM BD Biosciences; Cat #3562311
  • MEF conditioned medium supplemented with 8 ng/ml bFGF (PeproTech Inc.; Cat #100-18B) with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg
  • Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase. Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human embryonic stem cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma.
  • Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and seeded onto reduced growth factor MATRIGELTM-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat #6005182) in volumes of 0.1 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout the duration of assay.
  • Assay was initiated by aspirating the culture medium from each well and adding back aliquots (100 ⁇ l) of test medium. Test conditions were performed in quadruplicate over a total four-day assay period, feeding daily by aspirating and replacing the medium from each well with fresh test medium.
  • Test compounds in this assay included six of the compounds of the present invention: Compound 181, Compound 180, Compound 19, Compound 202, Compound 40, and Compound 34, and a commercial GSK3i inhibitor BIO (EMD Chemicals, Inc.; Cat #361550).
  • Control conditions were as follows: 1) growth medium alone; 2) 20 ng/ml Wnt3a only R&D Systems; Cat #1324-WN/CF); 3) 100 ng/ml activin A (PeproTech; Cat #120-14); 4) 100 ng/ml activin A and 20 ng/ml Wnt3a; 5) 100 ng/ml GDF-8 (R&D Systems, Cat #788-G8); 6) 100 ng/ml GDF-8 and 20 ng/ml Wnt3a.
  • Test compounds were diluted two-fold in series to yield a concentration range from 78 nM to 10 ⁇ M in the final assay.
  • Experimental test samples combined each individual compound dilution series with 100 ng/ml activin A or 100 ng/ml GDF-8, both treatment sets in the absence of Wnt3a.
  • some wells continued to be treated with 20 ng/ml Wnt3a or diluted test compound in combination with either activin A or GDF-8.
  • activin A or GDF-8 treatment continued on the second and third day of assay, but Wnt3a or diluted test compound was removed.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • 5 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 and 4500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control.
  • results High content analysis results are shown for SOX17 expression in FIGS. 10-14 and resulting cell number at the conclusion of assay in FIGS. 15-19 .
  • results are shown for SOX17 expression resulting from control treatments using activin A or GDF-8, either alone or in combination with Wnt3a.
  • Activin A treatments resulted in significantly higher SOX17 expression than was observed with GDF-8 treatment.
  • FIG. 15 shows that activin A treatment resulted in a higher number of cells at the conclusion of assay than was seen with GDF-8 treatment, regardless of whether Wnt3a was present for one or three days during assay.
  • GDF-8 definitive endoderm differentiation
  • a commercial inhibitor of GSK3 included a commercial inhibitor of GSK3 as well as compounds of the present invention.
  • a step-wise differentiation protocol was applied to cells treated with GDF-8 in combination with various small molecules. The efficacy of differentiation was determined by gene expression for biomarkers representative the pancreatic endoderm, or pancreatic endocrine lineages.
  • a parallel control sample of cells treated with activin A and Wnt3a was maintained for comparison purposes throughout the step-wise differentiation process.
  • Preparation of cells for assay Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM (BD Biosciences; Cat #356231)-coated dishes in MEF conditioned medium with passage on average every four days. Passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen, Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the monolayer with MEF conditioned culture medium and gentle scraping to recover cell clusters. Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase.
  • H1 human embryonic stem cell line Stock cultures of human embryonic stem cells (H1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELTM (BD Biosciences; Cat #356231)-coated dishes in MEF conditioned medium with passage on average every four days. Passage
  • Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human embryonic stem cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma.
  • Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and plated onto reduced growth factor MATRIGEL-coated 24-well, black wall culture plates (Arctic White; Cat #AWLS-303012) in volumes of 0.5 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout the duration of assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back an aliquot (0.5 ml) of test medium. Test conditions for the first step of differentiation were conducted over a three-day period, feeding daily by aspirating and replacing the medium from each well with fresh test medium. On the first day of assay, 100 ng/ml activin A (PeproTech; Cat #120-14) or 100 ng/ml GDF-8 (R&D Systems, Cat #788-G8) was added to respective assay wells where each growth factor was diluted into RPMI-1640 medium (Invitrogen; Cat #22400) with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (Proliant Inc.
  • RPMI-1640 medium Invitrogen; Cat #22400
  • Albumin Bovine Fraction V Fatty Acid Free
  • Step 2 of the differentiation protocol was carried out over two days.
  • Cells were fed daily by aspirating the medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM:F12 medium (Invitrogen; Cat #11330-032) containing 2% FAF BSA, 50 ng/ml FGF7 (PeproTech; Cat #100-19), and 250 nM cyclopamine-KAAD (Calbiochem; Cat #239804).
  • Step 3 of the differentiation protocol was carried out over seven days.
  • Cells were fed daily by aspirating medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM-high glucose (Invitrogen; Cat #10569) supplemented with 0.1% Albumax (Invitrogen; Cat #11020-021), 0.5 ⁇ Insulin-Transferrin-Selenium (ITS-X; Invitrogen; Cat #51500056), 50 ng/ml FGF7, 100 ng/ml Noggin (R&D Systems; Cat #3344-NG), 250 nM KAAD-cyclopamine, and 2 ⁇ M all-trans retinoic acid (RA) (Sigma-Aldrich; Cat #R2625).
  • DMEM-high glucose Invitrogen; Cat #10569
  • Albumax Invitrogen; Cat #11020-021
  • ITS-X Insulin-Transferrin-Selenium
  • FGF7 100 ng/ml Noggin
  • Step 4 of the differentiation protocol was carried out over three days.
  • Cells were fed daily by aspirating the medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM-high glucose supplemented with 0.1% Albumax, 0.5 ⁇ Insulin-Transferrin-Selenium, 100 ng/ml Noggin, and 1 ⁇ M Alk 5 inhibitor (Axxora; Cat #ALX-270-445).
  • Axxora Cat #ALX-270-445.
  • cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation. Other culture wells were subjected to high content image analysis for protein expression levels of Pdx1.
  • Step 5 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 0.1% Albumax, 0.5 ⁇ Insulin-Transferrin-Selenium, and 1 ⁇ M Alk 5 inhibitor. Medium in each well was aspirated and replaced with a fresh aliquot (0.5 ml) on all days.
  • cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation.
  • Other culture wells were subjected to high content image analysis for protein expression levels of insulin and glucagon.
  • FACS Analysis Cells for FACS analysis were blocked in a 1:5 solution of 0.5% human gamma-globulin (Sigma; Cat #G-4386) in PBS (Invitrogen; Cat #14040-133): BD FACS staining buffer—BSA (BD; Cat #554657) for 15 minutes at 4° C. Cells were then stained with antibodies for CD9 PE (BD; Cat #555372), CD99 PE (Caltag; Cat #MHCD9904) and CXCR4 APC (R&D Systems; Cat #FAB173A) for 30 minutes at 4° C.
  • BSA BD FACS staining buffer—BSA (BD; Cat #554657) for 15 minutes at 4° C. Cells were then stained with antibodies for CD9 PE (BD; Cat #555372), CD99 PE (Caltag; Cat #MHCD9904) and CXCR4 APC (R&D Systems; Cat #FAB173A) for 30 minutes at 4° C.
  • BD FACS staining buffer After a series of washes in BD FACS staining buffer, the cells were stained for viability with 7-AAD (BD; Cat #559925) and run on a BD FACSArray. A mouse IgG1K Isotype control antibody for both PE and APC was used to gate percent positive cells.
  • RNA samples were purified by binding to a silica-gel membrane (Rneasy Mini Kit, Qiagen, Calif.) in the presence of an ethanol-containing, high-salt buffer followed by washing to remove contaminants.
  • the RNA was further purified using a TURBO DNA-free kit (Ambion, INC), and high-quality RNA was then eluted in water. Yield and purity were assessed by A260 and A280 readings on a spectrophotometer.
  • CDNA copies were made from purified RNA using an ABI (ABI, CA) high capacity cDNA archive kit.
  • reagents were purchased from Applied Biosystems. Real-time PCR reactions were performed using the ABI PRISM® 7900 Sequence Detection System. TAQMAN® UNIVERSAL PCR MASTER MIX® (ABI, CA) was used with 20 ng of reverse transcribed RNA in a total reaction volume of 20 ⁇ l. Each cDNA sample was run in duplicate to correct for pipetting errors. Primers and FAM-labeled TAQMAN® probes were used at concentrations of 200 nM. The level of expression for each target gene was normalized using a human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) endogenous control previously developed by Applied Biosystems.
  • GPDH human glyceraldehyde-3-phosphate dehydrogenase
  • Primer and probe sets are listed in Table 12. After an initial incubation at 50° C. for 2 min followed by 95° C. for 10 min, samples were cycled 40 times in two stages—a denaturation step at 95° C. for 15 sec followed by an annealing/extension step at 60° C. for 1 min. Data analysis was carried out using GENEAMP®7000 Sequence Detection System software. For each primer/probe set, a Ct value was determined as the cycle number at which the fluorescence intensity reached a specific value in the middle of the exponential region of amplification. Relative gene expression levels were calculated using the comparative Ct method.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • 5 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Other primary antibodies used for analysis included 1:200 dilution rabbit anti-human insulin (Cell Signaling; Cat #C27C9), and 1:1500 dilution mouse anti-human glucagon (Sigma-Aldrich; Cat #G2654).
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 and 4500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control.
  • PCR results for representative differentiation markers are shown in Table 14 for cells harvested from each step of differentiation. Samples treated with GDF-8 and Wnt3a or with GDF-8 and a small molecule showed similar expression levels of markers associated with endodermal and endocrine differentiation.
  • FIG. 20 panel A shows FACS analysis for the definitive endoderm marker, CXCR4, after the first step of differentiation.
  • Treatment of human embryonic stem cells with GDF-8 and Wnt3a yielded a similar percentage of CXCR4 positive cells compared to treatment with activin A and Wnt3a.
  • Treatment of human embryonic stem cells with GDF-8 and a compound of the present invention (Compound 19, Compound 202, Compound 40, or GSK3 inhibitor 1 ⁇ BIO) also yielded an equivalent or slightly higher percentage of CXC4 positive cells.
  • FIG. 20 , panel B shows high content image analysis for normalized SOX17 protein expression in human embryonic stem cells after three days differentiation to definitive endoderm.
  • treatment with GDF-8 resulted in a lower cell number at the conclusion of the first step of differentiation.
  • GDF-8 treatment in combination with Wnt3a or with the small molecule inhibitors clearly induced expression of SOX17, a marker of definitive endoderm.
  • treatment with GDF-8 and Compound 40 yielded cell numbers in culture and SOX17 expression equivalent to treatment with activin A and Wnt3a.
  • FIG. 20 panel C shows high content image analysis for relative cell numbers recovered from cultures treated through differentiation step 5. As observed earlier at the end of step 1, some treatments caused a drop in cell recovery relative to treatment with activin A and Wnt3a. This decrease in cell number was seen specifically with treatment groups using GDF-8 with GSK3 inhibitor BIO and also using GDF-8 with Compound 19. Additional GDF-8 treatment groups had cell recoveries similar to treatment with activin A and Wnt3a.
  • panels D-F normalized protein levels of insulin and glucagon are shown, along with their respective ratio for each treatment group.
  • GDF-8 and activin A were tested in combination with GDF-8 and activin A for definitive endoderm differentiation. These included a commercial inhibitor of GSK3 as well as the compounds of the present invention.
  • a step-wise differentiation protocol was applied to cells treated with GDF-8 in combination with various small molecules. The efficacy of differentiation was determined by gene expression for biomarkers representative of the pancreatic endoderm and pancreatic endocrine lineages. A parallel control sample of cells treated with activin A and Wnt3a was maintained for comparison purposes throughout the step-wise differentiation process.
  • Clusters were centrifuged at low speed to collect a cell pellet and remove residual dispase. Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human ES cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma.
  • Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and plated onto reduced growth factor MATRIGELTM coated 24-well, black wall culture plates (Arctic White; Cat #AWLS-303012) in volumes of 0.5 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back an aliquot (0.5 ml) of test medium. Test conditions for the first step of differentiation were conducted over a three-day period, feeding daily by aspirating and replacing the medium from each well with fresh test medium. On the first day of assay, 100 ng/ml activin A (PeproTech; Cat #120-14) or 100 ng/ml GDF-8 (R&D Systems, Cat #788-G8) was added to respective assay wells where each growth factor was diluted into RPMI-1640 medium (Invitrogen; Cat #: 22400) with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat #152401).
  • Wnt3a 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF) was also included.
  • 100 ng/ml activin A or 100 ng/ml GDF-8 was diluted into RPMI-1640 medium supplemented with 2% FAF BSA, omitting Wnt3a from all samples.
  • Wnt3a was replaced with a given concentration of small molecule compound, added only on the first day of definitive endoderm differentiation.
  • Step 2 of the differentiation protocol was carried out over two days.
  • Cells were fed daily by aspirating the medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM:F12 medium (Invitrogen; Cat #11330-032) containing 2% FAF BSA, 50 ng/ml FGF7 (PeproTech; Cat #100-19), and 250 nM cyclopamine-KAAD (Calbiochem; Cat #239804).
  • Step 3 of the differentiation protocol was carried out over four days.
  • Cells were fed daily by aspirating medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM-high glucose (Invitrogen; Cat #10569) supplemented with 0.1% Albumax (Invitrogen; Cat #: 11020-021), 0.5x Insulin-Transferrin-Selenium (ITS-X; Invitrogen; Cat #51500056), 50 ng/ml FGF7, 100 ng/ml Noggin (R&D Systems; Cat #3344-NG), 250 nM KAAD-cyclopamine, and 2 ⁇ M all-trans retinoic acid (RA) (Sigma-Aldrich; Cat #R2625).
  • DMEM-high glucose Invitrogen; Cat #10569
  • Albumax Invitrogen; Cat #: 11020-021
  • ITS-X Insulin-Transferrin-Selenium
  • FGF7 100 ng/ml
  • Step 4 of the differentiation protocol was carried out over three days.
  • Cells were fed daily by aspirating the medium from each well and replacing with a fresh aliquot (0.5 ml) of DMEM-high glucose supplemented with 0.1% Albumax, 0.5 ⁇ Insulin-Transferrin-Selenium, 100 ng/ml Noggin, and 1 ⁇ M Alk 5 inhibitor (Axxora; Cat #ALX-270-445).
  • Axxora Cat #ALX-270-445
  • Step 5 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 0.1% Albumax, 0.5 ⁇ Insulin-Transferrin-Selenium, and 1 ⁇ M Alk 5 inhibitor. Medium in each well was aspirated and replaced with a fresh aliquot (0.5 ml) on all days.
  • cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation.
  • Other culture wells were subjected to high content image analysis for protein expression levels of insulin and glucagon.
  • FACS Analysis Cells for FACS analysis were blocked in a 1:5 solution of 0.5% human gamma-globulin (Sigma; Cat #G-4386) in PBS (Invitrogen; Cat #14040-133): BD FACS staining buffer—BSA (BD; Cat #554657) for 15 minutes at 4° C. Cells were then stained with antibodies for CD9 PE (BD; Cat #555372), CD99 PE (Caltag; Cat #MHCD9904) and CXCR4 APC (R&D Systems; Cat #FAB173A) for 30 minutes at 4° C.
  • BSA BD FACS staining buffer—BSA (BD; Cat #554657) for 15 minutes at 4° C. Cells were then stained with antibodies for CD9 PE (BD; Cat #555372), CD99 PE (Caltag; Cat #MHCD9904) and CXCR4 APC (R&D Systems; Cat #FAB173A) for 30 minutes at 4° C.
  • BD FACS staining buffer After a series of washes in BD FACS staining buffer, the cells were stained for viability with 7-AAD (BD; Cat #559925) and run on a BD FACSArray. A mouse IgG1K Isotype control antibody for both PE and APC was used to gate percent positive cells.
  • RNA samples were purified by binding to a silica-gel membrane (Rneasy Mini Kit, Qiagen, Calif.) in the presence of an ethanol-containing, high-salt buffer followed by washing to remove contaminants.
  • the RNA was further purified using a TURBO DNA-free kit (Ambion, INC), and high-quality RNA was then eluted in water. Yield and purity were assessed by A260 and A280 readings on a spectrophotometer.
  • CDNA copies were made from purified RNA using an ABI (ABI, CA) high capacity cDNA archive kit.
  • reagents were purchased from Applied Biosystems. Real-time PCR reactions were performed using the ABI PRISM® 7900 Sequence Detection System. TAQMAN® UNIVERSAL PCR MASTER MIX® (ABI, CA) was used with 20 ng of reverse transcribed RNA in a total reaction volume of 20 ⁇ l. Each cDNA sample was run in duplicate to correct for pipetting errors. Primers and FAM-labeled TAQMAN® probes were used at concentrations of 200 nM. The level of expression for each target gene was normalized using a human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) endogenous control previously developed by Applied Biosystems.
  • GPDH human glyceraldehyde-3-phosphate dehydrogenase
  • Primer and probe sets are listed in Table 12. After an initial incubation at 50° C. for 2 min followed by 95° C. for 10 min, samples were cycled 40 times in two stages—a denaturation step at 95° C. for 15 sec followed by an annealing/extension step at 60° C. for 1 min. Data analysis was carried out using GENEAMP®7000 Sequence Detection System software. For each primer/probe set, a Ct value was determined as the cycle number at which the fluorescence intensity reached a specific value in the middle of the exponential region of amplification. Relative gene expression levels were calculated using the comparative Ct method.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well.
  • 5 ⁇ g/ml Hoechst 33342 (Invitrogen; Cat #H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in 100 ⁇ l/well PBS for imaging.
  • Secondary antibodies used for analysis included 1:400 dilution Alexa Fluor 647 chicken anti-mouse IgG (Invitrogen; Cat #A-21463), 1:200 dilution Alexa Fluor 488 donkey anti-goat IgG (Invitrogen; Cat #A 11055), 1:1000 dilution Alexa Fluor 647 chicken anti-rabbit IgG (Invitrogen; Cat #A21443), and 1:1000 dilution Alexa Fluor 488 chicken anti-mouse IgG (Invitrogen; Cat #A21200).
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by the area of the cell. Background was eliminated based on acceptance criteria for gray-scale ranges between 200 and 4500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control.
  • Results Results for representative differentiation markers are shown in FIG. 21 and Table 15 for cells harvested from each step of differentiation.
  • FIGS. 21A and B flow cytometric results for CXCR4 are shown for various treatments during the first step of definitive endoderm differentiation.
  • FIG. 21A shows the effects on CXCR4 expression from treatment with various compounds in combination with activin A.
  • FIG. 21B shows the effects on CXCR4 from treatment with various compounds in combination with GDF-8.
  • Compounds of the present invention in combination with activin A did not enhance CXCR4 expression. However, all of the compounds of the present invention tested in this Example enhanced CXCR4 expression in combination with GDF-8.
  • FIGS. 21C and 21D normalized RT-PCR values for various differentiation markers at the end of step one of differentiation are shown for treatments applied during step one of the protocol, using selected compounds of the present invention in combination with activin A ( FIG. 21C ) or in combination with GDF-8 ( FIG. 21D ). Similar normalized RT-PCR values were evaluated at the conclusion of step three of the differentiation protocol ( FIGS. 21E and 21F ) and at the end of step four of the differentiation protocol ( FIGS. 21G and 21H ) and at the end of step 5 of the differentiation protocol ( FIGS. 21I and 21J ).
  • Cell passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen; Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the cell monolayer with MEF conditioned medium and gentle scraping to recover cell clusters.
  • Cell clusters were centrifuged at low speed in MEF conditioned medium to remove residual dispase and then evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF (PeproTech Inc.; Cat #100-18B) for seeding on reduced growth factor MATRIGEL (BD Biosciences; Cat #356231)-coated 6-well plates (Nunc; Cat #140685) at a 1:3 ratio using volumes of 2.5 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout the time in culture.
  • Step 1 was conducted over three days to generate definitive endoderm cells.
  • differentiation was initiated by aspirating spent culture medium and adding an equal volume of RPMI-1640 basal medium (Invitrogen; Cat #22400) with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (Proliant Biologicals; Cat #SKU 68700) and 8 ng/ml bFGF.
  • cells were exposed to 100 ng/ml activin A (PeproTech; Cat #120-14) with 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF).
  • activin A PeproTech; Cat #120-14
  • Wnt3a R&D Systems; Cat #1324-WN/CF
  • cells were exposed to 100 ng/ml GDF-8 (R&D Systems; Cat #788-G8) with 2.5 ⁇ M Compound 40.
  • cells were exposed to 100 ng/ml GDF-8 (R&D Systems; Cat #788-G8) with 2.5 ⁇ M Compound 202.
  • step 1 of differentiation cells in all treatment groups were fed with RPMI-1640 containing 2% FAF BSA, 8 ng/ml bFGF and either 100 ng/ml activin A (treatment group 1) or 100 ng/ml GDF-8 (treatment groups 2 and 3), without the addition of Wnt3a or a compound of the present invention.
  • treatment group 1 100 ng/ml activin A
  • GDF-8 100 ng/ml GDF-8
  • Step 2 of the differentiation protocol was conducted over three days.
  • Cells for all treatment groups were fed daily with DMEM:F12 (Invitrogen; Cat #11330-032) supplemented with 2% FAF BSA and 50 ng/ml FGF7 (PeproTech; Cat #100-19).
  • Step 3 of the differentiation protocol was conducted over four days.
  • Cells for all treatment groups were fed daily with DMEM-high glucose (Invitrogen; Cat #10569) supplemented with 1% B27 (Invitrogen; Cat #: 17504-044), 50 ng/ml FGF7, 100 ng/ml Noggin (R&D Systems; Cat #3344-NG), 250 nM KAAD-cyclopamine (Calbiochem; Cat #239804), and 2 ⁇ M all-trans retinoic acid (RA) (Sigma-Aldrich; Cat #R2625).
  • Step 4 of the differentiation protocol was conducted over three days.
  • Cells for all treatment groups were fed daily for the first two days with DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin, and 1 ⁇ M ALK5 inhibitor (Axxora; Cat #ALX-270-445).
  • Axxora Cat #ALX-270-445
  • cells were lifted from the substratum by using a 20 ⁇ l tip (Rainin; Cat #RT-L10F) and a cell scraper (Corning; Cat #3008), then transferred to a 50 ml tube.
  • the cells were allowed to sediment by gravity, and the supernatant was aspirated without disturbing the cell pellet.
  • Cells were resuspended in DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin and 1 ⁇ M ALK5 inhibitor, then cultured overnight in six-well Costar Ultra Low Attachment Microplates (Corning Inc., Cat #3471). On the following day, cells in suspension culture were collected and counted. Aliquots of 10 ⁇ 10 6 cells/mouse were used for transplantation. Aliquots of 0.5 ⁇ 10 6 cells were collected for RT-PCR analysis.
  • FIG. 22 panel A shows flow cytometric results for definitive endoderm cells generated at the end of step 1 for each of the respective treatment groups.
  • Treatment with activin A and Wnt3a or treatment with GDF-8 and a compound of the present invention resulted in cells expressing similar levels of CXCR4 (greater than 85%) at the end of step 1, suggesting that an equivalent definitive endoderm population of cells was derived from each treatment group.
  • FIG. 22 Results for RT-PCR analysis for cells from each treatment group at the conclusion of step 4 of the differentiation protocol are shown in FIG. 22 , panel B.
  • mice Five to six-week-old male scid-beige mice (C.B-Igh-1b/GbmsTac-Prkdc scid -Lyst bg N7) were purchased from Taconic Farms. Mice were housed in microisolator cages with free access to sterilized food and water. In preparation for surgery, mice were identified by ear tagging, their body weight was measured, and their blood glucose was determined using a hand held glucometer (LifeScan; One Touch).
  • mice On the day of surgery, mice were anesthetized with a mixture of isolflurane and oxygen, and the surgical site was shaved with small animal clippers. Mice were dosed with 0.1 mg.kg Buprenex subcutaneously 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 incision was made through the skin and muscle layers. The left kidney was externalized and kept moist with 0.9% sodium chloride. A 24G ⁇ 3 ⁇ 4′′ I.V. 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.
  • mice During preoperative preparation of the mice, cells for transplant were centrifuged in a 1.5 mL microfuge tube, and most of the supernatant was removed, leaving sufficient medium to collect the pellet of cells. The cells were collected into a Rainin Pos-D positive displacement pipette tip, and the pipette was inverted to allow the cells to settle by gravity. Excess medium was dispensed leaving a packed cell preparation for transplant. For transplantation, the Pos-D pipette tip was placed firmly in the hub of the catheter, and the cells were dispensed from the pipette through the catheter under the kidney capsule for delivery to the distal pole of the kidney.
  • the lumen of the catheter was flushed with a small volume of culture medium to deliver any remaining cells, and the catheter was withdrawn.
  • the kidney capsule was sealed with a low temperature cautery, and the kidney was returned to its original anatomical position.
  • the muscle was closed with continuous sutures using 5-0 VICRYL sutures, and the skin was closed with wound clips.
  • the mouse was removed from anesthesia and allowed to fully recover. Mice were dosed with 1.0 mg.kg Metacam subcutaneously post-operatively.
  • mice were weighed once per week and blood glucose was measured twice per week. At various intervals following transplantation, mice were dosed with 3 g/kg glucose IP, and blood was drawn 60 minutes following glucose injection via the retro-orbital sinus into microfuge tubes containing a small amount of heparin. The blood was centrifuged, and the plasma was placed into a second microfuge tube, frozen on dry ice, for storage at ⁇ 80° C. until the human C-peptide assay was performed. Human C-peptide levels were determined using the Mercodia/ALPCO Diagnotics Ultrasensitive C-peptide ELISA according to the manufacturer's instructions.
  • ELISA results for human C-peptide are shown in FIG. 23 for mice transplanted with cells from each of the respective treatment groups. No circulating human C-peptide was detected at four weeks post-transplant for any mice receiving cells from any of the treatment groups. At 8-weeks post-transplant, detectable C-peptide was found in one of two mice receiving cells treated with activin A and Wnt3a; one of three mice receiving cells treated with GDF-8 and Compound 40; and two of three mice receiving cells treated with GDF-8 and Compound 202. These results suggest that an equivalent endocrine precursor cell population could be derived from the differentiation protocol with GDF-8 and a small molecule and that the cells further matured in vivo to a glucose responsive, insulin secreting cell.
  • Cell passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (Invitrogen; Cat #17105-041) for 5 to 7 minutes at 37° C. followed by rinsing the cell monolayer with MEF conditioned medium and gentle scraping to recover cell clusters.
  • dispase Invitrogen; Cat #17105-041
  • Cell clusters were centrifuged at low speed in MEF conditioned medium to remove residual dispase and then evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF (PeproTech Inc.; Cat #100-18B) for seeding on reduced growth factor MATRIGELTM (BD Biosciences; Cat #356231)-coated 6-well plates (Nunc; Cat #140685) at a 1:3 ratio using volumes of 2.5 ml/well. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Plates were maintained at 37° C., 5% CO 2 throughout culture.
  • Step 1 was conducted over three days to generate cells expressing markers characteristic of the definitive endoderm lineage.
  • differentiation was initiated by aspirating spent culture medium and adding an equal volume of RPMI-1640 basal medium (Invitrogen; Cat #22400) with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (Proliant Biologicals; Cat #SKU 68700) and 8 ng/ml bFGF.
  • duplicate sets of cells were treated with 100 ng/ml GDF-8 (R&D Systems; Cat #788-G8) and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF).
  • duplicate sets of cells were treated with 100 ng/ml GDF-8 and 2.5 ⁇ M Compound 40.
  • cells in all treatment groups were fed with RPMI-1640 containing 2% FAF BSA, 8 ng/ml bFGF and 100 ng/ml GDF-8 but without the addition of Wnt3a or Compound 40.
  • one well from each treatment group was collected for FACS analysis.
  • Step 2 of the differentiation protocol was carried out over three days.
  • Cells for all treatment groups were fed daily with DMEM:F12 (Invitrogen; Cat #11330-032) supplemented with 2% FAF BSA and 50 ng/ml FGF7 (PeproTech; Cat #100-19).
  • Step 3 of the differentiation protocol was carried out over four days.
  • Cells for all treatment groups were fed daily with DMEM-high glucose (Invitrogen; Cat #10569) supplemented with 1% B27 (Invitrogen; Cat #: 17504-044), 50 ng/ml FGF7, 100 ng/ml Noggin (R&D Systems; Cat #3344-NG), 250 nM KAAD-cyclopamine (Calbiochem; Cat #239804), and 2 ⁇ M all-trans retinoic acid (RA) (Sigma-Aldrich; Cat #R2625).
  • Step 4 of the differentiation protocol was carried out over three days.
  • Cells for all treatment groups were fed daily with DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin and 1 ⁇ M ALK5 inhibitor (Axxora; Cat #ALX-270-445), and 100 ng/ml GDF-8 (R&D Systems; Cat #788-G8) during the first two days.
  • Axxora 100 ng/ml Noggin and 1 ⁇ M ALK5 inhibitor
  • GDF-8 R&D Systems
  • Step 4 of the differentiation protocol was carried out over three days.
  • Cells for all treatment groups were fed daily with DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin and 1 ⁇ M ALK5 inhibitor (Axxora; Cat #ALX-270-445), and 100 ng/ml GDF-8 (R&D Systems; Cat #788-G8) during the first two days.
  • cells were harvested from the 6-well plates using a 20
  • Cells were resuspended in DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin, and 1 ⁇ M ALK5 inhibitor, then cultured overnight in six-well Costar Ultra Low Attachment Microplates (Corning Inc., Cat #3471). On the following day, cells in suspension culture were collected and counted. Aliquots of 10 ⁇ 10 6 cells/mouse were used for transplantation. Aliquots of 0.5 ⁇ 10 6 cells were collected for RT-PCR analysis.
  • FIG. 24A shows flow cytometric results for definitive endoderm cells generated at the end of step 1 for each of the respective treatment groups.
  • Results for treatment with GDF-8 and Wnt3a or treatment with GDF-8 and Compound 40 expressed similar levels of CXCR4 at the end of step 1, suggesting that an equivalent and robust definitive endoderm population of cells resulted from each treatment group.
  • Duplicate treatment sets were in strong agreement. Results prior to transplant for RT-PCR analysis at the conclusion of step 4 of the differentiation protocol are shown in FIG. 24B .
  • mice Five to six-week-old male scid-beige mice (C.B-Igh-1bGbmsTac-Prkdc scid -Lyst bg N7) were purchased from Taconic Farms. Mice were housed in microisolator cages with free access to sterilized food and water. In preparation for surgery, mice were identified by ear tagging, their body weight was measured, and their blood glucose was determined using a hand held glucometer (LifeScan; One Touch). On the day of surgery, mice were anesthetized with a mixture of isolflurane and oxygen, and the surgical site was shaved with small animal clippers.
  • mice were dosed with 0.1 mg.kg Buprenex subcutaneously 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 incision was made through the skin and muscle layers.
  • the left kidney was externalized and kept moist with 0.9% sodium chloride.
  • a 24G ⁇ 3 ⁇ 4′′ I.V. 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.
  • cells for transplant were centrifuged in a 1.5 mL microfuge tube, and most of the supernatant was removed, leaving sufficient medium to collect the pellet of cells.
  • the cells were collected into a Rainin Pos-D positive displacement pipette tip, and the pipette was inverted to allow the cells to settle by gravity. Excess medium was dispensed leaving a packed cell preparation for transplant.
  • the Pos-D pipette tip was placed firmly in the hub of the catheter, and the cells were dispensed from the pipette through the catheter under the kidney capsule for delivery to the distal pole of the kidney.
  • the lumen of the catheter was flushed with a small volume of culture medium to deliver any remaining cells, and the catheter was withdrawn.
  • the kidney capsule was sealed with a low temperature cautery, and the kidney was returned to its original anatomical position.
  • the muscle was closed with continuous sutures using 5-0 vicryl, and the skin was closed with wound clips.
  • the mouse was removed from anesthesia and allowed to fully recover. Mice were dosed with 1.0 mg.kg Metacam subcutaneously post-operatively.
  • mice were weighed once per week and blood glucose was measured twice per week. At various intervals following transplantation, mice were dosed with 3 g/kg glucose IP, and blood was drawn 60 minutes following glucose injection via the retro-orbital sinus into microfuge tubes containing a small amount of heparin. The blood was centrifuged, and the plasma was placed into a second microfuge tube, frozen on dry ice, for storage at ⁇ 80° C. until the human C-peptide assay was performed. Human C-peptide levels were determined using the Mercodia/ALPCO Diagnotics Ultrasensitive C-peptide ELISA according to the manufacturer's instructions. ELISA results for human C-peptide are shown in FIGS.
  • mice transplanted with cells from each of the respective treatment groups Similar levels of human C-peptide were detectable at 8 weeks post-transplant for each treatment category, indicating that an equivalent endocrine precursor cell population could be derived from the differentiation protocol using GDF-8 and Wnt3a or GDF-8 and a compound of the present invention.
  • a subset of 14 proprietary small molecules, known to have specificity for the CDK, GSK3, and/or TRK signaling pathways were evaluated for their potential to differentiate human embryonic stem cells to cells expressing markers characteristic of the definitive endoderm lineage.
  • Cells were allowed to attach and then recover log phase growth over a 1 to 3 day time period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • Assay was initiated by aspirating culture medium from each well followed by three washes in PBS (Invitrogen; Cat #14190) to remove residual growth factors. On the first day of assay, test volumes of 200 ⁇ l per well were added to each well using DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.5% FCS (HyClone; Cat #SH30070.03) and 100 ng/ml GDF-8 (R&D Systems, Cat #788-G8) plus 2.5 ⁇ M compound. A parallel set of test samples were treated in an identical manner but omitting GDF-8 from the medium.
  • test volumes of 100 ⁇ l per well were added to each well using DMEM:F12 base medium supplemented with 2% FCS plus 100 ng/ml GDF-8 (R&D Systems, Cat #788-G8). GDF-8 was omitted from test samples that did not get treated with GDF-8 on the first day of assay.
  • Positive control samples contained the same base medium supplemented with FCS and 100 ng/ml recombinant human activin A (PeproTech; Cat #120-14) throughout the four day assay along with Wnt3a (20 ng/ml) addition on days 1 and 2.
  • Negative control samples contained DMEM:F12 base medium supplemented with FCS.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell multiplied by area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 and 3500. Average data from triplicate wells were collected. The percentage of treated wells relative to the positive control was calculated.
  • Results for this screen are shown in Table 17. None of the small molecules induced significant SOX17 expression in the absence of GDF-8 during the four day differentiation process. Compound 34 served as an experimental control and induced significant SOX17 expression in the presence of GDF-8, equivalent to levels observed with the positive control using activin A and Wnt3a. The remaining compounds of the present invention tested in this example showed a range of activities with weak to moderate induction of SOX17 expression. Of note, differentiation activity in this subset of compounds was observed in association with selectivity for all three enzymatic signal pathways, making it difficult to conclusively determine a clear mechanism of action.
  • an analog search was conducted and 118 analogues were found. Initial screening determined that some analogues were able to induce definitive endoderm differentiation in the absence of activin A in combination with other growth factors. It was important to determine if these analogues could also induce definitive endoderm differentiation in combination with only GDF-8.
  • Cells were allowed to attach and then recover log phase growth over a 1 to 3 day time period, feeding daily with MEF conditioned medium supplemented with 8 ng/ml bFGF (R&D Systems; Cat #233-FB). Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • test volumes of 200 ⁇ l per well were added to each well using DMEM:F12 base medium (Invitrogen; Cat #11330-032) supplemented with 0.5% FCS (HyClone; Cat #SH30070.03) and 200 ng/ml GDF-8 (R&D Systems, Cat #788-G8) plus 2.5 ⁇ M compound.
  • test volumes of 100 ⁇ l per well were added to each well using DMEM:F12 base medium supplemented with 2% FCS plus 200 ng/ml GDF-8 (R&D Systems, Cat #788-G8).
  • Positive control samples contained the same base medium supplemented with FCS and 100 ng/ml recombinant human activin A (PeproTech; Cat #120-14) throughout the four-day assay along with Wnt3a (20 ng/ml) on days 1 and 2.
  • Negative control samples contained DMEM:F12 base medium supplemented with FCS, adding Wnt3a on days 1 and 2 but omitting treatment with activin A.
  • Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 488. Exposure times were optimized from positive control wells and from untreated negative control wells stained with secondary antibody alone. Images from 15 fields per well were acquired to compensate for any cell loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX17 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei was determined based on gray-scale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set.
  • Total SOX17 protein expression was reported as total intensity or integrated intensity, defined as total fluorescence of the cell times area of the cell. Background was eliminated based on acceptance criteria of gray-scale ranges between 200 to 3500. Total intensity data were normalized by dividing total intensities for each well by the average total intensity for the positive control. Normalized data were calculated for averages and standard deviations for each replicate set.
  • H1 p49C3 cells were routinely grown on Cytodex3 beads (GE Healthcare Life Sciences, NJ) in a 125 ml spinner flask, according to the methods described in U.S. Patent Application No. 61/116,447. After seven days, cells and beads were transferred to a 6 well plate (Vendor; Cat #XXX) at a ratio of 30 cm 2 bead surface area per well, and the plate was placed on a rocking platform.
  • Cells on beads in the positive control treatment well were differentiated with addition of 100 ng/ml activin A (PeproTech; Cat #120-14) and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF) for two days followed by 100 ng/ml activin A and 8 ng/ml bFGF (PeproTech Inc.; Cat #: 100-18B) for one day in RPMI-1640 (Invitrogen; Cat #: 22400) with 2% Fatty Acid Free BSA (MP Biomedicals, Inc; Cat #152401) using volumes of 2 ml/well.
  • Compound 34 at a final concentration of 2.5 ⁇ M was added to a negative control treatment well (designated CMP alone) in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) for three days in the absence of any other growth factor treatment.
  • a third treatment well (designated CMP+8) received Compound 34at 2.5 ⁇ M plus 50 ng/ml GDF-8 (R&D Systems, Cat #788-G8) in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) for three days.
  • a fourth treatment well received Compound 34 at 2.5 ⁇ M with 50 ng/ml GDF-8 and 50 ng/ml PDGF-D in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) for three days.
  • a fifth treatment well received Compound 34at 2.5 ⁇ M with 50 ng/ml GDF-8, 50 ng/ml PDGF-D, and 50 ng/ml VEGF in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) for three days.
  • a sixth treatment well (designated CMP+8+D+V+M) received Compound 34at 2.5 ⁇ M with 50 ng/ml GDF-8, 50 ng/ml PDGF-D, 50 ng/ml VEGF, and 20 ng/ml Muscimol in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) for three days. All media and treatments were exchanged daily.
  • Results are shown in FIG. 25 .
  • panel A similar numbers of cells were recovered for all treatment groups undergoing differentiation.
  • panel B cells treated with the Compound 34 alone did not differentiate into CXCR4 positive cells.
  • the positive control treatment adding activin A and Wnt3a during differentiation, induced expression of CXCR4 in 68% of the resulting cell population.
  • Compound 34 added with the various growth factor combinations induced CXCR4 expression in 50% of the cells, on average.
  • equivalent levels of CXCR4 expression were observed during treatment with Compound 34 in combination with a single growth factor, GDF-8, or in combination with multiple growth factors that included GDF-8. This proves that Compound 34 in combination with at least GDF-8 can substitute for activin A and Wnt3a to promote definitive endoderm differentiation.
  • This example also shows that the treatment procedure is effective for cells grown and differentiated on microcarrier beads.
  • GDF-8 is able to replace activin A to differentiate human embryonic stem cells to cells expressing markers characteristic of the definitive endoderm lineage. It was important to know the relative potencies of GDF-8 and activin A with respect to definitive endoderm formation. A dose response assay was conducted using equivalent concentrations of each growth factor to compare results during human embryonic stem cell differentiation.
  • the compounds of the present invention used in combination with GDF-8 during definitive endoderm differentiation were evaluated for their ability to induce cell proliferation. Results were compared to treatment with activin A or GDF-8 alone.
  • Cell clusters were split at a 1:3 or 1:4 ratio for routine maintenance culture or a 1:1 ratio for immediate assay. All human embryonic stem cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotypic phenotype and for absence of mycoplasma contamination.
  • Cell clusters used in the assay were evenly resuspended in MEF conditioned medium supplemented with 8 ng/ml bFGF and seeded onto reduced growth factor MATRIGELTM-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat #6005182) in volumes of 100 ⁇ l/well.
  • MEF conditioned medium supplemented with 8 ng/ml bFGF was used for initial seeding and expansion.
  • Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium.
  • a background set of wells in each assay plate was not seeded with cells but was treated throughout assay with basal media conditions. Plates were maintained at 37° C., 5% CO 2 in a humidified box throughout the duration of assay.
  • the assay was initiated by aspirating the culture medium from each well and adding back a final aliquot (100 ⁇ l) of test medium. Test conditions were performed in triplicate over a total three-day assay period, feeding daily by aspirating and replacing the medium from each well with fresh test medium. Identical assays were set up simultaneously in parallel for evaluation at the end of 24, 48, and 72 hours.
  • Control conditions included the following, with final growth factor concentrations as indicated: 1) basal medium with 2% FAF BSA; 2) 100 ng/ml activin A (PeproTech; Cat #120-14) with 8 ng/ml bFGF (PeproTech; Cat #100-18B); 3) 100 ng/ml activin A with 8 ng/ml bFGF and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF); 4) 100 ng/ml GDF-8 (R&D Systems, Cat #788-G8) with 8 ng/ml bFGF; 5) GDF-8 with 8 ng/ml bFGF and 20 ng/ml Wnt3a.
  • MTS Assay At the conclusion of 24, 48, or 72 hours of culture, one set of assay plates was subjected to a MTS assay (Promega; Cat #G3581), following the manufacturer's instructions. In brief, 20 ⁇ l of MTS was added to each well, and assay plates were incubated at 37° C., 5% CO 2 for four hours prior to taking OD490 readings. Statistical measures were calculated minus background (i.e. treatment wells without cells) to determine mean values for each triplicate set in addition to a standard error of the mean.
  • the MTS assay is a measure of cellular metabolic activity in the enzymatic reduction of a tetrazolium compound to a formazan product.
  • the MTS assay can be used as a comparative indicator of cell viability.
  • MTS assays evaluated in parallel at sequential time points can add additional information regarding increases in cellular metabolic activity which in turn can be correlated with cell proliferation for each treatment condition.
  • FIG. 26 panel A shows OD490 readings for all control treatments over the three day assay period. Cells treated with conditioned medium showed little change in OD490 over three days, indicating that cell numbers in this treatment group remained static.
  • FIG. 26 , panel B through FIG. 26 , panel I show MTS assay results for treatment with a small molecule inhibitor in combination with GDF-8.
  • OD490 readings from treatments with a compound of the present invention and GDF-8 were equivalent to or exceeded results from treatment with activin A.
  • an optimal concentration of each small molecule combined with GDF-8 resulted in improved OD490 readings over the three day assay relative to treatment with GDF-8 alone. This suggests that the compounds of the present invention are important for inducing proliferation and expansion of a cell population during definitive endoderm differentiation.
  • H1 p45 cells were grown on Cytodex3 beads (GE Healthcare; Cat #17-0485-01) in a 6 well ultra low attachment plate (Costar; Cat #3471) placed on a rocking platform at about 1 rotation every 10 seconds (Vari Mix, Thermo Scientific, Cat # M79735 ). MEF conditioned media was changed daily for six days. Then the media was changed to the following treatments to initiate endoderm differentiation.
  • Cells on beads in the positive control treatment well were differentiated with addition of 100 ng/ml activin A (PeproTech; Cat #120-14), 8 ng/ml bFGF (PeproTech Inc.; Cat #: 100-18B), and 20 ng/ml Wnt3a (R&D Systems; Cat #1324-WN/CF) for one day followed by 100 ng/ml activin A and 8 ng/ml bFGF (PeproTech Inc.; Cat #: 100-18B) for two days in RPMI-1640 (Invitrogen; Cat #: 22400) with 2% Fatty Acid Free BSA (Proliant Biomedicals, Inc; SKU #68700) using volumes of 2 ml/well.
  • a second treatment well (designated GDF-8+MCX) received Compound 202 at 2.5 ⁇ M plus 200 ng/ml GDF-8 (R&D Systems, Cat #788-G8) and 8 ng/ml bFGF for one day followed by two days with 200 ng/ml GDF-8 and 8 ng/ml bFGF in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) media.
  • a third treatment well (designated GDF-8+Wnt) received 200 ng/ml GDF-8 with 20 ng/ml Wnt3a and 8 ng/ml bFGF for one day followed by two days with 200 ng/ml GDF-8 and 8 ng/ml bFGF in RPMI-1640 with 2% Fatty Acid Free BSA (2 ml/well) media. All media and treatments were exchanged daily.
  • stage 3 the endodermal genes PDX1, HNF4 alpha, and CDX2 are expressed in the cells ( FIGS. 27C , D).
  • Treatment of the cells with GDF-8 and a compound of the present invention during stage one of differentiation resulted in better expression of Pdx1 than the control differentiation treatment.
  • endodermal genes were up regulated further ( FIGS. 27E , F).
US12/494,789 2008-06-30 2009-06-30 Differentiation of Pluripotent Stem Cells Abandoned US20100015711A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US12/494,789 US20100015711A1 (en) 2008-06-30 2009-06-30 Differentiation of Pluripotent Stem Cells
US13/434,370 US9593305B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US13/434,392 US20120190112A1 (en) 2008-06-30 2012-03-29 Differentiation of Pluripotent Stem Cells
US13/434,409 US9593306B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US15/457,864 US10351820B2 (en) 2008-06-30 2017-03-13 Methods for making definitive endoderm using at least GDF-8
US15/457,893 US10233421B2 (en) 2008-06-30 2017-03-13 Differentiation of pluripotent stem cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US7690008P 2008-06-30 2008-06-30
US7691508P 2008-06-30 2008-06-30
US7690808P 2008-06-30 2008-06-30
US12/494,789 US20100015711A1 (en) 2008-06-30 2009-06-30 Differentiation of Pluripotent Stem Cells

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US13/434,392 Continuation US20120190112A1 (en) 2008-06-30 2012-03-29 Differentiation of Pluripotent Stem Cells
US13/434,409 Continuation US9593306B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US13/434,370 Continuation US9593305B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells

Publications (1)

Publication Number Publication Date
US20100015711A1 true US20100015711A1 (en) 2010-01-21

Family

ID=41100882

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/494,789 Abandoned US20100015711A1 (en) 2008-06-30 2009-06-30 Differentiation of Pluripotent Stem Cells
US13/434,370 Active 2029-07-11 US9593305B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US13/434,409 Active 2029-09-19 US9593306B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US13/434,392 Abandoned US20120190112A1 (en) 2008-06-30 2012-03-29 Differentiation of Pluripotent Stem Cells
US15/457,893 Active US10233421B2 (en) 2008-06-30 2017-03-13 Differentiation of pluripotent stem cells
US15/457,864 Active 2029-12-09 US10351820B2 (en) 2008-06-30 2017-03-13 Methods for making definitive endoderm using at least GDF-8

Family Applications After (5)

Application Number Title Priority Date Filing Date
US13/434,370 Active 2029-07-11 US9593305B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US13/434,409 Active 2029-09-19 US9593306B2 (en) 2008-06-30 2012-03-29 Differentiation of pluripotent stem cells
US13/434,392 Abandoned US20120190112A1 (en) 2008-06-30 2012-03-29 Differentiation of Pluripotent Stem Cells
US15/457,893 Active US10233421B2 (en) 2008-06-30 2017-03-13 Differentiation of pluripotent stem cells
US15/457,864 Active 2029-12-09 US10351820B2 (en) 2008-06-30 2017-03-13 Methods for making definitive endoderm using at least GDF-8

Country Status (15)

Country Link
US (6) US20100015711A1 (es)
EP (2) EP2310492B1 (es)
JP (1) JP5734183B2 (es)
KR (4) KR20180018839A (es)
CN (1) CN102159703B (es)
AU (1) AU2009267137A1 (es)
BR (1) BRPI0914116A2 (es)
CA (1) CA2729121C (es)
ES (2) ES2552240T3 (es)
HK (2) HK1156665A1 (es)
MX (1) MX2011000125A (es)
PL (2) PL2942392T3 (es)
RU (2) RU2533805C2 (es)
WO (1) WO2010002846A1 (es)
ZA (1) ZA201100758B (es)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070254359A1 (en) * 2006-04-28 2007-11-01 Lifescan, Inc. Differentiation of human embryonic stem cells
US20090325293A1 (en) * 2008-04-24 2009-12-31 Janet Davis Treatment of pluripotent cells
US20100112693A1 (en) * 2008-10-31 2010-05-06 Alireza Rezania Differentiation of Human Embryonic Stem Cells
US20110091971A1 (en) * 2008-06-30 2011-04-21 Janet Davis Differentiation of Pluripotent Stem Cells
US20110151561A1 (en) * 2009-12-23 2011-06-23 Janet Davis Differentiation of human embryonic stem cells
US20110151560A1 (en) * 2009-12-23 2011-06-23 Jean Xu Differentiation of human embryonic stem cells
WO2012030539A3 (en) * 2010-08-31 2012-05-31 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
WO2012030538A3 (en) * 2010-08-31 2012-06-07 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
WO2013169769A1 (en) 2012-05-07 2013-11-14 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endoderm
WO2013184888A1 (en) 2012-06-08 2013-12-12 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
WO2014105546A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
WO2014106141A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Suspension and clustering of human pluripotent cells for differentiation into pancreatic endocrine cells
WO2014105543A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
US8778673B2 (en) 2004-12-17 2014-07-15 Lifescan, Inc. Seeding cells on porous supports
US8785184B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US8785185B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9012218B2 (en) 2008-10-31 2015-04-21 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9062290B2 (en) 2007-11-27 2015-06-23 Lifescan, Inc. Differentiation of human embryonic stem cells
US9074189B2 (en) 2005-06-08 2015-07-07 Janssen Biotech, Inc. Cellular therapy for ocular degeneration
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US9096832B2 (en) 2007-07-31 2015-08-04 Lifescan, Inc. Differentiation of human embryonic stem cells
US9181528B2 (en) 2010-08-31 2015-11-10 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
WO2015175307A1 (en) 2014-05-16 2015-11-19 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
WO2016100035A1 (en) 2014-12-19 2016-06-23 Janssen Biotech, Inc. Suspension culturing of pluripotent stem cells
US20160237401A1 (en) * 2013-10-09 2016-08-18 Cambridge Enterprise Limited In Vitro Production of Foregut Stem Cells
US9434920B2 (en) 2012-03-07 2016-09-06 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US9593305B2 (en) 2008-06-30 2017-03-14 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9752125B2 (en) 2010-05-12 2017-09-05 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US20170280121A1 (en) * 2014-04-01 2017-09-28 Canon Kabushiki Kaisha Imaging apparatus and image processing system
WO2017180361A1 (en) 2016-04-14 2017-10-19 Janssen Biotech, Inc. Differentiation of pluripotent stem cells to intestinal midgut endoderm cells
WO2017222879A1 (en) 2016-06-21 2017-12-28 Janssen Biotech, Inc. Generation of human pluripotent stem cell derived functional beta cells showing a glucose-dependent mitochondrial respiration and two-phase insulin secretion response
US9969972B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Pluripotent stem cell culture on micro-carriers
US9969981B2 (en) 2010-03-01 2018-05-15 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
US9969973B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
US10030229B2 (en) 2013-06-11 2018-07-24 President And Fellows Of Harvard College SC-β cells and compositions and methods for generating the same
US10066203B2 (en) 2008-02-21 2018-09-04 Janssen Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US10076544B2 (en) 2009-07-20 2018-09-18 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10190096B2 (en) 2014-12-18 2019-01-29 President And Fellows Of Harvard College Methods for generating stem cell-derived β cells and uses thereof
US10253298B2 (en) 2014-12-18 2019-04-09 President And Fellows Of Harvard College Methods for generating stem cell-derived beta cells and methods of use thereof
US10358628B2 (en) 2011-12-22 2019-07-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
US10443042B2 (en) 2014-12-18 2019-10-15 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived beta cells and uses thereof
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11466256B2 (en) 2018-08-10 2022-10-11 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
US11945795B2 (en) 2017-11-15 2024-04-02 Vertex Pharmaceuticals Incorporated Islet cell manufacturing compositions and methods of use

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2412800A1 (en) * 2010-07-29 2012-02-01 Koninklijke Nederlandse Akademie van Wetenschappen Liver organoid, uses thereof and culture method for obtaining them
CN102428172A (zh) 2009-03-20 2012-04-25 新加坡科技研究局 多潜能和多能细胞在微载体上的培养
US9234170B2 (en) * 2010-04-25 2016-01-12 Mount Sinai School Of Medicine Generation of anterior foregut endoderm from pluripotent cells
US9046992B2 (en) 2010-10-01 2015-06-02 Z124 Gesture controls for multi-screen user interface
AU2011308567B2 (en) 2010-10-01 2015-09-03 Fundacion Centro Nacional De Investigaciones Oncologicas, Carlos Iii Manipulation of stem cell function by p53 isoforms
EP2723852B1 (en) 2011-06-21 2021-11-17 Novo Nordisk A/S Efficient induction of definitive endoderm from pluripotent stem cells
US20140242038A1 (en) * 2011-10-11 2014-08-28 The Trustees Of Columbia University In The City Of New York Method for generating beta cells
EP2829606A4 (en) 2012-03-22 2016-02-24 Takara Bio Inc PROCESS FOR PRODUCING VIRAL VECTOR
WO2013146480A1 (ja) * 2012-03-29 2013-10-03 タカラバイオ株式会社 遺伝子導入方法
JP6470687B2 (ja) 2012-09-03 2019-02-13 ノヴォ ノルディスク アー/エス 小分子を用いた多能性幹細胞からの膵臓内胚葉の作製
ME03042B (me) 2012-11-19 2018-10-20 Novartis Ag Jedinjenja i kompozicije za tretman parazitskih oboljenja
EP2961829A4 (en) 2013-02-27 2016-08-17 Univ California GENERATION OF THYME PEPPER THERAPEUTIC CELLS IN VITRO
EP2964230A4 (en) 2013-03-07 2016-10-26 Califia Bio Inc KINASEHEMMER WITH MIXED ABSTRACT AND TREATMENT PROCESS THEREWITH
SG11201603045VA (en) * 2013-11-01 2016-05-30 Janssen Biotech Inc Suspension and clustering of human pluripotent stem cells for differentiation into pancreatic endocrine cells
KR101686229B1 (ko) * 2015-04-03 2016-12-13 연세대학교 산학협력단 D형 혈소판유래성장인자로 처리된 지방줄기세포를 유효성분으로 함유하는 발모 촉진용 조성물
US20210255185A1 (en) * 2018-06-08 2021-08-19 Novartis Ag Cell-based assay for measuring drug product potency
EP3976236A1 (en) 2019-05-31 2022-04-06 W.L. Gore & Associates Inc. A biocompatible membrane composite
WO2020243665A1 (en) 2019-05-31 2020-12-03 W. L. Gore & Associates, Inc. A biocompatible membrane composite
JP7328362B2 (ja) 2019-05-31 2023-08-16 ダブリュ.エル.ゴア アンド アソシエイツ,インコーポレイティド 制御された酸素拡散距離を伴う細胞カプセル化デバイス
US20220233298A1 (en) 2019-05-31 2022-07-28 W. L. Gore & Associates, Inc. A biocompatible membrane composite
US11344859B2 (en) 2019-11-27 2022-05-31 Smartdyelivery Gmbh Reactor for the preparation of a formulation
EP4103183A4 (en) * 2020-02-14 2024-02-28 Salk Inst For Biological Studi MACROCYCLIC ULK1/2 INHIBITORS

Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4557264A (en) * 1984-04-09 1985-12-10 Ethicon Inc. Surgical filament from polypropylene blended with polyethylene
US5567612A (en) * 1986-11-20 1996-10-22 Massachusetts Institute Of Technology Genitourinary cell-matrix structure for implantation into a human and a method of making
US5759830A (en) * 1986-11-20 1998-06-02 Massachusetts Institute Of Technology Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5770417A (en) * 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5834308A (en) * 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6022743A (en) * 1986-04-18 2000-02-08 Advanced Tissue Sciences, Inc. Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
US6306424B1 (en) * 1999-06-30 2001-10-23 Ethicon, Inc. Foam composite for the repair or regeneration of tissue
US6331298B1 (en) * 1992-03-28 2001-12-18 Renovo Limited Wound healing and treatment of fibrotic disorders
US6333029B1 (en) * 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US20020072117A1 (en) * 2000-01-11 2002-06-13 Chunhui Xu Human feeder cells that support proliferation of undifferentiated pluripotent stem cells
US6458589B1 (en) * 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
US6458593B1 (en) * 1999-01-21 2002-10-01 Vitro Diagnostics, Inc. Immortalized cell lines and methods of making the same
US6509369B2 (en) * 1997-12-29 2003-01-21 Ortho-Mcneil Pharmaceutical, Inc. Anti-inflammatory compounds
US20030082155A1 (en) * 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20030138948A1 (en) * 2001-12-07 2003-07-24 Fisk Gregory J. Islet cells from human embryonic stem cells
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
US6626950B2 (en) * 2001-06-28 2003-09-30 Ethicon, Inc. Composite scaffold with post anchor for the repair and regeneration of tissue
US6656488B2 (en) * 2001-04-11 2003-12-02 Ethicon Endo-Surgery, Inc. Bioabsorbable bag containing bioabsorbable materials of different bioabsorption rates for tissue engineering
US20040015805A1 (en) * 2002-07-22 2004-01-22 Kidd David A. Layout quality analyzer
US20040062753A1 (en) * 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
US20040132729A1 (en) * 1998-05-11 2004-07-08 Francesco Salituro Inhibitors of p38
US20040171623A1 (en) * 2001-07-19 2004-09-02 Reynolds Rachel Heulwen Chemical compounds
US6793945B2 (en) * 1999-01-08 2004-09-21 Sky High, Llc Aqueous anti-apoptotic compositions
US6800480B1 (en) * 1997-10-23 2004-10-05 Geron Corporation Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
US20040209901A1 (en) * 2000-10-23 2004-10-21 Smithkline Beecham Corporation Novel compounds
US20040220393A1 (en) * 2002-11-23 2004-11-04 Ward Donna T. Modulation of HIF1alpha and HIF2alpha expression
US20040241761A1 (en) * 1999-12-13 2004-12-02 Nora Sarvetnick Markers for identification and isolation of pancreatic islet alpha and beta cell progenitors
US20050037488A1 (en) * 2001-08-06 2005-02-17 Maisam Mitalipova Alternative compositions and methods for the culture of stem cells
US20050148070A1 (en) * 2000-03-09 2005-07-07 Thomson James A. Cultivation of primate embryonic stem cells
US20050233446A1 (en) * 2003-12-31 2005-10-20 Parsons Xuejun H Defined media for stem cell culture
US20050244962A1 (en) * 2000-03-09 2005-11-03 Thomson James A Cultivation of primate embryonic stem cells
US20050266554A1 (en) * 2004-04-27 2005-12-01 D Amour Kevin A PDX1 expressing endoderm
US20060003446A1 (en) * 2002-05-17 2006-01-05 Gordon Keller Mesoderm and definitive endoderm cell populations
US20060148081A1 (en) * 2003-12-23 2006-07-06 Olivia Kelly Expansion of definitive endoderm cells
US20070259423A1 (en) * 2006-05-02 2007-11-08 Jon Odorico Method of differentiating stem cells into cells of the endoderm and pancreatic lineage
US20090298178A1 (en) * 2008-06-03 2009-12-03 D Amour Kevin Allen Growth factors for production of definitive endoderm
US8187878B2 (en) * 2004-08-13 2012-05-29 University Of Georgia Research Foundation, Inc. Methods for increasing definitive endoderm differentiation of pluripotent human embryonic stem cells with PI-3 kinase inhibitors

Family Cites Families (230)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3209652A (en) 1961-03-30 1965-10-05 Burgsmueller Karl Thread whirling method
AT326803B (de) 1968-08-26 1975-12-29 Binder Fa G Maschenware sowie verfahren zur herstellung derselben
US3935067A (en) 1974-11-22 1976-01-27 Wyo-Ben Products, Inc. Inorganic support for culture media
CA1201400A (en) 1982-04-16 1986-03-04 Joel L. Williams Chemically specific surfaces for influencing cell activity during culture
US4499802A (en) 1982-09-29 1985-02-19 Container Graphics Corporation Rotary cutting die with scrap ejection
US4537773A (en) 1983-12-05 1985-08-27 E. I. Du Pont De Nemours And Company α-Aminoboronic acid derivatives
US5215893A (en) 1985-10-03 1993-06-01 Genentech, Inc. Nucleic acid encoding the ba chain prodomains of inhibin and method for synthesizing polypeptides using such nucleic acid
US5089396A (en) 1985-10-03 1992-02-18 Genentech, Inc. Nucleic acid encoding β chain prodomains of inhibin and method for synthesizing polypeptides using such nucleic acid
US4737578A (en) 1986-02-10 1988-04-12 The Salk Institute For Biological Studies Human inhibin
NZ229354A (en) 1988-07-01 1990-09-26 Becton Dickinson Co Treating polymer surfaces with a gas plasma and then applying a layer of endothelial cells to the surface
EP0363125A3 (en) 1988-10-03 1990-08-16 Hana Biologics Inc. Proliferated pancreatic endocrine cell product and process
SU1767433A1 (ru) 1989-11-27 1992-10-07 Пермский государственный медицинский институт Способ определени инсулинорезистентности имунного генеза у больных сахарным диабетом I типа
US5837539A (en) 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
EP0628639B1 (en) 1991-04-25 1999-06-23 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin 6 receptor
US5449383A (en) 1992-03-18 1995-09-12 Chatelier; Ronald C. Cell growth substrates
CA2114282A1 (en) 1993-01-28 1994-07-29 Lothar Schilder Multi-layered implant
JP3525221B2 (ja) 1993-02-17 2004-05-10 味の素株式会社 免疫抑制剤
WO1994022915A1 (en) * 1993-03-31 1994-10-13 Pro-Neuron, Inc. Inhibitor of stem cell proliferation and uses thereof
JP2813467B2 (ja) 1993-04-08 1998-10-22 ヒューマン・セル・カルチャーズ・インコーポレーテッド 細胞培養法および培地
US5523226A (en) 1993-05-14 1996-06-04 Biotechnology Research And Development Corp. Transgenic swine compositions and methods
GB9310557D0 (en) 1993-05-21 1993-07-07 Smithkline Beecham Plc Novel process and apparatus
TW257671B (es) 1993-11-19 1995-09-21 Ciba Geigy
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
US6703017B1 (en) 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US6083903A (en) 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
DE69525971T3 (de) 1994-12-29 2013-01-10 Chugai Seiyaku K.K. Verwendung eines pm-1 antikörpers oder eines mh 166 antikörpers zur verstärkung des anti-tumor-effektes von cisplatin oder carboplatin
US5718922A (en) 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
US5908782A (en) 1995-06-05 1999-06-01 Osiris Therapeutics, Inc. Chemically defined medium for human mesenchymal stem cells
US5681561A (en) 1995-06-07 1997-10-28 Life Medical Sciences, Inc. Compositions and methods for improving autologous fat grafting
WO1998030679A1 (en) 1997-01-10 1998-07-16 Life Technologies, Inc. Embryonic stem cell serum replacement
IL132318A0 (en) 1997-04-24 2001-03-19 Ortho Mcneil Pharm Inc Substituted imidazoles useful in the treatment of inflammatory diseases
AU8476698A (en) 1997-07-03 1999-01-25 Osiris Therapeutics, Inc. Human mesenchymal stem cells from peripheral blood
US6670127B2 (en) 1997-09-16 2003-12-30 Egea Biosciences, Inc. Method for assembly of a polynucleotide encoding a target polypeptide
ES2340857T3 (es) 1997-09-16 2010-06-10 Centocor Ortho Biotech Inc. Metodo para la sintesis quimica completa y emsamblaje de genes y genomas.
ATE316795T1 (de) 1998-03-18 2006-02-15 Osiris Therapeutics Inc Mesenchymale stammzellen für die prävention und behandlung von immunantworten bei transplantationen
US6413773B1 (en) 1998-06-01 2002-07-02 The Regents Of The University Of California Phosphatidylinositol 3-kinase inhibitors as stimulators of endocrine differentiation
US7410798B2 (en) 2001-01-10 2008-08-12 Geron Corporation Culture system for rapid expansion of human embryonic stem cells
US6617159B1 (en) * 1998-11-09 2003-09-09 Consorzio Per La Gestione Del Centro Di Biotechnologie Avanzate Serum free medium for chondrocyte cells
US6610540B1 (en) 1998-11-18 2003-08-26 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US6815203B1 (en) 1999-06-23 2004-11-09 Joslin Diabetes Center, Inc. Methods of making pancreatic islet cells
EP1224259A4 (en) 1999-09-27 2005-04-27 Univ Florida INVERSION OF INSULIN DEPENDENT DIABETES BY ISOLATED STEM CELLS, PROGENITOR ISLANDIC CELLS, AND INSULAR TYPE STRUCTURES
US6685936B2 (en) 1999-10-12 2004-02-03 Osiris Therapeutics, Inc. Suppressor cells induced by culture with mesenchymal stem cells for treatment of immune responses in transplantation
US6436704B1 (en) 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
CN1449439A (zh) 2000-06-26 2003-10-15 株式会社雷诺再生医学研究所 细胞级分包括能分化为神经细胞的细胞
ES2263681T3 (es) 2000-12-08 2006-12-16 Ortho-Mcneil Pharmaceutical, Inc. Compuestos de pirrolina indazolil-substituidos como inhibidores de la kinasa.
JP2004526676A (ja) 2000-12-08 2004-09-02 オーソ−マクニール・フアーマシユーチカル・インコーポレーテツド キナーゼ阻害剤として有用な大員複素環式化合物
EP1366148A2 (en) 2001-01-24 2003-12-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE DEPARTMENT OF HEALTH & HUMAN SERVICES Differentiation of stem cells to pancreatic endocrine cells
WO2002059131A1 (en) 2001-01-25 2002-08-01 Millennium Pharmaceuticals, Inc. Formulation of boronic acid compounds
DE10117563B4 (de) 2001-04-07 2006-04-13 Interessengemeinschaft für Rundfunkschutzrechte GmbH Schutzrechtsverwertung & Co. KG. Verfahren zur Verstärkung eines amplituden- und/oder phasenmodulierten Hochfrequenzsignals nach dem Prinzip des Envelope Elimination und Restoration und Schaltungsanordnung
EP1379626A2 (en) 2001-04-19 2004-01-14 DeveloGen Aktiengesellschaft für entwicklungsbiologische Forschung A method for differentiating stem cells into insulin-producing cells
CN100516204C (zh) 2001-04-24 2009-07-22 味之素株式会社 干细胞及其分离方法
WO2002092756A2 (en) 2001-05-15 2002-11-21 Rappaport Family Institute For Research In The Medical Sciences Insulin producing cells derived from human embryonic stem cells
KR100418195B1 (ko) 2001-07-05 2004-02-11 주식회사 우리기술 전력케이블의 다중절연진단장치 및 그 방법
US6617152B2 (en) 2001-09-04 2003-09-09 Corning Inc Method for creating a cell growth surface on a polymeric substrate
EP1298201A1 (en) 2001-09-27 2003-04-02 Cardion AG Process for the production of cells exhibiting an islet-beta-cell-like state
EP1444345A4 (en) 2001-10-18 2004-12-08 Ixion Biotechnology Inc TRANSFORMATION OF STEM CELLS AND LIVER PROGENITORS INTO FUNCTIONAL CELLS OF PANCREAS
CA2468171C (en) 2001-11-15 2015-10-06 Children's Medical Center Corporation Methods of isolation, expansion and differentiation of fetal stem cells from chorionic villus, amniotic fluid, and placenta and therapeutic uses thereof
CA2849201A1 (en) 2001-12-07 2003-07-03 Macropore Biosurgery, Inc. Adipose-derived cell processing unit
WO2003054169A1 (en) 2001-12-21 2003-07-03 Thromb-X Nv Compositions for the in vitro derivation and culture of embryonic stem (es) cell lines with germline transmission capability
WO2003055992A2 (en) 2001-12-28 2003-07-10 Cellartis Ab A method for the establishment of a pluripotent human blastocyst-derived stem cell line
US20030162290A1 (en) 2002-01-25 2003-08-28 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
US20030180268A1 (en) 2002-02-05 2003-09-25 Anthony Atala Tissue engineered construct for supplementing or replacing a damaged organ
JPWO2003087349A1 (ja) 2002-04-17 2005-08-18 大塚製薬株式会社 間葉系細胞から膵β細胞を形成する方法
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
ES2300573T3 (es) 2002-05-08 2008-06-16 Janssen Pharmaceutica Nv Inhibidores de cinasa sustituidos con pirrolina.
GB0210539D0 (en) 2002-05-08 2002-06-19 Univ Edinburgh Control of es cell self renewal and lineage specification, and medium therefor
CN1819838A (zh) 2002-05-28 2006-08-16 贝克顿·迪金森公司 人类胰腺腺泡细胞体外扩增并转分化为胰岛素产生细胞的方法
JP2005531609A (ja) 2002-06-05 2005-10-20 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ キナーゼ阻害剤としてのシスインドリル−マレイミド誘導体
GB0212976D0 (en) 2002-06-06 2002-07-17 Tonejet Corp Pty Ltd Ejection method and apparatus
CN1171991C (zh) 2002-07-08 2004-10-20 徐如祥 人神经干细胞的培养方法
US7838290B2 (en) 2002-07-25 2010-11-23 The Scripps Research Institute Hematopoietic stem cells and methods of treatment of neovascular eye diseases therewith
JP2005534345A (ja) 2002-07-29 2005-11-17 エス セル インターナショナル ピーティーイー リミテッド インスリン陽性、グルコース応答性細胞の分化のための多段階方法
US20040063204A1 (en) 2002-08-14 2004-04-01 Lijun Yang Bone marrow cell differentiation
US7371576B2 (en) 2002-09-06 2008-05-13 Reneuron, Inc. CD56 positive human adult pancreatic endocrine progenitor cells
US9969977B2 (en) 2002-09-20 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
AU2003285172A1 (en) 2002-11-08 2004-06-03 The Johns Hopkins University Human embryonic stem cell cultures, and compositions and methods for growing same
US20060040385A1 (en) 2002-12-05 2006-02-23 Technion Research & Development Foundation Ltd. Cultured human pancreatic islets, and uses thereof
PT2457999T (pt) 2002-12-16 2019-01-28 Technion Res & Dev Foundation Sistema de cultura para células estaminais pluripotentes
US20050118148A1 (en) 2002-12-20 2005-06-02 Roland Stein Compositions and methods related to mammalian Maf-A
RU2359671C2 (ru) 2003-01-29 2009-06-27 Такеда Фармасьютикал Компани Лимитед Способ получения препарата с покрытием
NZ541749A (en) 2003-01-29 2009-06-26 Takeda Pharmaceutical Process for producing coated preparation comprising pioglitazone hydrochloride and a coating material
WO2004073633A2 (en) * 2003-02-14 2004-09-02 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for modulating the development of stem cells
US20070154981A1 (en) 2003-02-14 2007-07-05 The Board Of Trustees Of The Leland Stanford Junior University Insulin-producing cells derived from stem cells
AU2004219851B2 (en) * 2003-03-12 2009-12-17 Reliance Life Sciences Pvt. Ltd. Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells
US20070020242A1 (en) 2003-03-27 2007-01-25 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic pathway
US20060194315A1 (en) 2003-03-31 2006-08-31 Condie Brian G Compositions and methods for the control, differentiaton and/or manipulation of pluripotent cells through a gamma-secretase signaling pathway
US20090203141A1 (en) 2003-05-15 2009-08-13 Shi-Lung Lin Generation of tumor-free embryonic stem-like pluripotent cells using inducible recombinant RNA agents
EP2338980B1 (en) 2003-06-27 2015-04-22 DePuy Synthes Products, LLC Regeneration and repair of neural tissue using postpartum umbilical cord -derived cells
IL161903A0 (en) 2003-07-17 2005-11-20 Gamida Cell Ltd Ex vivo progenitor and stem cell expansion for usein the treatment of disease of endodermally- deri ved organs
ITRM20030395A1 (it) 2003-08-12 2005-02-13 Istituto Naz Per Le Malattie Infettive Lazz Terreno di coltura per il mantenimento, la proliferazione e il differenziamento di cellule di mammifero.
US7569385B2 (en) 2003-08-14 2009-08-04 The Regents Of The University Of California Multipotent amniotic fetal stem cells
US7157275B2 (en) 2003-08-15 2007-01-02 Becton, Dickinson And Company Peptides for enhanced cell attachment and growth
CA2536067A1 (en) 2003-08-27 2005-03-10 Stemcells California, Inc. Enriched pancreatic stem cell and progenitor cell populations, and methods for identifying, isolating and enriching for these populations
US7468391B2 (en) 2003-12-17 2008-12-23 Allergan, Inc. Methods for treating retinoid responsive disorders using selective inhibitors of CYP26A and CYP26B
US20060030042A1 (en) 2003-12-19 2006-02-09 Ali Brivanlou Maintenance of embryonic stem cells by the GSK-3 inhibitor 6-bromoindirubin-3'-oxime
US7541185B2 (en) 2003-12-23 2009-06-02 Cythera, Inc. Methods for identifying factors for differentiating definitive endoderm
CN109628371B (zh) 2003-12-23 2021-02-19 维亚希特公司 定形内胚层
EA200601231A1 (ru) 2003-12-23 2007-06-29 Китера, Инк. Дефинитивная энтодерма
TWI334443B (en) 2003-12-31 2010-12-11 Ind Tech Res Inst Method of single cell culture of undifferentiated human embryonic stem cells
US7794704B2 (en) 2004-01-23 2010-09-14 Advanced Cell Technology, Inc. Methods for producing enriched populations of human retinal pigment epithelium cells for treatment of retinal degeneration
US20080241107A1 (en) 2004-01-23 2008-10-02 Copland Iii John A Methods and Compositions For Preparing Pancreatic Insulin Secreting Cells
WO2005080551A2 (en) 2004-02-12 2005-09-01 University Of Newcastle Upon Tyne Stem cells
WO2005080598A1 (ja) 2004-02-19 2005-09-01 Dainippon Sumitomo Pharma Co., Ltd. 体細胞核初期化物質のスクリーニング方法
WO2005086860A2 (en) 2004-03-09 2005-09-22 Gang Xu Methods for generating insulin-producing cells
WO2005086845A2 (en) 2004-03-10 2005-09-22 Regents Of The University Of California Compositions and methods for growth of embryonic stem cells
US7892835B2 (en) 2004-03-23 2011-02-22 Daiichi Sankyo Company, Limited Pluripotent stem cell growing method
WO2005097980A2 (en) * 2004-03-26 2005-10-20 Geron Corporation New protocols for making hepatocytes from embryonic stem cells
KR20070029681A (ko) 2004-04-01 2007-03-14 위스콘신 얼럼나이 리서어치 화운데이션 줄기 세포의 내배엽 및 이자 혈통으로의 분화
KR101278421B1 (ko) 2004-04-27 2013-07-15 비아싸이트, 인크. Pdx1 발현 내배엽
WO2006016999A1 (en) 2004-07-09 2006-02-16 Cythera, Inc. Methods for identifying factors for differentiating definitive endoderm
US20080268533A1 (en) 2004-08-25 2008-10-30 University Of Georgia Research Foundation, Inc. Methods and Compositions Utilizing Myc and Gsk3Beta to Manipulate the Pluripotency of Embryonic Stem Cells
DE102004043256B4 (de) 2004-09-07 2013-09-19 Rheinische Friedrich-Wilhelms-Universität Bonn Skalierbarer Prozess zur Kultivierung undifferenzierter Stammzellen in Suspension
ATE549395T1 (de) 2004-09-08 2012-03-15 Wisconsin Alumni Res Found Medium und kultur von embryonalen stammzellen
JP2008518585A (ja) 2004-09-08 2008-06-05 ウイスコンシン アラムニ リサーチ ファンデーション ヒト胚幹細胞の培養
EP1841857A1 (en) 2004-12-30 2007-10-10 Stemlifeline, Inc. Methods and compositions relating to embryonic stem cell lines
JP2008538276A (ja) 2005-01-28 2008-10-23 ノヴァセラ・リミテッド 胚幹細胞培養のための方法
CA2613812A1 (en) 2005-01-31 2006-08-10 Es Cell International Pte Ltd. Directed differentiation of embryonic stem cells and uses thereof
US20060182724A1 (en) 2005-02-15 2006-08-17 Riordan Neil H Method for expansion of stem cells
SG160373A1 (en) 2005-03-04 2010-04-29 John Oaeneil Adult pancreatic derived stromal cells
GB0505970D0 (en) 2005-03-23 2005-04-27 Univ Edinburgh Culture medium containing kinase inhibitor, and uses thereof
CN100425694C (zh) 2005-04-15 2008-10-15 北京大学 诱导胚胎干细胞向胰腺细胞分化的方法
US7998938B2 (en) 2005-04-15 2011-08-16 Geron Corporation Cancer treatment by combined inhibition of proteasome and telomerase activities
US20080208351A1 (en) 2005-04-26 2008-08-28 Aarhus Universitet Biocompatible Material for Surgical Implants and Cell Guiding Tissue Culture Surfaces
WO2006126574A1 (ja) 2005-05-24 2006-11-30 Kumamoto University Es細胞の分化誘導方法
AU2006202209B2 (en) 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
CA2610598A1 (en) 2005-06-10 2006-12-21 Irm Llc Compounds that maintain pluripotency of embryonic stem cells
WO2006138433A2 (en) 2005-06-14 2006-12-28 The Regents Of The University Of California Induction of cell differentiation by class i bhlh polypeptides
EP1931764A1 (en) 2005-06-21 2008-06-18 GE Healthcare Bio-Sciences AB Method for cell culture
KR20130100221A (ko) 2005-06-22 2013-09-09 제론 코포레이션 인간 배아 줄기 세포의 현탁 배양
JP5345388B2 (ja) 2005-06-30 2013-11-20 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ 環式アニリノ−ピリジノトリアジン
WO2007016485A2 (en) 2005-07-29 2007-02-08 Athersys, Inc. Use of a gsk-3 inhibitor to maintain potency of cultured cells
US20090087907A1 (en) 2005-07-29 2009-04-02 Alice Pebay Compositions and Methods for Growth of Pluripotent Cells
US20080194024A1 (en) * 2005-07-29 2008-08-14 Mays Robert W Culture of Non-Embryonic Cells at High Cell Density
WO2007025234A2 (en) 2005-08-26 2007-03-01 The Trustees Of Columbia University In The City Of New York Generation of pancreatic endocrine cells from primary duct cell cultures and methods of use for treatment of diabetes
US8476070B2 (en) 2005-08-29 2013-07-02 Technion Research & Development Foundation Limited Media for culturing stem cells
AU2006285468A1 (en) 2005-09-02 2007-03-08 Agency For Science, Technology And Research Method of deriving progenitor cell line
US9165427B2 (en) 2005-09-08 2015-10-20 Bally Gaming, Inc. Wagering game with multiple bonus triggering feature and bonus accrual feature
SG151259A1 (en) 2005-09-12 2009-04-30 Es Cell Int Pte Ltd Cardiomyocyte production
SG169324A1 (en) 2005-10-14 2011-03-30 Univ Minnesota Differentiation of non-embryonic stem cells to cells having a pancreatic phenotype
US7732202B2 (en) 2005-10-21 2010-06-08 International Stem Cell Corporation Oxygen tension for the parthenogenic activation of human oocytes for the production of human embryonic stem cells
DK1957636T3 (en) 2005-10-27 2018-10-01 Viacyte Inc PDX1-EXPRESSING DORSAL AND VENTRAL FORTARM ENDODERM
ES2367525T3 (es) 2005-12-13 2011-11-04 Kyoto University Factor de reprogramación celular.
WO2007082963A1 (es) 2006-01-18 2007-07-26 Fundación Instituto Valenciano De Infertilidad Líneas de células madre embrionarias humanas y métodos para usar las mismas
EP1994141B1 (en) 2006-02-23 2017-11-15 ViaCyte, Inc. Compositions and methods useful for culturing differentiable cells
EP1999253B1 (en) 2006-03-02 2019-05-22 Viacyte, Inc. Endocrine precursor cells, pancreatic hormone-expressing cells and methods of production
US7695965B2 (en) 2006-03-02 2010-04-13 Cythera, Inc. Methods of producing pancreatic hormones
GB0615327D0 (en) 2006-03-30 2006-09-13 Univ Edinburgh Culture medium containing kinase inhibitors and uses thereof
EP2021462B1 (en) 2006-04-28 2019-01-09 Lifescan, Inc. Differentiation of human embryonic stem cells
US8741643B2 (en) * 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
US8685730B2 (en) 2006-05-02 2014-04-01 Wisconsin Alumni Research Foundation Methods and devices for differentiating pluripotent stem cells into cells of the pancreatic lineage
WO2007136673A2 (en) 2006-05-19 2007-11-29 Medistem Laboratories, Inc. Treatment of disc degenerative disease and compositions for same
WO2007139929A2 (en) 2006-05-25 2007-12-06 The Burnham Institute For Medical Research Methods for culture and production of single cell populations of human embryonic stem cells
CN101541953A (zh) 2006-06-02 2009-09-23 佐治亚大学研究基金会 通过从人胚胎干细胞获得的定形内胚层细胞的分化得到胰和肝内胚层细胞及组织
US20090298169A1 (en) 2006-06-02 2009-12-03 The University Of Georgia Research Foundation Pancreatic and Liver Endoderm Cells and Tissue by Differentiation of Definitive Endoderm Cells Obtained from Human Embryonic Stems
WO2007149182A2 (en) 2006-06-19 2007-12-27 Geron Corporation Differentiation and enrichment of islet-like cells from human pluripotent stem cells
CN100494359C (zh) 2006-06-23 2009-06-03 中日友好医院 神经干细胞三维立体培养体外扩增的方法
US20080003676A1 (en) 2006-06-26 2008-01-03 Millipore Corporation Growth of embryonic stem cells
EP2046946B8 (en) 2006-06-26 2017-01-25 Lifescan, Inc. Pluripotent stem cell culture
WO2008004990A2 (en) 2006-07-06 2008-01-10 Es Cell International Pte Ltd Method for stem cell culture and cells derived therefrom
AU2007277364B2 (en) 2006-07-26 2010-08-12 Viacyte, Inc. Methods of producing pancreatic hormones
US9040297B2 (en) 2006-08-02 2015-05-26 Technion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
KR101331510B1 (ko) 2006-08-30 2013-11-20 재단법인서울대학교산학협력재단 저농도의 포도당을 함유하는 인간 배아줄기세포용 배지조성물 및 이를 이용한 인간 배아 줄기세포로부터 인슐린생산 세포 또는 세포괴로 분화시키는 방법, 그리고그로부터 유도된 인슐린 생산 세포 또는 세포괴
JP2008099662A (ja) 2006-09-22 2008-05-01 Institute Of Physical & Chemical Research 幹細胞の培養方法
US20080091234A1 (en) 2006-09-26 2008-04-17 Kladakis Stephanie M Method for modifying a medical implant surface for promoting tissue growth
EP2114895A4 (en) 2006-10-17 2011-06-22 Stiefel Laboratories TALARAZOLMETABOLITEN
WO2008048647A1 (en) * 2006-10-17 2008-04-24 Cythera, Inc. Modulation of the phosphatidylinositol-3-kinase pathway in the differentiation of human embryonic stem cells
CA2666789C (en) 2006-10-18 2016-11-22 Yong Zhao Embryonic-like stem cells derived from adult human peripheral blood and methods of use
WO2008056779A1 (fr) 2006-11-09 2008-05-15 Japan As Represented By The President Of International Medical Center Of Japan Procédé destiné à la culture et au passage d'une cellule souche embryonnaire de primate, et procédé destiné à induire la différenciation de la cellule souche embryonnaire
TW200836749A (en) 2007-01-09 2008-09-16 Vioquest Pharmaceuticals Inc Compositions including triciribine and bortezomib and derivatives thereof and methods of use thereof
EP2126045A4 (en) * 2007-01-30 2010-05-26 Univ Georgia EARLY MESODERM CELLS, STABLE POPULATION OF MESENDODERM CELLS WITH THE ABILITY TO GENERATE ENDODERM AND MESODERM CELL LINES AND MULTIPOTENTIAL MIGRATION CELLS
GB0703188D0 (en) 2007-02-19 2007-03-28 Roger Land Building Large scale production of stem cells
WO2008148105A1 (en) 2007-05-25 2008-12-04 Medistem Laboratories, Inc. Endometrial stem cells and methods of making and using same
US7354310B1 (en) * 2007-06-07 2008-04-08 Fci Americas Technology, Inc. Electrical connector housing cover
EP2173863B1 (en) 2007-06-29 2018-10-10 FUJIFILM Cellular Dynamics, Inc. Automated method and apparatus for embryonic stem cell culture
KR101555824B1 (ko) 2007-07-18 2015-09-25 라이프스캔, 인코포레이티드 인간 배아 줄기 세포의 분화
MX2010001335A (es) 2007-07-31 2010-06-02 Lifescan Inc Diferenciacion de celulas madre embrionarias humanas.
CN102317443B (zh) 2007-07-31 2018-09-14 生命扫描有限公司 用人饲养细胞进行的多能干细胞分化
EP2190413B1 (en) 2007-08-24 2015-01-28 Stichting Het Nederlands Kanker Instituut Compositions for the treatment of neoplastic diseases
US20110151447A1 (en) 2007-11-06 2011-06-23 Children's Medical Center Corporation Method to produce induced pluripotent stem (ips) cells from non-embryonic human cells
KR101592182B1 (ko) 2007-11-27 2016-02-05 라이프스캔, 인코포레이티드 인간 배아 줄기 세포의 분화
SG154367A1 (en) 2008-01-31 2009-08-28 Es Cell Int Pte Ltd Method of differentiating stem cells
WO2009096049A1 (ja) 2008-02-01 2009-08-06 Kyoto University 人工多能性幹細胞由来分化細胞
US20100330677A1 (en) 2008-02-11 2010-12-30 Cambridge Enterprise Limited Improved Reprogramming of Mammalian Cells, and Cells Obtained
KR101597731B1 (ko) 2008-02-21 2016-02-26 센토코 오르토 바이오테크 인코포레이티드 세포 부착, 배양 및 탈리를 위한 방법, 표면 개질 플레이트 및 조성물
JPWO2009110215A1 (ja) 2008-03-03 2011-07-14 独立行政法人科学技術振興機構 繊毛細胞の分化誘導方法
US8716018B2 (en) 2008-03-17 2014-05-06 Agency For Science, Technology And Research Microcarriers for stem cell culture
RU2359030C1 (ru) 2008-03-19 2009-06-20 Общество С Ограниченной Ответственностью "Лаборатория Клеточных Технологий" Способ получения эндотелиальных клеток из эмбриональных стволовых клеток человека (варианты)
US8338170B2 (en) 2008-04-21 2012-12-25 Viacyte, Inc. Methods for purifying endoderm and pancreatic endoderm cells derived from human embryonic stem cells
WO2009131568A1 (en) 2008-04-21 2009-10-29 Cythera, Inc. Methods for purifying endoderm and pancreatic endoderm cells derived from human embryonic stem cells
WO2009132083A2 (en) 2008-04-22 2009-10-29 President And Fellows Of Harvard College Compositions and methods for promoting the generation of pdx1+ pancreatic cells
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US7939322B2 (en) 2008-04-24 2011-05-10 Centocor Ortho Biotech Inc. Cells expressing pluripotency markers and expressing markers characteristic of the definitive endoderm
WO2009154606A1 (en) 2008-06-03 2009-12-23 Cythera, Inc. Growth factors for production of definitive endoderm
KR20180018839A (ko) 2008-06-30 2018-02-21 얀센 바이오테크 인코포레이티드 만능 줄기 세포의 분화
DE102008032236A1 (de) 2008-06-30 2010-04-01 Eberhard-Karls-Universität Tübingen Isolierung und/oder Identifizierung von Stammzellen mit adipozytärem, chondrozytärem und pankreatischem Differenzierungspotential
US20100028307A1 (en) 2008-07-31 2010-02-04 O'neil John J Pluripotent stem cell differentiation
US9683215B2 (en) 2008-08-22 2017-06-20 President And Fellows Of Harvard College Methods of reprogramming cells
KR101798474B1 (ko) 2008-10-31 2017-11-16 얀센 바이오테크 인코포레이티드 인간 배아 줄기 세포의 췌장 내분비 계통으로의 분화
ES2634445T3 (es) 2008-10-31 2017-09-27 Janssen Biotech, Inc. Diferenciación de las células madre embrionarias humanas con el linaje endocrino pancreático
DK2356213T3 (da) 2008-11-04 2019-09-09 Viacyte Inc Stamcelleaggregatsuspensionssammensætninger og fremgangsmåder til differentiering deraf
US8008075B2 (en) 2008-11-04 2011-08-30 Viacyte, Inc. Stem cell aggregate suspension compositions and methods of differentiation thereof
JP2012508584A (ja) 2008-11-14 2012-04-12 ヴィアサイト,インコーポレイテッド ヒト多能性幹細胞由来膵臓細胞のカプセル化
CN102307991B (zh) 2008-11-20 2017-08-15 森托科尔奥索生物科技公司 微载体上的多能干细胞培养
DK2356218T3 (en) 2008-12-05 2017-08-21 Inserm (Institut Nat De La Santé Et De La Rech Médicale) METHOD AND MEDIUM FOR NEURAL DIFFERENTIZATION OF PLURIPOTENT CELLS
GB2485113B (en) 2009-07-20 2016-12-28 Janssen Biotech Inc Differentiation of human embryonic stem cells into cells of the pancreatic endoderm lineage
US10076544B2 (en) 2009-07-20 2018-09-18 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
AR078805A1 (es) 2009-10-29 2011-12-07 Centocor Ortho Biotech Inc Celulas madre pluripotentes
FI20096288A0 (fi) 2009-12-04 2009-12-04 Kristiina Rajala Formulations and methods for culturing stem cells
RU2701335C2 (ru) 2009-12-23 2019-09-25 Янссен Байотек, Инк. Способ получения популяции панкреатических эндокринных клеток, соэкспрессирующих nkx6.1 и инсулин, и способ лечения диабета
ES2633648T3 (es) 2009-12-23 2017-09-22 Janssen Biotech, Inc. Diferenciación de células madre embrionarias humanas
CA2794473A1 (en) 2010-02-03 2011-08-11 National Cancer Center Induced hepatic stem cell and process for production thereof, and applications of the cell
US20120322152A1 (en) 2010-03-02 2012-12-20 Michael Raghunath Culture Additives To Boost Stem Cell Proliferation And Differentiation Response
CN102959076B (zh) 2010-03-31 2015-09-16 斯克里普斯研究所 重编程细胞
US9234170B2 (en) 2010-04-25 2016-01-12 Mount Sinai School Of Medicine Generation of anterior foregut endoderm from pluripotent cells
BR112012028855A2 (pt) 2010-05-12 2015-09-22 Janssen Biotech Inc diferenciação das células-tronco embrionárias humanas
WO2012019122A2 (en) 2010-08-05 2012-02-09 Wisconsin Alumni Research Foundation Simplified basic media for human pluripotent cell culture
MX348537B (es) 2010-08-31 2017-06-07 Janssen Biotech Inc Diferencia de celulas madre pluripotentes.
US9528090B2 (en) 2010-08-31 2016-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
MY177150A (en) 2011-02-28 2020-09-08 Stempeutics Res Malaysia Sdn Bhd Isolation and expansion of adult stem cells, their therapeutic composition and uses thereof
WO2013055834A2 (en) 2011-10-11 2013-04-18 The New York Stem Cell Foundation Er stress relievers in beta cell protection
US9670463B2 (en) 2011-10-14 2017-06-06 Children's Medical Center Corporation Inhibition and enhancement of reprogramming by chromatin modifying enzymes
BR112014015417A8 (pt) 2011-12-22 2017-07-04 Janssen Biotech Inc diferenciação de células-tronco embrionárias humanas em células positivas de insulina hormonal únicas
US10519422B2 (en) 2012-02-29 2019-12-31 Riken Method of producing human retinal pigment epithelial cells
CN108103005A (zh) 2012-06-08 2018-06-01 詹森生物科技公司 人胚胎干细胞向胰腺内分泌细胞的分化
JP6470687B2 (ja) 2012-09-03 2019-02-13 ノヴォ ノルディスク アー/エス 小分子を用いた多能性幹細胞からの膵臓内胚葉の作製
CA2896658C (en) 2012-12-31 2021-06-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
US8859286B2 (en) 2013-03-14 2014-10-14 Viacyte, Inc. In vitro differentiation of pluripotent stem cells to pancreatic endoderm cells (PEC) and endocrine cells
EP2970892A1 (en) 2013-03-15 2016-01-20 The Jackson Laboratory Isolation of non-embryonic stem cells and uses thereof
AU2014248167B2 (en) 2013-04-03 2019-10-10 FUJIFILM Cellular Dynamics, Inc. Methods and compositions for culturing endoderm progenitor cells in suspension
TWI768808B (zh) 2021-04-01 2022-06-21 友達光電股份有限公司 遮光元件基板以及顯示裝置

Patent Citations (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4557264A (en) * 1984-04-09 1985-12-10 Ethicon Inc. Surgical filament from polypropylene blended with polyethylene
US6022743A (en) * 1986-04-18 2000-02-08 Advanced Tissue Sciences, Inc. Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
US5567612A (en) * 1986-11-20 1996-10-22 Massachusetts Institute Of Technology Genitourinary cell-matrix structure for implantation into a human and a method of making
US5759830A (en) * 1986-11-20 1998-06-02 Massachusetts Institute Of Technology Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5770417A (en) * 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US6331298B1 (en) * 1992-03-28 2001-12-18 Renovo Limited Wound healing and treatment of fibrotic disorders
US5834308A (en) * 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6200806B1 (en) * 1995-01-20 2001-03-13 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6800480B1 (en) * 1997-10-23 2004-10-05 Geron Corporation Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
US6509369B2 (en) * 1997-12-29 2003-01-21 Ortho-Mcneil Pharmaceutical, Inc. Anti-inflammatory compounds
US20040132729A1 (en) * 1998-05-11 2004-07-08 Francesco Salituro Inhibitors of p38
US6793945B2 (en) * 1999-01-08 2004-09-21 Sky High, Llc Aqueous anti-apoptotic compositions
US6458593B1 (en) * 1999-01-21 2002-10-01 Vitro Diagnostics, Inc. Immortalized cell lines and methods of making the same
US6365149B2 (en) * 1999-06-30 2002-04-02 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6333029B1 (en) * 1999-06-30 2001-12-25 Ethicon, Inc. Porous tissue scaffoldings for the repair of regeneration of tissue
US6534084B1 (en) * 1999-06-30 2003-03-18 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6306424B1 (en) * 1999-06-30 2001-10-23 Ethicon, Inc. Foam composite for the repair or regeneration of tissue
US20030082155A1 (en) * 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20040241761A1 (en) * 1999-12-13 2004-12-02 Nora Sarvetnick Markers for identification and isolation of pancreatic islet alpha and beta cell progenitors
US20020072117A1 (en) * 2000-01-11 2002-06-13 Chunhui Xu Human feeder cells that support proliferation of undifferentiated pluripotent stem cells
US6642048B2 (en) * 2000-01-11 2003-11-04 Geron Corporation Conditioned media for propagating human pluripotent stem cells
US20050148070A1 (en) * 2000-03-09 2005-07-07 Thomson James A. Cultivation of primate embryonic stem cells
US20050244962A1 (en) * 2000-03-09 2005-11-03 Thomson James A Cultivation of primate embryonic stem cells
US6458589B1 (en) * 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
US20040209901A1 (en) * 2000-10-23 2004-10-21 Smithkline Beecham Corporation Novel compounds
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
US6656488B2 (en) * 2001-04-11 2003-12-02 Ethicon Endo-Surgery, Inc. Bioabsorbable bag containing bioabsorbable materials of different bioabsorption rates for tissue engineering
US6626950B2 (en) * 2001-06-28 2003-09-30 Ethicon, Inc. Composite scaffold with post anchor for the repair and regeneration of tissue
US20040171623A1 (en) * 2001-07-19 2004-09-02 Reynolds Rachel Heulwen Chemical compounds
US20050037488A1 (en) * 2001-08-06 2005-02-17 Maisam Mitalipova Alternative compositions and methods for the culture of stem cells
US20060040387A1 (en) * 2001-12-07 2006-02-23 Fisk Gregory J Islet cells from human embryonic stem cells
US20030138948A1 (en) * 2001-12-07 2003-07-24 Fisk Gregory J. Islet cells from human embryonic stem cells
US20060003446A1 (en) * 2002-05-17 2006-01-05 Gordon Keller Mesoderm and definitive endoderm cell populations
US20040015805A1 (en) * 2002-07-22 2004-01-22 Kidd David A. Layout quality analyzer
US20040062753A1 (en) * 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
US20040220393A1 (en) * 2002-11-23 2004-11-04 Ward Donna T. Modulation of HIF1alpha and HIF2alpha expression
US20060148081A1 (en) * 2003-12-23 2006-07-06 Olivia Kelly Expansion of definitive endoderm cells
US20050233446A1 (en) * 2003-12-31 2005-10-20 Parsons Xuejun H Defined media for stem cell culture
US20070010011A1 (en) * 2003-12-31 2007-01-11 The Burnham Institute Defined media for pluripotent stem cell culture
US20050266554A1 (en) * 2004-04-27 2005-12-01 D Amour Kevin A PDX1 expressing endoderm
US8187878B2 (en) * 2004-08-13 2012-05-29 University Of Georgia Research Foundation, Inc. Methods for increasing definitive endoderm differentiation of pluripotent human embryonic stem cells with PI-3 kinase inhibitors
US20070259423A1 (en) * 2006-05-02 2007-11-08 Jon Odorico Method of differentiating stem cells into cells of the endoderm and pancreatic lineage
US20090298178A1 (en) * 2008-06-03 2009-12-03 D Amour Kevin Allen Growth factors for production of definitive endoderm

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Rebbapragada et al., Mol Cell Biol, 2003, 23: 7230-7242. *
Ring et al., Diabetes, 2003, 52: 588-595. *
Tsuchida et al., Mol. Cell. Endocrinol., 2004, 220: 59-65. *

Cited By (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8778673B2 (en) 2004-12-17 2014-07-15 Lifescan, Inc. Seeding cells on porous supports
US9074189B2 (en) 2005-06-08 2015-07-07 Janssen Biotech, Inc. Cellular therapy for ocular degeneration
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
US9725699B2 (en) 2006-04-28 2017-08-08 Lifescan, Inc. Differentiation of human embryonic stem cells
US20070254359A1 (en) * 2006-04-28 2007-11-01 Lifescan, Inc. Differentiation of human embryonic stem cells
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US10316293B2 (en) 2007-07-01 2019-06-11 Janssen Biotech, Inc. Methods for producing single pluripotent stem cells and differentiation thereof
US9096832B2 (en) 2007-07-31 2015-08-04 Lifescan, Inc. Differentiation of human embryonic stem cells
US9744195B2 (en) 2007-07-31 2017-08-29 Lifescan, Inc. Differentiation of human embryonic stem cells
US10456424B2 (en) 2007-07-31 2019-10-29 Janssen Biotech, Inc. Pancreatic endocrine cells and methods thereof
US9969982B2 (en) 2007-11-27 2018-05-15 Lifescan, Inc. Differentiation of human embryonic stem cells
US9062290B2 (en) 2007-11-27 2015-06-23 Lifescan, Inc. Differentiation of human embryonic stem cells
US11001802B2 (en) 2008-02-21 2021-05-11 Nunc A/S Surface of a vessel with polystyrene, nitrogen, oxygen and a static sessile contact angle for attachment and cultivation of cells
US10066203B2 (en) 2008-02-21 2018-09-04 Janssen Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US20090325293A1 (en) * 2008-04-24 2009-12-31 Janet Davis Treatment of pluripotent cells
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US9845460B2 (en) 2008-04-24 2017-12-19 Janssen Biotech, Inc. Treatment of pluripotent cells
US10351820B2 (en) 2008-06-30 2019-07-16 Janssen Biotech, Inc. Methods for making definitive endoderm using at least GDF-8
US10233421B2 (en) 2008-06-30 2019-03-19 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9593306B2 (en) 2008-06-30 2017-03-14 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US20110091971A1 (en) * 2008-06-30 2011-04-21 Janet Davis Differentiation of Pluripotent Stem Cells
US9593305B2 (en) 2008-06-30 2017-03-14 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9234178B2 (en) 2008-10-31 2016-01-12 Janssen Biotech, Inc. Differentiation of human pluripotent stem cells
US9752126B2 (en) 2008-10-31 2017-09-05 Janssen Biotech, Inc. Differentiation of human pluripotent stem cells
US20100112693A1 (en) * 2008-10-31 2010-05-06 Alireza Rezania Differentiation of Human Embryonic Stem Cells
US9012218B2 (en) 2008-10-31 2015-04-21 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9388387B2 (en) 2008-10-31 2016-07-12 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9969972B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Pluripotent stem cell culture on micro-carriers
US9969973B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
US8785184B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10076544B2 (en) 2009-07-20 2018-09-18 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US8785185B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10471104B2 (en) 2009-07-20 2019-11-12 Janssen Biotech, Inc. Lowering blood glucose
US9133439B2 (en) 2009-12-23 2015-09-15 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9150833B2 (en) 2009-12-23 2015-10-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10704025B2 (en) 2009-12-23 2020-07-07 Janssen Biotech, Inc. Use of noggin, an ALK5 inhibitor and a protein kinase c activator to produce endocrine cells
US20110151560A1 (en) * 2009-12-23 2011-06-23 Jean Xu Differentiation of human embryonic stem cells
US20110151561A1 (en) * 2009-12-23 2011-06-23 Janet Davis Differentiation of human embryonic stem cells
US9593310B2 (en) 2009-12-23 2017-03-14 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9969981B2 (en) 2010-03-01 2018-05-15 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
US10329534B2 (en) 2010-03-01 2019-06-25 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
US9752125B2 (en) 2010-05-12 2017-09-05 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
PH12018500672A1 (en) * 2010-05-12 2019-03-11 Janssen Biotech Inc Differentation of human embryonic stem cells
US9181528B2 (en) 2010-08-31 2015-11-10 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US20180237752A1 (en) * 2010-08-31 2018-08-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US20170009212A1 (en) * 2010-08-31 2017-01-12 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9528090B2 (en) 2010-08-31 2016-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9506036B2 (en) 2010-08-31 2016-11-29 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9458430B2 (en) 2010-08-31 2016-10-04 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
CN108517310A (zh) * 2010-08-31 2018-09-11 詹森生物科技公司 人胚胎干细胞的分化
CN103154239A (zh) * 2010-08-31 2013-06-12 詹森生物科技公司 人胚胎干细胞的分化
WO2012030538A3 (en) * 2010-08-31 2012-06-07 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
AU2011296381B2 (en) * 2010-08-31 2016-03-31 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9951314B2 (en) * 2010-08-31 2018-04-24 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
WO2012030539A3 (en) * 2010-08-31 2012-05-31 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
CN103154239B (zh) * 2010-08-31 2018-05-15 詹森生物科技公司 人胚胎干细胞的分化
CN108517310B (zh) * 2010-08-31 2022-02-15 詹森生物科技公司 人胚胎干细胞的分化
US11377640B2 (en) 2011-12-22 2022-07-05 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US10358628B2 (en) 2011-12-22 2019-07-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US9434920B2 (en) 2012-03-07 2016-09-06 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US9593307B2 (en) 2012-03-07 2017-03-14 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US20140162359A1 (en) * 2012-05-07 2014-06-12 Janssen Biotech, Inc. Differentiation of Human Embryonic Stem Cells into Pancreatic Endoderm
WO2013169769A1 (en) 2012-05-07 2013-11-14 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endoderm
EP3957714A1 (en) 2012-06-08 2022-02-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
US10066210B2 (en) 2012-06-08 2018-09-04 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
EP3450542A1 (en) 2012-06-08 2019-03-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endodrine cells
EP2859091A1 (en) 2012-06-08 2015-04-15 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
EP3450543A1 (en) 2012-06-08 2019-03-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endodrine cells
WO2013184888A1 (en) 2012-06-08 2013-12-12 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
US10208288B2 (en) 2012-06-08 2019-02-19 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
EP3450544A1 (en) 2012-06-08 2019-03-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endodrine cells
EP4039798A1 (en) 2012-12-31 2022-08-10 Janssen Biotech, Inc. Suspension and clustering of human pluripotent cells
US10138465B2 (en) 2012-12-31 2018-11-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using HB9 regulators
WO2014105543A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
WO2014106141A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Suspension and clustering of human pluripotent cells for differentiation into pancreatic endocrine cells
US10377989B2 (en) 2012-12-31 2019-08-13 Janssen Biotech, Inc. Methods for suspension cultures of human pluripotent stem cells
WO2014105546A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
EP4219683A1 (en) 2012-12-31 2023-08-02 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
US10344264B2 (en) 2012-12-31 2019-07-09 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
US10947511B2 (en) 2012-12-31 2021-03-16 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using thyroid hormone and/or alk5, an inhibitor of tgf-beta type 1 receptor
US10030229B2 (en) 2013-06-11 2018-07-24 President And Fellows Of Harvard College SC-β cells and compositions and methods for generating the same
US11078463B2 (en) 2013-06-11 2021-08-03 President And Fellows Of Harvard College SC-beta cells and compositions and methods for generating the same
US11827905B2 (en) 2013-06-11 2023-11-28 President And Fellows Of Harvard College SC-beta cells and compositions and methods for generating the same
US10655106B2 (en) 2013-06-11 2020-05-19 President And Fellows Of Harvard College SC-beta cells and compositions and methods for generating the same
US11162078B2 (en) 2013-06-11 2021-11-02 President And Fellows Of Harvard College SC-beta cells and compositions and methods for generating the same
US11104883B2 (en) 2013-06-11 2021-08-31 President And Fellows Of Harvard College SC-beta cells and compositions and methods for generating the same
US20160237401A1 (en) * 2013-10-09 2016-08-18 Cambridge Enterprise Limited In Vitro Production of Foregut Stem Cells
US10000740B2 (en) * 2013-10-09 2018-06-19 Cambridge Enterprise Limited In vitro production of foregut stem cells
US20170280121A1 (en) * 2014-04-01 2017-09-28 Canon Kabushiki Kaisha Imaging apparatus and image processing system
WO2015175307A1 (en) 2014-05-16 2015-11-19 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
US10006006B2 (en) 2014-05-16 2018-06-26 Janssen Biotech, Inc. Use of small molecules to enhance MAFA expression in pancreatic endocrine cells
EP3954759A1 (en) 2014-05-16 2022-02-16 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
US10870832B2 (en) 2014-05-16 2020-12-22 Janssen Biotech, Inc. Use of small molecules to enhance MAFA expression in pancreatic endocrine cells
US11085026B2 (en) 2014-12-18 2021-08-10 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived beta cells and uses thereof
US10190096B2 (en) 2014-12-18 2019-01-29 President And Fellows Of Harvard College Methods for generating stem cell-derived β cells and uses thereof
US10253298B2 (en) 2014-12-18 2019-04-09 President And Fellows Of Harvard College Methods for generating stem cell-derived beta cells and methods of use thereof
US11085025B2 (en) 2014-12-18 2021-08-10 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived beta cells and uses thereof
US10443042B2 (en) 2014-12-18 2019-10-15 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived beta cells and uses thereof
US11155787B2 (en) 2014-12-18 2021-10-26 President And Fellows Of Harvard College Methods for generating stem cell-derived beta cells and methods of use thereof
US10927350B2 (en) 2014-12-18 2021-02-23 President And Fellows Of Harvard College Methods for generating stem cell-derived beta cells and uses thereof
US11085027B2 (en) 2014-12-18 2021-08-10 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived beta cells and uses thereof
US20160215268A1 (en) * 2014-12-19 2016-07-28 Janssen Biotech, Inc. Suspension Culturing of Pluripotent Stem Cells
WO2016100035A1 (en) 2014-12-19 2016-06-23 Janssen Biotech, Inc. Suspension culturing of pluripotent stem cells
WO2017180361A1 (en) 2016-04-14 2017-10-19 Janssen Biotech, Inc. Differentiation of pluripotent stem cells to intestinal midgut endoderm cells
US10420803B2 (en) 2016-04-14 2019-09-24 Janssen Biotech, Inc. Differentiation of pluripotent stem cells to intestinal midgut endoderm cells
WO2017222879A1 (en) 2016-06-21 2017-12-28 Janssen Biotech, Inc. Generation of human pluripotent stem cell derived functional beta cells showing a glucose-dependent mitochondrial respiration and two-phase insulin secretion response
EP4194548A1 (en) 2016-06-21 2023-06-14 Janssen Biotech, Inc. Generation of human pluripotent stem cell derived functional beta cells showing a glucose-dependent mitochondrial respiration and two-phase insulin secretion response
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11945795B2 (en) 2017-11-15 2024-04-02 Vertex Pharmaceuticals Incorporated Islet cell manufacturing compositions and methods of use
US11466256B2 (en) 2018-08-10 2022-10-11 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
US11525120B2 (en) 2018-08-10 2022-12-13 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation

Also Published As

Publication number Publication date
ZA201100758B (en) 2016-07-27
WO2010002846A1 (en) 2010-01-07
US20120190112A1 (en) 2012-07-26
US10351820B2 (en) 2019-07-16
US20120190111A1 (en) 2012-07-26
RU2533805C2 (ru) 2014-11-20
US9593306B2 (en) 2017-03-14
US20170183625A1 (en) 2017-06-29
HK1156665A1 (en) 2012-06-15
KR20160101215A (ko) 2016-08-24
KR20170052706A (ko) 2017-05-12
JP5734183B2 (ja) 2015-06-17
ES2552240T3 (es) 2015-11-26
CN102159703A (zh) 2011-08-17
EP2310492A1 (en) 2011-04-20
US20170183624A1 (en) 2017-06-29
EP2942392A1 (en) 2015-11-11
ES2697798T3 (es) 2019-01-28
MX2011000125A (es) 2011-02-25
US10233421B2 (en) 2019-03-19
PL2310492T3 (pl) 2015-12-31
KR20110029155A (ko) 2011-03-22
KR101829310B1 (ko) 2018-02-14
KR20180018839A (ko) 2018-02-21
US9593305B2 (en) 2017-03-14
EP2310492B1 (en) 2015-07-22
KR101734501B1 (ko) 2017-05-11
RU2011103197A (ru) 2012-08-10
CA2729121A1 (en) 2010-01-07
EP2942392B1 (en) 2018-10-03
CA2729121C (en) 2019-04-09
AU2009267137A1 (en) 2010-01-07
PL2942392T3 (pl) 2019-02-28
KR101651661B1 (ko) 2016-08-26
US20120196365A1 (en) 2012-08-02
BRPI0914116A2 (pt) 2019-09-17
RU2673750C2 (ru) 2018-11-29
CN102159703B (zh) 2015-11-25
HK1216905A1 (zh) 2016-12-09
RU2014123686A (ru) 2015-12-20
JP2011526786A (ja) 2011-10-20

Similar Documents

Publication Publication Date Title
US10351820B2 (en) Methods for making definitive endoderm using at least GDF-8
US11369642B2 (en) Methods for lowering blood glucose
AU2016202260B2 (en) Differentiation of human embryonic stem cells
KR102025158B1 (ko) 인간 배아 줄기 세포의 췌장 내분비 계통으로의 분화
US8785184B2 (en) Differentiation of human embryonic stem cells
AU2018201993B2 (en) Differentiation of pluripotent stem cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: CENTOCOR ORTHO BIOTECH INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DAVIS, JANET;LIU, JIAJIAN;PARMENTER, CHRISTINE;AND OTHERS;SIGNING DATES FROM 20100212 TO 20100327;REEL/FRAME:025342/0085

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: JANSSEN BIOTECH, INC., PENNSYLVANIA

Free format text: CHANGE OF NAME;ASSIGNOR:CENTOCOR ORTHO BIOTECH INC.;REEL/FRAME:041332/0881

Effective date: 20110622