AU2018201993A1 - Differentiation of pluripotent stem cells - Google Patents

Differentiation of pluripotent stem cells Download PDF

Info

Publication number
AU2018201993A1
AU2018201993A1 AU2018201993A AU2018201993A AU2018201993A1 AU 2018201993 A1 AU2018201993 A1 AU 2018201993A1 AU 2018201993 A AU2018201993 A AU 2018201993A AU 2018201993 A AU2018201993 A AU 2018201993A AU 2018201993 A1 AU2018201993 A1 AU 2018201993A1
Authority
AU
Australia
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.)
Granted
Application number
AU2018201993A
Other versions
AU2018201993B2 (en
Inventor
Pascal Ghislain Andre Bonnet
Janet Davis
Jiajian Liu
Christine Parmenter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Biotech Inc
Original Assignee
Janssen Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2009/049183 external-priority patent/WO2010002846A1/en
Application filed by Janssen Biotech Inc filed Critical Janssen Biotech Inc
Priority to AU2018201993A priority Critical patent/AU2018201993B2/en
Publication of AU2018201993A1 publication Critical patent/AU2018201993A1/en
Application granted granted Critical
Publication of AU2018201993B2 publication Critical patent/AU2018201993B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

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

Abstract

The present invention is directed to methods to differentiate pluripotent stem cells. In particular, 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.

Description

DIFFERENTIATION OF PLURIPOTENT STEM CELLS
The present application is a divisional application of Australian Application No. 2015261644, which is incorporated in its entirety herein by reference.
The present invention claims priority to application serial number 61/076,900, fded June 30, 2008, application serial number 61/076,908, fded June 30, 2008, and application serial number 61/076,915, fded June 30, 2008.
FIELD OF THE INVENTION
The present invention is directed to methods to differentiate pluripotent stem cells. In particular, 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.
BACKGROUND
Advances in cell-replacement therapy for Type I diabetes mellitus and a shortage of transplantable islets of Langerhans have focused interest on developing sources of insulin-producing cells, or β cells, appropriate for engraftment. One approach is the generation of functional β cells from pluripotent stem cells, such as, for example, embryonic stem cells.
In vertebrate embryonic development, 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.
Formation of the pancreas arises from the differentiation of definitive endoderm into pancreatic endoderm. Cells of the pancreatic endoderm express the pancreatic-duodenal homeobox gene, PDX1. In the absence of PDX1, the pancreas fails to develop beyond the formation of ventral and dorsal buds. Thus, 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.
Cells bearing the features of islet cells have reportedly been derived from embryonic cells of the mouse. For example, Liimelsky etal. ( Science 292:1389, 2001) report differentiation of mouse embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Soria et at. (Diabetes 49:157, 2000) report that insulin-secreting cells derived from mouse embryonic stern cells normalize glycemia in streptozotocin-induced diabetic mice.
In one example, Hori etal. (PNAS 99: 16105, 2002) discloses that treatment of mouse embryonic stem cells with inhibitors of phosphoinositide 3-kinase (LY294002) produced cells that resembled β cells.
In another example, Blyszczuk et al. (PNAS 100:998, 2003) reports the generation of insulin-producing cells from mouse embryonic stem cells constitutively expressing Pax4.
Micallcf et al. reports that retinoic acid can regulate the commitment of embryonic stem cells to form Pdxl positive pancreatic endoderm. Retinoic acid is most effective at inducing Pdxl 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 etal. reports a mouse embryonic stem cell line over-expressing Pdxl. Their results show that exogenous Pdxl expression clearly enhanced the expression of insulin, somatostatin, glucokinase, ncurogenin3, p48, Pax6, and HNF6 genes in the resulting differentiated cells (Diabetes 53: 1030, 2004).
Skoudy et al. reports that activin A (a member of the TGF-β superfamily) up-regulates the expression of exocrine pancreatic genes (p48 and amylase) and endocrine genes (Pdxl, insulin, and glucagon) in mouse embryonic stem cells.
The maximal effect was observed using 1 nM activin A. They also observed that the expression level of insulin and Pdxl mRNA was not affected by retinoic acid: however, 3 nM FGF7 treatment resulted in an increased level of the transcript for Pdxl (Biochem. J. 379: 749, 2004).
Shiraki et al. studied the effects of growth factors that specifically enhance differentiation of embryonic stem cells into Pdxl positive cells. They observed that TGFP2 rcproducibly yielded a higher proportion of Pdxl positive cells (Genes Cells. 2005 June; 10(6): 503-16).
Gordon et al. demonstrated the induction of brachyury [positive]/ FlNF-3bcta [positive] endoderm cells from mouse embryonic stem cells in the absence of scrum and in the presence of activin along with an inhibitor of Wnt signaling (US 2006/0003446A 1).
Gordon et al. (PNAS, Vol 103, page 16806, 2006) states: "Wnt and TGF-beta/ nodal/ activin signaling simultaneously were required for the generation of the anterior primitive streak."
However, the mouse model of embryonic stem cell development may not exactly mimic the developmental program in higher mammals, such as, for example, humans.
Thomson et al. isolated embryonic stem cells from human blastocysts (Science 282:! 14, 1998). Concurrently, Gearhart and coworkers derived human embryonic germ (hEG) cel! lines from fetal gonadal tissue (Shamblott et al.. Proc. Natl. Acad. Sci. USA 95:13726, 1998). Unlike mouse embryonic stem cells, which can be prevented from differentiating simply by culturing with Leukemia Inhibitory Factor (L1F), human embryonic stem cells must be maintained under very special conditions (U.S. Pat. No. 6,200,806; WO 99/20741; WO 01/51616). 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 scrum (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'Amouret al. (Nature Biotechnology--24, 1392-1401 (2006)) 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 cn route to cells that express endocrine hormones."
In another example, Fisk et al. reports a system for producing pancreatic islet cells from human embryonic stem cells (US2006/0040387A1). In this case, 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-β antagonists such as Noggin in combination with EGF or bctacellulin to generate PDX1 positive cells. The terminal differentiation was induced by nicotinamide.
In one example, Bcnvcnistry etal. 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. For example. 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-β 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).
In another example, Arab K. Y. et al states: "Activins are multifunctional growth factors belonging to the transforming growth factor-β 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).
Therefore, there still remains a significant need for alternatives for activin A to facilitate the differentiation of pluripotcnt stem cells.
SUMMARY
In one embodiment, the present invention provides a method to differentiate pluripotcnt 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.
In one embodiment, the medium comprising a sufficient amount of GDF-8 also contains at least one other compound. In one embodiment, the at least one other compound is an aniline-pyridinotriazine. In an alternate embodiment, the at least one other compound is a cyclic aniline-pyridinotriazine.
BRIEF DESCRIPTION OF THE FIGURES
Figure 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 SOX 17 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).
Figure 2 shows the dose response relationship of activin A and GDF8 used to differentiate cells of the human embryonic stem cell line ΗI 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 20ng/ml Wnt3a on the first day of assay. Differentiation was determined by measuring SOX 17 intensity using a fluorescent antibody probe and high content analysis on a GE Healthcare ΓΝ Cell Analyzer.
Figure 3 shows the expression of CXCR4 in cells following the first step of differentiation, according to the methods described in Example 12. HI cells were treated with lOOng/m! activin A or 200ng/ml GDF-8 for a total of three days in combination with 20ng/ml Wtit3a for the first day or 2.5μΝ1 Compound 34 or 2.5μΜ 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.
Figure 4 shows the expression of SOX17 in cells after three days differentiation to definitive endoderm according to the methods described in Example 12. HI cells were treated for a total of three days with lOOng/mi activin A or 200ng/ml GDF-8 in combination with 20ng/ml Wnt3a for the first day or 2.5μΜ Compound 34 or 2.5μΜ Compound 56 for all three days. Differentiation was determined by measuring SOX 17 intensity (black bars) and resulting cell number (white bars) with fluorescent antibody probes and high content analysis on a GE Healthcare IN Cell Analyzer.
Figure 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. HI cells were treated for a total of three days with lOOng/ml activin A or 200ng/ml GDF-8 in combination with 20ng/ml Wnt3a for the first day or 2.5μΜ Compound 34 or 2.5uM Compound 56 for all three days followed by subsequent differentiation through the second and third 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.
Figure 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. Η I ceils were treated fora total of three days with lOOng/ml activin A or 200ng/ml GDF-8 in combination with 20ng/ml Wnt3a for the first day or 2.5μΜ Compound 34 or 2.5μΜ 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.
Figure 7 shows the protein expression for insulin and glucagon and cell number in cells differentiated according to the methods described in Example 12. HI cells were treated for a total of three days with lOOng/ml activin A or 200ng/ml GDF-8 in combination with 20ng/ml Wnt3a for the first day or 2.5μΜ Compound 34 or 2.5μΜ 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.
Figure 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. HI cells were treated for a total of four days with I OOng/ml of activin A or ! OOng/ml of a GDF- growth factor in combination with 20ng/m! Wnt3a for the first day or 2.5μΜ Compound 34 or 2.5μΜ Compound 56 for the first two days of assay. SOX17 protein expression (black bars) and cell numbers (white bars), 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. Panel 8A shows a series of control conditions for differentiation in the absence of any growth factors (ΜΟΝΕ), 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.
Figure 9 shows SOX 17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 14, HI cells were treated fora total of three days with lOOng/ml of activin A or various growth factors at the concentrations shown in combination with 20ng/m! Wnt3a or 2.5μΜ Compound 34 for the first day of assay. SOX 17 protein expression (black bars) and cell numbers (white bars), as determined with fluorescent antibody probes and high content analysis, arc depicted for each treatment group. For comparative purposes, values arc 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). Pane! 9B shows differentiation w'ith GDF-8 (Vendor PeproTech), at the concentrations shown, in combination with 20ng/ml Wnt3a. Panel 9C shows differentiation with GDF-8 (Vendor Shenendoah), at the concentrations shown, in combination with 20ng/ml Wnt3a. Panel 9D shows differentiation with TGFpi, 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 w ith 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 Wnt3a or Compound 34.
Figure 10 shows SOX 17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15. ΗI cells were treated for a total of three days in various timed exposures with lOOng/ml of activin A or lOOng/ml GDF-8 in combination with 20ng/ml Wnt3a. SOX 17 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 w ith 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.
Figure 11 shows SOX 17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a total of three days in various timed exposures with lOOng/ml of activin A in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19(Pancl C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), orGSK3 inhibitor BIO (Panel G)) at the concentrations shown, where test compound was added only on the first day of assay. Protein expression for SOX 17, as determined with fluorescent antibody probes and high content analysis, is depicted by total intensity values.
Figure 12 shows SOX 17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15. HI ceils were treated for a total of three days in various timed exposures with lOOng/m! of activin A in combination with test compound (Compound 181 (Panel A), Compound 180 (Panel B), Compound 19(Pancl C), Compound 202 (Panel D), Compound 40 (Panel E), Compound 34 (Panel F), or GSK.3 inhibitor BIO (Panel G)) al the concentrations shown, where test compound was added for all three days of assay. Protein expression for SOX17, as determined with fluorescent antibody probes and high content analysis, is depicted by total intensity values.
Figure 13 shows SOX17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15. Η1 cells were treated for a total of three days in various timed exposures with lOOng/ml ofGDF-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, as determined with fluorescent antibody probes and high content analysis, is depicted by total intensity values.
Figure 14 shows SOX 17 protein expression in human embryonic stem cells after differentiation to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a total of three days in various timed exposures with iOOng/ml ofGDF-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 SOX 17, as determined with fluorescent antibody probes and high content analysis, is depicted by total intensity values.
Figure 15 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a total of three days in various timed exposures with lOOng/ml of activin A or lOOng/ml GDF-8 in combination with 20ng/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.
Figure 16 shows cel! number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a totai of three days in various timed exposures with lOOng/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 (Pane! 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.
Figure 17 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a total of three days in various timed exposures with 1 OOng/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.
Figure 18 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a total of three days in various timed exposures with lOOng/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, arc shown.
Figure 19 shows cell number yields after differentiation of human embryonic stem cells to definitive endoderm, according to the methods described in Example 15. HI cells were treated for a total of three days in various timed exposures with lOOng/ml of GDF-8 in combination with test compound (Compound 181 (Panel A), Compound 180 (Pane! B), Compound !9 (Pane! 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.
Figure 20 shows the expression of various protein markers in cells throughout multiple steps of differentiation according to the methods described in Example 16. HI cells were treated with IQOng/ml activin A or lOOng/ml GDF-8 for a total of three days in combination with 20ng/ml Wnt3a for the first day or 2.5μΜ various compounds (Compound 19, Compound 202, Compound 40, or GSK3 inhibitor BIO) added only on the first day. Figure 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. Figure 20, panel B shows high content image analysis for normalized SOX 17 protein expression (black bars) and recovered cell numbers (white bars) resulting from the first step of differentiation, testing the corresponding treatments shown. Figure 20, panel C shows high content image analysis for relative cell numbers recovered from cultures treated through differentiation step 5. Figure 20, panel D shows high content image analysis for glucagon protein expression from cultures treated through differentiation step 5. Figure 20, panel E shows high content image analysis for insulin protein expression from cultures treated through differentiation step 5. Figure 20, panel F shows the ratio of glucagon to insulin expression in cells from cultures treated through differentiation step 5. For comparison purposes, expression values in panels B, C, D, E, and F are normalized to the control treatment with activin A and Wnt3a during step 1.
Figure 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. HI cells were treated with lOOng/ml activin A or lOOng/ml GDF-8 fora total of three days in combination with 20ng/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. In subsequent panels of Figure 21, 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). At the conclusion of 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.
Figure 22 shows the expression of various protein and RT-PCR markers in cells treated according to the methods described in Example 18. HI cells were treated with lOOng/ml activin A or lOOng/ml GDF-8 for a total of three days in combination with 20ng/ml Wnt3a for the first day or 2.5μΜ Compound 40 or 2.5μΜ Compound 202 only on the first day. Figure 22, panel A shows FACS analysis for the definitive endodertn 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. In Figure 22, 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.
Figure 23 shows the level of C-peptide detected in SCLD-beige mice that received cells at the end of step four of the differentiation protocol as described in Example 18.
Figure 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-pcptidc 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.
Figure 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 lOOng/ml activin A with 20ng/ml Wnt3a (AA/Wnt3a) or with various treatments combining GDF-8 as shown: 50ng/ml GDF-8with 2.5μΜ Compound 34 (Cmp 34+8); or 50ng/ml GDF-8 with 2.5μΜ Compound 34 and 50ng/ml PDGF (Cmp 34+8+D); or 50ng/ml GDF-8 with 2.5μΜ Compound 34 and 50ng/ml PDGF and 50ng/ml VEGF (Cmp 34+8+D+V); or 50ng/ml GDF-8 with 2.5μΜ Compound 34 and 50ng/ml PDGF and 50ng/ml VEGF and 20ng/ml muscimol (Cmp 34+8+D+V+M).
Figure 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, 2days, and 3 days after initiating the differentiation assay.
Figure 27 shows the expression of various proteins and genes from cells grown on mierocarrier 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.
DETAILED DESCRIPTION
For clarity of disclosure, and not by way of limitation, the detailed description of the invention is divided into the following subsections that describe or illustrate certain features, embodiments, or applications of the present invention.
Definitions
Stem cells are undifferentiated cells defined by their ability at the single cell level to both seif-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) pluripotcnt, 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 (c.g., platelets) that arc 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).
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. As used herein, 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. "β-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, ISLI, HNF-3 beta, MAFA, PAX4, or PAX6. Cells expressing markers characteristic of the β cell lineage include β cells. “Cells expressing markers characteristic of the definitive endoderm lineage”, or “Stage 1 cells”, or “Stage 1”, as used herein, refers to cells expressing at least one of the following markers: SOX 17, 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. Ceils expressing markers characteristic of the definitive endodcrm lineage include primitive streak precursor cells, primitive streak cells, mesendoderm cells and definitive endodcrm cells. “Cells expressing markers characteristic of the pancreatic endodcrm lineage”, as used herein, 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 endodcrm cells, primitive gut tube cells, and posterior foregut cells. “Cells expressing markers characteristic of the pancreatic endocrine lineage”, or “Stage 5 cells”, or “Stage 5”, as used herein, refers to cells expressing at least one of the following markers: NGN3, NEUROD, ISL i, PDX!, NK.X6.1, 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 β-ccll lineage. “Definitive endodcrm”, as used herein, refers to cells which bear the characteristics of cells arising from the cpiblast during gastrulation and which form the gastrointestinal tract and its derivatives. Definitive endoderm cells express the following markers: F1NF-3 beta, GATA4, SOX-17, Cerberus, OTX2, goosecoid, C-K.it, CD99, or MIXL1. “Extraembryonic endoderm”, as used herein, refers to a population of cells expressing at least one of the following markers: SOX7, AFP, or SPARC. "Markers", as used herein, are nucleic acid or polypeptide molecules that are differentially expressed in a cell of interest. In this context, differential expression means an increased level for a positive marker and a decreased level for a negative marker. The detectable level of the marker nucleic acid or polypeptide is 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”, as used herein, refers to a cell expressing at least one of the following markers: C'D48, eomesodermin (EOMES), SOX 17, DK.K4, HNF-3 beta, GSC, FGF17, or GATA-6. “Pancreatic endocrine cell”, or “pancreatic hormone expressing cell”, as used herein, refers to a cell capable of expressing at least one of the following hormones: insulin, glucagon, somatostatin, and pancreatic polypeptide. “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, 1SL1, PDX1, PAX4, or NKX2.2. “Pancreatic hormone producing cell”, as used herein, refers to a cell capable of producing at least one of the following hormones: insulin, glucagon, somatostatin, and pancreatic polypeptide. “Pancreatic hormone secreting cell”, as used herein, 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: PDX I, HNF 1, PTF-1 alpha, HNF6, HB-9, or PROX-1. “Pre-primitive streak cell”, as used herein, refers to a cell expressing at least one of the following markers: Nodal, or FGF8. “Primitive 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: HNF 1, HNF-4 alpha. “Primitive streak cell”, as used herein, refers to a cell expressing at least one of the following markers: Brachyury, Mix-like homeobox protein, or FGF4.
Isolation, expansion, and culture of pluripotent stem cells
Characterization of pluripotent stem cells
The 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. Alternatively, 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 piimate 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.
Sources of pluripotent stem cells
The types of pluripotent stem ceils that may be used include established lines of pluripotent cells derived from tissue formed after gestation, including prc-cmbryonic 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 HI, H7, and H9 (WiCell). Also contemplated is use of the compositions of this disclosure during the initial establishment or stabilization of such cells, in which case the source cells would be primary pluripotent cells taken directly from the source tissues. Also suitable are cells taken from a pluripotent stem cell population already cultured in the absence of feeder cells. Also suitable arc mutant human embryonic stem cell lines, such as, for example, BGOlv (BresaGen, Athens, GA).
In one embodiment, human embryonic stem cells are prepared as described by Thomson et al. (U.S. Pat. No. 5,843,780; Science 282:1145, 1998: Curr. Top. Dev. Biol. 38:133 ff„ 1998; Proc. Natl. Acad. Sci. U.S.A. 92:7844, 1995).
In one embodiment, pluripotent stem cells arc prepared as described by Takahashi et al. (Cell 131: 1-12,2007).
Culture of pluripotent stem cells
In one embodiment, pluripotent stem cells are typically cultured on a layer of feeder cells that support the pluripotent stem cells in various ways. Alternatively, pluripotent stem cells arc 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. Alternatively, 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. In one embodiment, 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. In one embodiment, the suitable culture substrate is MATRIGEL8 (Becton Dickenson). MATRIGEL* is a soluble preparation from Engelbreth-Holm-Swarm tumor cells that gels at room temperature to form a reconstituted basement membrane.
Other extracellular matrix components and component mixtures are suitable as an alternative. Depending on the cel! type being proliferated, this may include laminin, fibroncctin, 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, Dulbccco's modified Eagle's medium (DMEM), Gibco # 11965-092;
Knockout Dulbccco's modified Eagle's medium (K.0 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; β- mercaptoethanol, Sigma # M7522; human recombinant basic fibroblast growth factor (bFGF), Gibco # 13256-029.
Formation of pancreatic hormone producing cells from pluripotent stem cells
In one embodiment, the present invention provides a method for producing pancreatic hormone producing cells from pluripotent stem cells, comprising the steps of: a. Culturing pluripotent stem cells, b. Differentiating the pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage, c. Differentiating the cells expressing markers characteristic of the definitive endoderm lineage into cells expressing markers characteristic of the pancreatic endoderm lineage, and d. Differentiating the cells expressing markers characteristic of the pancreatic endoderm lineage into ceils expressing markers characteristic of the pancreatic endocrine lineage.
In one aspect of the present invention, the pancreatic endocrine cell is a pancreatic hormone producing cell. In an alternate aspect, the pancreatic endocrine cell is a cell expressing markers characteristic of the β-cell lineage. A cell expressing markers characteristic of the β-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. In one aspect of the present invention, a cell expressing markers characteristic of the β-cell lineage is a β-cell.
Pluripotent stem cells suitable for use in the present invention include, for example, the human embryonic stem cell line H9 (N1H code: WA09), the human embryonic stem cell line HI (NIH code: WA01), the human embryonic stem cell line H7 (N1H code: WA07), and the human embryonic stem cell line SA002 (Cellartis, Sweden). Also suitable for use in the present invention arc cells that express at least one of the following markers characteristic of pluripotent cells: ABCG2, cripto, CD9, FOXD3, Conne.\in43, Connexin45, OCT4, SOX2, Nanog, hTERT, UTF-1, ZFP42, SSEA-3, SSEA-4, Tral-60, orTral-BJ.
The pluripotent stem cells may be cultured on a feeder cell layer. Alternatively, 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 Bioscienccs under the trade name MATRIGEL™). Alternatively, the extracellular matrix may be growth factor-reduced MATRIGEL™. Alternatively, the extracellular matrix may be Sbronectin. In an alternate embodiment, 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. Examples of suitable methods for diluting the extracellular matrix and for coating the tissue culture substrate may be found in Kleinman, H.K., et a/., Biochemistry 25:312 (1986), and Hadley, M.A., et al„ J.Cell.Biol. 101:1511 (1985).
In otic embodiment, the extracellular matrix is MATRIGELim. In one embodiment, the tissue culture substrate is coated with MATRIGEL™ at a 1:10 dilution. In an alternate embodiment, the tissue culture substrate is coated with MATRIGEL.1 M at a 1:15 dilution. In an alternate embodiment, the tissue culture substrate is coated with MATRIGEL™ at a 1:30 dilution. In an alternate embodiment, the tissue culture substrate is coated with MATRIGEL™ at a 1:60 dilution.
In one embodiment, the extracellular matrix is growth factor-reduced MATRIGELIM. In one embodiment, the tissue culture substrate is coated with growth factor-reduced MATRIGELIM at a 1:10 dilution. In an alternate embodiment, the tissue culture substrate is coated with growth factor-reduced MATRIGELIM at a 1:15 dilution. In an alternate embodiment, the tissue culture substrate is coated with growth factor-reduced MATRJGELim at a 1:30 dilution. In an alternate embodiment, the tissue culture substrate is coated with growth factor-reduced MATRIGELIM at a 1:60 dilution.
Markers characteristic of the definitive endoderm lineage are selected from the group consisting of SOX 17, GATA4, HNF-3 beta, GSC, CERE Nodal, FGF8, Brachyury,
Mix-like homeobox protein, FGF4 CD48, oomcsodermin (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. In one aspect of the present invention, a cell expressing markers characteristic of the definitive endoderm lineage is a primitive streak precursor cell.
In an alternate aspect, a cell expressing markers characteristic of the definitive endoderm lineage is a mesendoderm cell. In an alternate aspect, a cell expressing markers characteristic of the definitive endoderm lineage is a definitive endoderm cell.
Markers characteristic of the pancreatic endoderm lineage are selected from the group consisting of PDX1, HNF-I 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. In one aspect of the present invention, a cell expressing markers characteristic of the pancreatic endoderm lineage is a pancreatic endoderm cell.
Markers characteristic of the pancreatic endocrine lineage are selected from the group consisting of NGN3, NEUROD, ISL1, PDX1, NKX6.1, PAX4, and PTF-1 alpha. In one embodiment, a pancreatic endocrine cell is capable of expressing at least one of the following hormones; insulin, glucagon, somatostatin, and pancreatic polypeptide. Suitable for use in the present invention is a cell that expresses at least one of the markers characteristic of the pancreatic endocrine lineage. In one aspect of the present invention, a cell expressing markers characteristic of the pancreatic endocrine lineage is a pancreatic endocrine cell. The pancreatic endocrine cell may be a pancreatic hormone expressing cell. Alternatively, the pancreatic endocrine cell may be a pancreatic hormone secreting cell.
Formation of cells expressing markers characteristic of the definitive endoderm lineage
In one aspect of the present invention, 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. Alternatively, 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. Alternatively, 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. Alternatively, 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. Alternatively, 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. Alternatively, 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. Alternatively, the pluripotent stem cells may be cultured in the medium containing a sufficient amount of GDF-8 for about one day.
In one embodiment, 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.
In one embodiment, the medium comprising a sufficient amount of GDF-8 also contains at least one other factor. In one embodiment, 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.
In one embodiment, 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.
In one embodiment, 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.
In one embodiment, 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.
In one embodiment, 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.
In one embodiment, 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.
In one embodiment, 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.
In one embodiment, 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.
In one embodiment, the muscimol is used at a concentration from about 1 μΜ to about 200 μΜ. In an alternate embodiment, the muscimol is used at a concentration from about 1 μΜ to about 20 μΜ. In an alternate embodiment, the muscimol is used at a concentration of about 20 μΜ.
In one embodiment, the PD98059 is used at a concentration from about 0.1 μΜ to about 10 μΜ. In an alternate embodiment, the PD98059 is used at a concentration from about 0.1 μΜ to about 1 μΜ. in asi alternate embodiment, the PD98059 is used at a concentration of about I μΜ.
In one embodiment, the LY294002 is used at a concentration from about 0.25 μΜ to about 25 μΜ. In an alternate embodiment, the LY294002 is used at a concentration from about 0.25 μΜ to about 2.5 μΜ. In an alternate embodiment, the LY294002 is used at a concentration of about 2.5 μΜ.
In one embodiment, the U0124 is used at a concentration from about 0.1 μΜ to about 10 μΜ. In an alternate embodiment, the U0124 is used at a concentration from about 0.1 μΜ to about 1 μΜ. In an alternate embodiment, the U0124 is used at a concentration of about 1 μΜ.
In one embodiment, the U0126 is used at a concentration from about 0.1 μΜ to about 10 μΜ. In an alternate embodiment, the U0126 is used at a concentration from about 0.1 μΜ to about 1 μΜ. In an alternate embodiment, the U0I26 is used at a concentration of about 1 μΜ.
In one embodiment, the sodium butyrate is used at a concentration from about 0.05 μΜ to about 5 μΜ. In an alternate embodiment, the sodium butyrate is used at a concentration from about 0.05 μΜ to about 0.5 μΜ. In an alternate embodiment, the sodium butyrate is used at a concentration of about 0.5 μΜ.
In an alternate embodiment, the at least one other factor is selected from the group consisting of: an aniline-pyridinotriazine, a cyclic aniline-pyridinotriazine, N-> [ 1 -(Phcnylmethyl)azepan-4-yl]methyl}-2-pyridin-3-ylacetamide, 4-{[4-(4-{[2-(Pyridin-2-yIamino)ethyl]amino}-l,3,5-triazin-2-yl)pyridin-2-yl]oxy}butan-l-ol, 3-({3-[4-({2-[Mcthyl(pyridin-2-yl )amino]ethy 1| amino)-1,3.5-triazin-2-yl]pyridin-2- yl }amino)propan-1 -ol, N~4'-[2-(3-Fluorophenyl)ethyl]-N~2--[3-(4-rncthylpiperazin- 1- yl)propyl]pyrido[2,3-d]pyrimidinc-2,4-diaminc, l-Methyl-N-[(4-pyridin-3-yl-2-i[3-(trifluoromethy 1 )phenyl]amino} -1,3-thiazol-5-yl )methyl]piperidine-4-carboxamide, 1,1-Dimethylethyl {2-[4-({5-[3-(3-hydroxypiOpyl)phenyl]-4H-l,2,4-triazol-3-yl i amino)phenyl]ethyl}carbamate, 1,1-Dimethylethyl {[3-( {5-[5-(3-hydroxypropyl)- 2- imethyloxy)phenyl]-l,3-oxazol-2-yI}amino)phenyl]methyl{carbamate, l-({5-[6-({4-[(4-Methylpipcrazin-l-yl)sulfonyl]phenyl | amino )pyrazin-2-yl]thiophen-2-yl} methyl )piperidin-4-ol, 1 -({4-[6-( {4-[(4-Methylpiperazin-1 -yl)sulfonyl]phenyl |amino)pyrazin-2-yl]thiophen-2-yl| methyl )piperidine-4-carboxamide, and 2-{[4-(l-Methylethyl)phenyl]aminoJ-N-(2-thiophen-2-yiethyl)-7,8-dihydropyrido[4,3-d]pyrimidine-6(5H)-carboxamide.
The compounds of the present invention
The present invention provides compounds that are capable of differentiating pluripotent stem cells into cells expressing markers characteristic of the definitive endoderm lineage. in one embodiment, 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):
Formula (1).
The N-oxide forms, the pharmaceutically acceptable addition salts and the stereochemicalIy isomeric forms thereof, wherein: m represents an integer from 1 to 4; n represents an integer from ! to 4; Z represents N or C; R and R each independently represent hydrogen, Het , cyano, halo, hydroxy, Ci-6alkoxy-, Ci^alkyl-, mono-or di(C|_ialky I )amino-carbony I-, mono-or di(Ci_ ^alkyl)amino-sulfonyl, C|.(,alkoxy-substitutcd with halo or R1 rcprcscntsCi^alkyI substituted with one or where possible two or more substituents selected from hydroxy or halo; RJ and RJ each independently represents hydrogen, Ci_*alfcyl, C^alkenyl, Het3, Het^-Ci^alkyl-, Hets-Cualkylcarbonyl-, mono-or di(Cj^alkyl)amino-C|_4alkyl-carbonyl-or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or CMalkyloxy-; R? and R each independently represent hydrogen, Chalky!, Hetf', Het7-C'i-ialkyl-, C2. 4alkenylcarbonyl-optionaIly substituted with Hets-CMalkylaminocarbonyl-, C2. ^alkcnylsulfonyl-, C|_ilkyloxyCnalkyl-or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or Ci-jalkyloxy-; R4, R5, R6 and R1" each independently represent hydrogen or Chalky I optionally substituted with hydroxy, Het9 or CMalkyloxy;
Het1 and Het" each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl. pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het1 and Her are optionally substituted with amino, hydroxy. Chalky!, hydroxy-C|_ialIcyl-, phenyl, phenyl-Ci^alkyl- CMalkyi-oxy-Ci-^alkyl-mono-or difCi^alkyl) amino-or amino-carbonyl-;
Het* and Hei0 each independently represent, heterocyclc selected from pyrrolidinyl or piperidinyl wherein said Het1 and Het0 are optionally substituted with one or where possible twO or more substituents selected from Ci^alkyl, C^cycloalkyl, hydroxy-C|, ^alkyl-, C|_ialkyloxyCMalkyl or polyhydroxy-CMalkyl-;
Hot9, Het and Het9 each independently represent a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or piperidinyl wherein said Het"1, Het7 and Het9 are optionally substituted with one or where possible two or more substituents selected from C^alkyl, C^cycloalkyl, hydroxy-Ci_ialkyl-, CMalkyloxyCi-ialky] or polyhydroxy-C ^alkyl-;
Het" represents a heterocyclc selected from morpholinyl, pyrrolidinyl, piperazinyl or pipendinyl wherein said Het5 is optionally substituted with one or where possible two or more substituents selected from Cwalkyl, C\.6cycloalkyl, hydroxy-Ci^alkyl-, C]_ 4alkyloxyC|_jalkyl or polyhydroxy-Ci_4alkyl-;
Het10, Het11 and Het1' each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het10, Het11 and Het1' are optionally substituted with amino, hydroxy, Ci^alkyl, hydroxy-Ci_ia]kyl-, phenyl, phenyl-C|. 4a 1 kyl-, Ci4alkyl-oxy-Ci4alkyl-, amino-carbonyl-or mono-or di(C|.4alky 1 )amino-;
Het12 represents a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyI, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het1' is optionally substituted with amino, hydroxy, Cwalkyl, hydroxy-Ci^alkyl-, phenyl, phenyl-Ci-ialkyi-, Cmalkyl-oxy-CMalkyi-; mono-or di(Ci-i4lkyl)amino-or amino-carbonyl-;
Hctu represents a heterocycle selected from morpholinyl; pyrrolidinyl; piperazinyl; imidazolyl; pyrrolyl; 2,3,4-triazapyrrolyl; 1,2,3-triazolyI; pyrazolyl; or piperidinyl wherein said Het14 is optionally substituted with one or where possible two or more substituents selected from CMalkyl, CN^cycloalkyl, hydroxy-CMalkyl-, Cj. 4alkyloxyC|_iaikyl or polyhydroxy-Ci4alkyl-; in particular Het14 represents a heterocycle selected from morpholinyl; pyrrolidinyl; pyrrolyl; 2,3,4-triazapyrrolyl; piperazinyl or piperidinyl wherein said Het14 is optionally substituted with one or where possible two or more substituents selected from C| 4alkyl, Cv(,cycloalkyl, hydroxy-Ci^alky!-, C|.4alkyloxyCi_.jalkyl or poiyhydroxy-CMalkyl-; more particular Het*4 represents a heterocycle selected from morpholinyl; pyrrolidinyl; piperazinyl or piperidinyl wherein said Het14 is optionally substituted with one or where possible two or more substituents selected from CMalkyl, C^cycloalkyl, hydroxy-CMalkyl-, CMalkyloxyC|.4alkyl or poiyhydroxy-CMalkyl-.
In one embodiment, the aniline-pyridinotriazinc is a compound of the Formula (1).
In one embodiment, the aniline-pyridinotriazinc is a compound of the Formula (2).
Formula (2): 3-{3-[(4-Pyridin-3- yl-l,3,5-triazin-2-yl)amino]phcnyl[propanoic acid. Referred to herein as “Compound 1”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (3).
Formula (3): 2-{3-[(4-Pyridin-3- yl-l,3,5-triazin-2-yl)amino]phenyl} ethanol. Referred to herein as “Compound 2”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (4).
Formula (4): 1,1 -Dimethylethyl {2-[3-({4-[2-(3-hydiOxyprop-l-yn-l-yl)pyridin-4-yl]-l,3,5-triazin-2-yl J amino )phenyl]ethyl {carbamate. Referred to herein as “Compound 3”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (5).
Formula (5): 1,1 -Dimethylethyl {4-[4-(4-{[3-(hydroxymethyI)phenyl]amino}-l,3,5-triazin-2-yl)pyridin-2-yljbutyl [carbamate. Referred to herein as “Compound 4”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (6).
Formula (6): 1,1 -Dimethylethyl {3-[! [5-(2-1 [3-bromo-5-(hydroxymethyl )phenyl]amino }pyrimidin-4-yl)-2-(methyloxy)phenyl]methyl} (methyl )amino]propyl J carbamate. Referred to herein as “Compound 5”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (7).
Formula (7): 4-([3-(3-Fluorophcnyl)-3H-[l,2,3]triazolo[4,5-d]pyrimidin-5-yl]amino}benzoic acid. Referred to herein as “Compound 6”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (8).
Formula (8): 2-Fluoro-5-[(3-phenyl-3H-[l,2,3]triazolo[4,5-d]pyrimidin-5-yl)amino]benzoic acid. Referred to herein as “Compound 7”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (9).
Formula (9): N-{[3-(5-{[3-(2-
Aminopyrimidin-4-y!)phcnyl]amino}-3H-[l,2,3]triazolo[4,5-d]pyrimidin-3-y I )pheny 1 ] m et hy 1} cycloprop ane car box amide. Referred to herein as “Compound 8”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (10).
Formula (10): 4-[( 1 -Cyclohcxyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino]-N-[3- (methyIoxy)propyl]bcnzenesuIfonamidc. Referred to herein as “Compound 9”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (11).
Formula (11): 4-Chloro-2-[(6-{[3- (chloromethyi)-4-mcthoxyphenyl]aminoJpyrirnidin-4-yl)arnino]phenol. Referred to herein as “Compound 10”.
In one embodiment, the aniline-pyridinotriazinc is a compound of the Formula (12).
Formula (12): 4-{[4-(4-Methyl-3,4-dihydroquinoxalin-l(2H)-yl)pyrimidin-2-yl]amino}-N-( l-mcthylpipcridin-4-yl)benzamide. Referred to herein as “Compound 11”.
In one embodiment, the aniline-pyridinotriazine is a compound of the Formula (13).
Formula (13): N-(2-Methoxy-4-| [(3-methoxypropyl)amino]methyl | phenyl )-4-( 1 H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-2-amine. Referred to herein as “Compound 12”.
In one embodiment, 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):
Formula (14).
The N-oxide forms, the pharmaceutically acceptable addition salts and the stereochemically isomeric forms thereof, wherein: m represents an integer from I to 4; n represents an integer from 1 to 4; Z represents Nor C; Y represcnts-NR2-C|.6alkyl-CO-NR·'-, -CMalkyl-NR9-CMalkyl-, Cusalkyl-CO-Het10-, -Hetll-CO-C|-6alkyl-, -Hetlz-Cl.6alkyl·, -CO-Hetl3-C|.6alkyl-, -CO-NRl0-CK)alkyl-, -Het'-Ci ealkyl-CO-NR’’-, or-Hct2-CO-NR('- wherein the-C|^alkyl-1 inker in-NR2-C'" 6alky I -CO-NR^or-Het'-Ci-r.al kyl-CO-NR3- is optionally substituted with one or where possible two or more substituents selected from hydroxy, methoxy, aminocarbonyl, halo, phenyl, indolyl, methylsulfide. thiol, hydroxyphenyl, cyanophenyl, amino and hydroxycarbonyl; X1 represents a direct bond, CMalkyl, CV^alkyloxy-, Ci_»a 1 ky 1-CO-, C2_»alkenyl, C2-^alkynyl, or CMalkyl-NR·-, wherein said Ci^alkyl or Cj-ialkcnyl is optionally substituted with one or where possible two or more halo substituents; X2 represents a direct bond, Ci^alkyl, CMalkyloxy-, CMalkyl-CO-, Ca^alkenyl, C2-^alkynyl, or CMalkyl-NR7-, wherein said CMalkyl or C2^alkenyl is optionally substituted with one or where possible two or more halo substituents; I Ο 14 R and R each independently represent hydrogen, Het , cyano, halo, hydroxy, Ci_ ealkoxy-, Cm alkyl-, mono-or di(CMalkyl)amino-carbonyl-, mono-or di(C|. 4alkyl)amino-sulfonyl, C|.6alkoxy-substitutcd with halo or R1 rcpresentsC'Malkyl substituted with one or where possible two or more substituents selected from hydroxy or halo; R" and R9 each independently represents hydrogen, Ci^alkyl, C^alkenyl, Her, Het")-Cl_4alkyl-, Hets-C|_ialkylcarbonyl-, mono-or difCi^alkyOamino-Ci^alkyl-carbonyl-or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino orCMalkyloxy-; R' and R7 each independently represent hydrogen, C|_4alkyl, Hct'\ Hct7-Ci-4alkyl-, C2-4alkenylcarbonyl-optionally substituted with Hcts-C|_4alkylaminocarbonyl-, C2-4aIkenylsulfonyl-, CnIkyloxyCMalkyl-or phenyl optionally substituted with one or where possible two or more substituents selected from hydrogen, hydroxy, amino or Ci_ialkyloxy-; R4, R5, R6 and Rhl each independently represent hydrogen or CMalkyl optionally substituted with hydroxy, Het9 or Ci_jalkyloxy;
Het1 and Hct2 each independently represent a hctcrocyclc selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidiny! wherein said Met' and Het2 are optionally substituted with amino, hydroxy, CMalkyl, hydroxy- C1.4 a 1 Icy I -, phenyl, phenyl-Ci_4alkyl-, C|.4alkyl-oxy-Ci. 4aikyl-mono-or di(C|_4alkyl) amino-or amino-carbonyl-;
Her and Het0 each independently represent, heterocycle selected from pyrrolidinyl or piperidinyl wherein said Het' and Het° are optionally substituted with one or where possible two or more substituents selected from C i^alkyl, C^cycloalkyl, hydroxy-Cfr 4alkyl-, Ci-ialkyloxyCi-jalkyl or polyhydroxy-CMalkyl-;
Het'1, Het7 and Hety each independently represent a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or piperidinyl wherein said Hef\ Het and Het9 are optionally substituted with one or where possible two or more substituents selected from Ci^alkyl, C^ocycloalkyl, hydroxy-C^alky!-, CMalkyloxyCi-4alkyl or polyhydroxy-C Malkyl-;
Het? represents a heterocycle selected from morpholinyl, pyrrolidinyl, piperazinyl or pipendinyl wherein said Het-' is optionally substituted with one or where possible two or more substituents selected from CMalkyl, C.^cycloalkyl, hydroxy-CMalkyl-, Ci_ jalky loxyC|_4alkyl or polyhydroxy-C’i_ialkyl-;
Het10, Het11 and Het14 each independently represent a heterocycle selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het10, Het11 and Het1’ are optionally substituted with amino, hydroxy, C.Malkyl, hydroxy-C 1.4aIkyI-, phenyl, phenyl-C|. 4alkyl-, CMalkyi-oxy-CMalkyl-, amino-carbonyl-or mono-or di(Ci_4a!kyI)amino-;
Het1’ represents a hcterocyclc selected from pyrrolidinyl, piperidinyl, piperazinyl, pyridinyl, pyrimidinyl, pyrazinyl, imidazolidinyl or pyrazolidinyl wherein said Het12 is optionally substituted with amino, hydroxy, C Malkyl, hydroxy-C Malkyl-, phenyl, phenyl-C|_4alkyl-, CMalkyl-oxy-CMalkyl-; mono-or di(CMalkyl)amino-or amino-carbonyl-;
Het11 represents a hcterocycle selected from morpholinyl; pyrrolidinyl; piperazinyl; imidazolyl; pyrroiyl; 2,3,4-triazapyrrolyl; 1,2,3-triazolyl; pyrazolyl; or piperidinyl wherein said Het14 is optionally substituted with one or where possible two or more substituents selected from Cualkyi, C^cycloalkyl, hydroxy-C Malkyl-, Cj. 4 alky loxyC Malkyl or polyhydroxy-CMalkyl-; in particular Het14 represents a hcterocyclc selected from morpholinyl; pyrrolidinyl; pyrroiyl; 2,3,4-triazapyrrolyl; piperazinyl or piperidinyl wherein said Het'4 is optionally substituted with one or where possible two or more substituents selected from CMalkyl, C^,cycloalkyl, hydroxy-C 14a 1 kyl-, C MalkyloxyC Malkyl or polyhydroxy-C Malkyl-; more particular Het14 represents a heterocycle selected from morpholinyl; pyrrolidinyl; piperazinyl or piperidinyl wherein said Het14 is optionally substituted with one or where possible two or more substituents selected from Ci^alkyl, C.MCycloalkyl, hydroxy-CMalkyl-, Ci.jalkyloxyCi^alkyl or polyhydroxy-C Malkyl-.
Compounds of Formula (7) are disclosed in W02007/003525, assigned to Janssen Pharmaceutica N.V.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (H).
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (15) .
Formula (15): 1,8,10,12,17,19,23,27,33-
Nonaazapentacyclo[25.2.2.1 ~3,7~. 1 -9,13-. 1 -14,18~]tetratriaconta- 3(34),4,6,9(33), 10.12,14(32),15,17-nonaen-24-onc. Referred to herein as “Compound 13”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (16) .
Formula (16): 10-Chloro-14-cthyl- 3,5,7,14,17,22,27-heptaazatetracycIof 19.3.1.1 -2,6-. 1 -8,12~]heptacosa-1(25),2(27),3,5,8(26),9,1 l,21,23-nonaen-16-one. Referred to herein as “Compound 14”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (17).
Formula (17): 14-Ethyl- 3,5,7,14,17,27-hexaazatctracyclo[ 19.3.1.1 -2,6-. 1 -8,12-]hcptacosa-1(25),2(27),3,5,8(26),9,1 l,21,23-nonaen-l6-one. Referred to herein as “Compound 15”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (18).
Formula (18): lO-Chloro-14- ethyl-3,5,7,14,17,27-hexaazatetracyclo[ 19.3.1.1-2,6-.1-8,12~]hcptacosa-1(25),2(27),3,5,8(26),9,1 l,21,23-nonaen-16-one. Referred to herein as “Compound 16”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (19).
Formula(19): 3,5,7,14,20,26,31 -
Hcptaazapcntacyclo[22.3.1.1 -2,6-. 1 '8,12-. 1 -14,18~]hcntriaconta- 1 (28),2(31 ),3,5,8(30),9,11,24,26-nonacn-l 9-onc. Referred to herein as “Compound 17”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (20).
Formula (20): (18S)- 3,5,7,14,20,26,30-Heptaazapentacyclo[22,3.1. i -2,6-.! -8,12-.0-14,18-]triaconta-1(28),2(30),3,5,8(29),9,1 l,24,26-nonaen-19-one. Referred to herein as “Compound 18”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (21).
Formula (21): 14-Methyl- 3,5,7,14,18,24,28-heptaazatctracyclo[20.3.1.1 -2,6-. 1 ~8,12~]octacosa-1(26),2(28),3,5,8(27),9,11,22i24-nonaen-17-one. Referred to herein as “Compound 19”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (22).
Formula (22): 14-Methyl- 3,5,7. 14, i 9,25,29-heptaazatetracyclo[21.3.1.1 -2,6-. 1 -8,12-]nonacosa-1(27),2(29),3,5,8(28),9,1 l,23,25-nonaen-18-one. Referred to herein as “Compound 20”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (23).
Formula (23): 14-Mcthyl- 3,5,7,14,18,22,29-heptaazatctracyclo[21.3.1.1 -2,6-. 1 -8,12~]nonacosa-1(27),2(29),3,5,8(28),9,1 l,23,25-nonaen-17-one. Referred to herein as “Compound 21”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (24) .
Formula (24): 1,8,10,12,16,22,30-Heptaazapentacyclo[22.2.2.1 -3,7-. 1 -9,13-.1-14,18~]hcntriaconta-3(31 ),4,6,9(30), 10,12,14(29), 15,17-nonaen-23-one. Refemed to herein as “Compound 22”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (25) .
Formula (25): 1.8.10.12.16.22.26.32-
Octaazapcntacyclo[24.2.2.1 -3,7-. 1 -9,13-. 1 -14,18-]tritriaconta- 3(33),4,6,9(32), 10,12,14(31), 15,17-nonaen-23-one. Referred to herein as “Compound 23”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (26).
Formula (26): 5-Chloro-17-fluoro- 1.8.10.12.22.26.32- heptaazapentacyclo[24.2.2.1 -3,7-.1-9,13-. 1 -14,18~]tritriaconta-3(33),4,6,9(32), 10,! 2,14(3 I), 15,17-nonacn-23-one. Referred to herein as “Compound 24”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (27).
Formula (27): 10-Chloro-14-cthyl- 22-fluoro-3,5 ,7, 14,17,27-hexaazatetracycio[ 19.3.1.1 -2,6-. 1 --8,12~]hcptacosa-1(25),2(27),3,5,8(26),9,11.21,23-nonacn-l6-onc. Referred to herein as “Compound 25”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (28).
Formula (28): 10-Chloro-25- fluoro-3,5,7,14,20,31 - hexaazapentacyclo[22.3.1.1-2,6-. 1 ~8,12—, 1—14,18-]hentriaconta-1(28),2(31),3,5,8(30),9,1 l,24,26-nonaen-19-onc. Referred to herein as “Compound 26”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (29).
Formula (29): 4-Chloro- 1,8,10,12.17,22,26,32- octaazapcntacyclo[24.2.2.1 ~3,7~. 1 ~9,13~. 1-1 4,18~]tritriaconta- 3(33 ),4,6,9(32), 10,12,14(31), 15,17-nonaen-23-one. Referred to herein as “Compound 27”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (30) .
Formula (30): 18-Methyl- 3,5,7,15.18,28-hexaazatetracyclo[20,3.1.1 -2,6-. 1 -8,12-]octacosa-1(26),2(28),3,5,8(27),9,1 l,22,24-nonaen-16-one. Referred to herein as “Compound 28”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (31) .
Formula (31): I8-Ethyl- 3,5,7,15,18,28-hcxaazatctracyclo[20.3.1.1 -2,6-. I -8,12-]octacosa-1(26),2(28),3,5,8(27),9,11,22,24-nonaen-16-onc. Referred to herein as "Compound 29”
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (32) .
Formula (32): 1,8,10,12,17.19,23,27,33-
Nonaazapentacyclo[25.2.2.1 -3,7-. 1 -9,13~. 1 -14,18-]tetratriaconta-3(34),4,6,9(33), 10,12,14(32), 15,17-nonaen-24-one. Referred to herein as "Compound 30”.
In one embodiment, the cyclic anilinc-pyridinotnazine is a compound of the Formula (33) .
Formula (33): 1,11,13,15,23,3 !-
Hexaazapcntacyclo[23.2.2.1 -5,9-. 1 - ! 0,! 4-.! -! 6,2(K]dotriaconla-5(32),6,8,10(31),11,13,16(30). 17,19-nonaen-24-one. Referred to herein as “Compound 31”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (34) .
Formula (34): 15-Ethyl- 13,14,15,16,18,19-hexahydro-! 13-6,2-(azeno )-7,11 -(metheno)-l ,3,5.15,18-bcnzopentaazacyclohenicosin-17(12FI)-onc. Referred to herein as “Compound 32”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (35) .
Formula (35): 20-Mcthyl- 3,5,7,15,20,30-hcxaazatctracyclo[22.3.1.1 -2,6'. 1 -8, l2-]triaconta-1(28),2(30),3,5,8(29),9,1 l,24,26-nonaen-16-one. Referred to herein as “Compound 33”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Fonnula (36) .
Fonnula (36): 5-C'hloro- 1,8,10,12,16,22,26,32- octaazapentacyclo[24.2.2.1 -3,7-. 1 -9,13-.1-14,18-]tritriaconta- 3(33),4,6,9(32), 10,12,14(3!), 15,17-nonacti-23-onc. Refened to herein as “Compound 34”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (37) .
Formula (37): 10-C'hloro-14- ethyI-3,5,7,14,17,23.27-hcptaazatctracyclo[ 19.3.1.1 -2,6-. 1 -8,12~]heptacosa-1 (25),2(27),3,5,8(26),9,11,21,23-nonacn-16-onc. Referred to herein as “Compound 35”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (38) .
Formula (38): (18S)-10-Chloro- 3,5,7,14,20,26,30-heptaazapentacyclo[22.3.1.1 -2,6-.1-8,12-.0-14,18~Jtriaconta-1(28),2(30),3,5,8(29),9,1 l,24,26-nonaen-19-one. Referred to herein as “Compound 36”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (39) .
Formula (39): 10-ChIoro- 3,5,7,14,20,26,31 -hcptaazapcntacyclo[22.3.1.1 ~2,6~. 1 -8,12~. I -14,18-]hcntriaconta-1(28),2(31),3,5,8(30),9,11,24,26-nonaen-19-one. Referred to herein as “Compound 37”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (40) .
Formula (40): 5-Chloro- 1,8,10,12,16,22,30- hcptaazapentacyclo [22.2.2.1-3,7-.1-9,13-.! -14,18~]hentriaconta- 3(31 ),·4,6,9(30), 10,12,14(29), 15,17-nonaen-23-one. Referred to herein as “Compound 38”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (41) .
Formula (41): 9-Mcthyl- 2,3.4,5,7,8,9,10-octahydro-16H-17,21 -(azeno)-11,15-(mctheno)pyrido[3,2- g][ 1,3,5,9,13,17]hexaazacyclotricosin-6( 1 H)-one. Referred to herein as “Compound 39”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula
(42) .
Formula (42): 14-Prop-2-en-l-yl- 3,5,7,14,17,23,27-hcptaazatetracyclo[ 19.3.1.1 -2,6-. 1 -8,12-]heptacosa-1 (25),2(27),3,5,8(26),9,11,21,23-nonaen-16-one. Referred to herein as “Compound 40”.
In one embodiment, the cyclic aniline-pyridinotriazinc is a compound of the Formula (43) .
Formula (43): 18-Oxo-14-oxa- 2,4,8,17,25-pentaazatctracyclo[l 7.3.1.1-3,7~. 1 ~9,13~]pentacosa-1(23),3(25),4,6,9(24), 10,12,19,21 -nonacne-6-carbonitrile. Referred to herein as “Compound 41"
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Fonnula (44) .
Fonnula (44): 14,21-Dioxa- 2,4,8,18,28-pcntaazatctracyclo[20.3.1.1 ~3,7~. I ~9,13~]octacosa- 1(26),3(28),4,6,9(27), 10,12,22,24-nonacn-19-one. Referred to herein as “Compound 42”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (45) .
Formula (45); 21-Methyl- 1,8,10,1 1,21,24,30- heptaazapentacyclo[22.2.2.1 -3,7-. 1 -9,12-. 1-13,17-]hentriaconta- 3(31 ),4,6,9,11,13(29), 14.16-octaen-23-onc. RefeiTed to herein as “Compound 43”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (46) .
Formula (46): (18S)-ll- (Morpholin-4-yIcarbonyl)-5,7,14,20,28-pentaazapcntacyclo[20.3.1.1 -2,6-.1-8,12-.0-14.18-]octacosa-1(26),2(28),3,5,8(27),9,1 l,22,24-nonaen-!9-onc. Referred to herein as “Compound 44”.
In one embodiment, the cyclic aniline-pyridinotriazine is a compound of the Formula (47) .
Formula (47): 10-Methoxy-17- methyl-2,14,15,17,18,19,20,22-octahydro-6H-19,21 -mcthano-7,11 -(metheno)-12-oxa-2,3,5,6,17,2l-hexaazacycioicosa[l,2,3-cd]indcn-l6( 13H)-onc. Referred to herein as “Compound 45”.
In one embodiment, the at least one other factor is a compound of the Formula (48):
Formula(48). N-j[l- (Phenylmethyl)azepan-4-yI]methyl}-2-pyridin-3-ylacctamide. Referred herein as “Compound 46”.
In one embodiment, the at least one other factor is a compound of the Formula (49):
Formula (49). 4- j [4-(4-{[2-(Pyridin-2-ylamino)cthyl]amino} -1,3,5-triazm-2-yl)pyridin-2-yl]oxy} butan-1 -ol Referred herein as “Compound 47”.
In one embodiment, the at least one other factor is a compound of the Formula (50):
Formula (50). 3-({3-[4-({2- [Methyl(pyridin-2-yl)amino]ethyl} amino )-1,3,5-triazin-2-yl]pyridin-2-yl}amino)propan-I-ol. Referred herein as “Compound 48”.
In one embodiment, the at least one other factor is a compound of the Formula (51 ):
Formula (51). N-4—[2-(3-
Fluoropheny I )ethyl ]-N~2—[3-(4-methy Ipiperazin-1 -yl )propyl]pyrido[2,3-d]pyrimidine-2,4-diamine. Referred herein as “Compound 49”.
In one embodiment, the at least one other factor is a compound of the Formula (52):
Formula (52). I -Methyl-N-[(4- pyridin-3-yl-2-{[3-(trifluoromethyl)phenyl]aminoi-] ,3-thiazol-5-yI)methyl]piperidine-4-carboxamide. Referred herein as ‘'Compound 50”.
In one embodiment, the at least one other factor is a compound of the Formula (53):
Formula (53). 1,1-Dimethylethyl {2-[4-({5-[3-(3-hydroxypropyl)phenyl]-4H-l,2,4-triazol-3-yl}amino)phenyl]ethyl}carbamate. Referred herein as “Compound 51”.
In one embodiment, the at least one other factor is a compound of the Formula (54):
Formula (54). 1,1 -Dimcthylethyl {[3-( {5-[5-(3-hydroxypropyl )-2-(methyloxy)phenyl]-1,3-oxazol-2-yl}amino)phenyl]methyl [carbamate. Referred herein as “Compound 52”.
In one embodiment, the at least one other factor is a compound of the Formula (55):
Formula (55). 1-( {5-[6-({4-[(4-
Methylpiperazin-1 -yl)sulfonyl]phcnyl J amino)pyrazin-2-yl]thiophen-2-yI}methyl)piperidin-4-ol. Referred herein as “Compound 53”.
In one embodiment, the at least one other factor is a compound of the Formula (56):
Formula (56). l-({4-[6-({4-[(4-
Methy Ipiperazin-1 -yl )sulfony IJphcnyl} amino)pyrazin-2-yl]thiophcn-2-yl}methyl )piperidine-4-carboxamide. Referred herein as “Compound 54”.
In one embodiment, the at least one other factor is a compound of the Formula (57):
Formula (57). 2-{[4-( 1 -
MethylcthyI)phcnyl]amino}-N-(2-thiophen-2-ylethyl)“7,8-dihydropyrido[4,3-d]pyrimidine-6(5H )-carboxamide. Referred herein as “Compound 55”.
In one embodiment, the at least one other factor is a compound of the Formula (58):
Formula (58). 6-[(2-J[4- (2,4-Dichloropheny I )-5-(4-mcthyl-1 H-imidazol-2-yl )pyrimidin-2-yl]amino|cthyl)amino]pyridinc-3-carbonitnlc. Referred herein as “Compound 56”.
In one embodiment, the at least one other factor is a compound of the Formula (59):
Formula (59). 4-(6-{[(3-
Chloroplieny!)mcthyl]aminO(irnidazo[l,2-b]pyridazin-3-yl)-N-[2-(dimethylamino)ethyl]benzamide. Referred herein as “Compound 57”.
Detection of cells expressing markers characteristic of the definitive endodenn lineage
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. PI impotent stem cells typically do not express such markers. Thus, differentiation of pi impotent 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.
Methods for assessing expression of protein and nucleic acid markers in cultured or isolated cells are standard in the art. These include quantitative reverse transcriptase polymerase chain reaction (RT-PCR), Northern blots, in situ hybridization (see, e.g.. Current Protocols in Molecular Biology (Ausubel et a/., eds. 200! supplement)), and 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)).
For example, characteristics of pluripotcnt stem cells arc well known to those skilled in the art, and additional characteristics of pluripotcnt stem cells continue to be identified. Pluripotcnt stem cell markers include, for example, the expression of one or more of the following: ABCG2, cripto, FOXD3, Connexin43, Connexin45, OC'T4, SOX2, Nanog, hTERT, UTF-1, ZFP42, SSEA-3, SSEA-4, Tra I -60, or Tra 1-81.
After treating pluripotent stem cells with the methods of the present invention, 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.
Formation of cells expressing markers characteristic of the pancreatic endoderm lineage
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 ait or by any method proposed in this invention.
For example, ceils expressing markers characteristic of the definitive endoderm lineage may be differentiated into cells expressing markers characteristic of the pancreatic endoderm lineage according to the methods disclosed in D’Amour et cil. Nature Biotechnology 24, 1392 - 1401 (2006).
For example, 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. An example of this method is disclosed in Nature Biotechnology 24, 1392 - 1401 (2006).
In one aspect of the present 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 retinoic acid and at least one fibroblast growth factor for a period of time, according to the methods disclosed in US patent application Ser. No. 11/736,908, assigned to LifeScan, Inc.
In one aspect of the present 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 retinoic acid and at least one fibroblast growth factor for a period of time, according to the methods disclosed in US patent application Ser. No. 11/779,311, assigned to LifeScan, Inc.
In one aspect of the present 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 according to the methods disclosed in US 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. Alternatively, 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 . Further, 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-β family, including TGF-βΙ, 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, - I I), glucagon like peptide-l 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 1 and II, copper chelators such as, for example, triethylenc pentamine, forskolin, Na-Butyrate, activin. bctaceliulin, ITS, noggin, neurite growth factor, nodal, valproic acid, trichostatin A, sodium butyrate, hepatocyte growth factor (HGF), sphingosine-1, VEGF, MG132 (EMD, C'A), N2 and B27 supplements (Gibco, CA), steroid alkaloid such as, for example, cyclopamine (EMD, CA), keratinocyte growth factor (K.GF), Dickkopf protein family, bovine pituitary extract, islet neogenesis-associated protein (INGAP), Indian hedgehog, sonic hedgehog, protcasome inhibitors, notch pathway inhibitors, sonic hedgehog inhibitors, or combinations thereof.
The at least one other additional factor may be supplied by conditioned media obtained from pancreatic cells lines such as, for example, PANC-1 (ATC'C No: CR.L-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).
Detection of cells expressing markers characteristic of the pancreatic endoderm linage
Markers characteristic of the pancreatic endoderm lineage are well known to those skilled in the art, and additional markers characteristic of the pancreatic endoderm 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 endoderm lineage. Pancreatic endoderm lineage specific markers include the expression of one or more transcription factors such as, for example, Hlxb9, PTF-la, PDX-1, HNF-6, HNF-lbcta.
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.
Methods for assessing expression of protein and nucleic acid markers in cultured or isolated cells are standard in the art. These include quantitative reverse transcriptase polymerase chain reaction (RT-PCR), Northern blots, in situ hybridization (see, c.g.. Current Protocols in Molecular Biology (Ausubel et a/., eds. 2001 supplement)), and immunoassays such as immunohistochcmical 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)).
Formation of cells expressing markers of the pancreatic endocrine lineage
Cells expressing markers characteristic of the pancreatic cndodcrm 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.
For example, 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 etal, Nature Biotechnology 24, 1392 - 1401 (2006).
For example, cells expressing markers characteristic of the pancreatic endoderm lineage arc 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 cxendin 4, then removing the medium containing DAPT and cxendin 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).
For example, 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 cxendin 4, then removing the medium containing cxendin 4 and subsequently culturing the cells in medium containing cxendin 1, IGF-1 and HGF. An example of this method is disclosed in D’ Amour et al, Nature Biotechnology, 2006.
For example, cells expressing markers characteristic of the pancreatic endoderm lineage arc further differentiated into cells expressing markers characteristic of the pancreatic endocrine iineage, 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.
For example, 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.
In one aspect of the present invention, 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 US patent application Ser. No. 11/736,908, assigned to LifcScan, Inc.
In one aspect of the present invention, cells expressing markers characteristic of the pancreatic endoderm lineage arc further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the cells expressing markers characteristic of the pancreatic endoderm lineage with a factor that inhibits the Notch signaling pathway, according to the methods disclosed in US patent application Ser. No. 11/779,311, assigned to LifeScan, Inc.
In one aspect of the present invention, 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 US patent application Ser. No. 60/953,178, assigned to LifcScan, Inc.
In one aspect of the present invention, cells expressing markers characteristic of the pancreatic endoderm lineage arc further differentiated into cells expressing markers characteristic of the pancreatic endocrine lineage, by treating the ceils expressing markers characteristic of the pancreatic endoderm lineage according to the methods disclosed in LIS 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.
Alternatively, 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-β family, including TGF-βΙ, 2, and 3, scrum 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 pcptidc-1 and II (GLP-I and II), GLP-1 and GLP-2 mimetobody, Exendin-4, retinoic acid, parathyroid hormone, insulin, progesterone, aprotinin, hydrocortisone, cthanolaminc, beta mercaptocthanol, epidermal growth factor (EGF), gastrin I and II, copper chelators such as, for example, tricthylcnc pcntaminc, forskolin, Na-Butyratc, activin, bctaccllulin, ITS, noggin, neurite growth factor, nodal, valproic acid, trichostatin A, sodium butyrate, hepatocyte growth factor (HGF), sphingosine-1, VEGF, MG132 (EMD, C’A), N2 and B27 supplements (Gibco, CA), steroid alkaloid such as, for example, cyclopamine (EMD, CA), keratinocyte growth factor (KGF), Dickkopf protein family, bovine pituitary extract, islet neogenesis-associated protein (INGAP), Indian hedgehog, sonic hedgehog, proteasome inhibitors, notch pathway inhibitors, sonic hedgehog inhibitors, or combinations thereof.
The at least one other additional factor may be supplied by conditioned media obtained from pancreatic cells lines such as, for example, PANC-I (ATCC No: CRL-1469), CAPAN-] (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).
Detection of cells expressing markers characteristic of the pancreatic endocrine linage
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 1SLI.
Markers characteristic of cells of the β cell lineage are well known to those skilled in the art, and additional markers characteristic of the β cell 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 β-ccll lineage, β cell lineage specific characteristics include the expression of one or more transcription factors such as, for example, PDX1, NKX2.2, NKX6.1, ISLI, PAX6, PAX4, NEUROD, HNF1 beta, F1NF6, 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. Alternatively, 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 β cell lineage.
Methods for assessing expression of protein and nucleic acid markers in cultured or isolated cells arc standard in the art. These include quantitative reverse transcriptase polymerase chain reaction (RT-PCR), Northern blots, in situ hybridization (see, e.g.. Current Protocols in Molecular Biology (Ausubel et at., eds. 2001 supplement)), and immunoassays such as immunohistochemicai analysis of sectioned material. Western blotting, and for markers that arc 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)).
In one aspect of the present invention, the efficiency of differentiation is determined by measuring the percentage of insulin positive cells in a given cell culture following treatment. In one embodiment, the methods of the present invention produce about 100% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 90% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 80% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 70% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 60% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 50% insulin positive cells in a given culture. In an alternate embodiment, the methods of the present invention produce about 40% insulin positive cells in a given culture. In an alternate embodiment, 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.
In one aspect of the present invention, 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. In one embodiment, cells produced by the methods of the present invention produce about lOOOng C-pcptide/pg DNA. in an alternate embodiment, cells produced by the methods of the present invention produce about 900ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 800ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 700ng C-pcptidc/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 600ng C-pcptidc/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 500ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 400ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 500ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 400ng C-peptidc/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 300ng C-pcptide./pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 200ng C-peptidc/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about lOOng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 90ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 80ng C-pcptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 70ng C-peptidc/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 60ng C-peptide/pg DNA. In an alternate embodiment, ceils produced by the methods of the present invention produce about 50ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 40ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 30ng C-peptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about 20ng C-pcptide/pg DNA. In an alternate embodiment, cells produced by the methods of the present invention produce about lOng C-pcptide/pg DNA.
Therapies
In one aspect, the present invention provides a method for treating a patient suffering from, or at risk of developing, Typel diabetes. This method involves culturing pluripotent stem cells, differentiating the pluripotent stem cells in vitro into a β-cell lineage, and implanting the cells of a β-cell lineage into a patient.
In yet another aspect, 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 β-cell lineage, and implanting the cells of 3β-οε11 lineage into the patient.
If appropriate, the patient can be further treated with pharmaceutical agents or bioactives that facilitate the survival and function of the transplanted cells. These agents may include, for example, insulin, members of the TGF-β family, including TGF-βΙ, 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-1,11) growth differentiation factor (such as, for example, GDF-5, -6, -7, -8,-10,-15), vascular endothelial cell-derived growth factor (VEGF), plciotrophin, endothelin, among others. Other pharmaceutical compounds can include, for example, nicotinamide, glucagon like peptide-1 (GLP-I) and II, GLP-1 and -2 mimetibody, Excndin-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.
The pluripotcnt stem cells may be differentiated into an insulin-producing cell prior to transplantation into a recipient. In a specific embodiment, the pluripotent stem cells are fully differentiated into β-cells prior to transplantation into a recipient. Alternatively, 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, β cells, may be implanted as dispersed cells or formed into clusters that may be infused into the hepatic portal vein. Alternatively, cells may be provided in biocompatiblc degradable polymeric supports, porous non-degradablc 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.
To enhance further differentiation, survival or activity of the implanted cells, additional factors, such as growth factors, antioxidants or anti-inflammatory agents, can be administered before, simultaneously with, or after the administration of the cells. In certain embodiments, 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.
In one aspect, this invention provides a method for treating a patient suffering from, or at risk of developing diabetes. This method involves culturing pluripotcnt stem cells, differentiating the cultured cells in vitro into a β-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. Alternatively, 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. In particular, synthetic and natural materials in the form of foams, sponges, gels, hydrogels, textiles, and non woven structures, which have been used in vitro and in vivo to reconstruct or regenerate biological tissue, as well as to deliver chcmotactic 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. Patent 5,770.417, U.S. Patent 6,022,743, U.S. Patent 5,567,612, U.S. Patent 5,759,830, U.S. Patent 6,626,950, U.S. Patent 6,534,084, U.S. Patent 6,306,424, U.S. Patent 6,365,149, U.S. Patent 6,599,323, U.S. Patent 6,656,488, U.S. Published Application 2004/0062753 Al, U.S. Patent 4,557,264and U.S. Patent 6,333,029.
To form a support incorporated with a pharmaceutical agent, the pharmaceutical agent can be mixed with the polymer solution prior to forming the support. Alternatively, 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. Alternatively, excipients may be added to the support to alter the release rate of the pharmaceutical agent. In an alternate embodiment, 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. Patent 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.
Patent 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. Patent 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 w ith at least one pharmaceutical compound that is an immunosuppressive compound, such as, for example, compounds disclosed in U. S. Published Application 2004/0171623.
The support may aiso be incorporated with at least one pharmaceutical compound that is a growth factor, such as, for example, members of the TGF-β family, including TGF-βΙ, 2, and 3, bone morphogcnic 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), plciotrophin, endothelin, among others. Other pharmaceutical compounds can include, for example, nicotinamide, hypoxia inducible factor 1-alpha, glucagon like peptide-1 (GLP-1), GLP-1 and GLP-2 mimetibody, and II, Exendin-4, nodal, noggin, NGF. retinoic acid, parathyroid hormone, tcnascin-C, tropoclastin, thrombin-derived peptides, cathclicidins, 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.
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. For example, spinner flasks have been used in seeding of chondrocytes onto polyglycolic acid scaffolds (Biotcchnol. 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. For example, Yang et at. developed a cell seeding method (J. Biomed. Mater. Res. 55(3): 379-86 (2001)), referred to as Centrifugational Cell Immobilization (CCI).
The present invention is further illustrated, but not limited by, the following examples.
EXAMPLES
For clarity of disclosure, and not by way of limitation, the detailed description of the invention is divided into the following subsections that describe or illustrate certain features, embodiments, or applications of the present invention.
Example 1
Human embryonic stem cell culture
The human embryonic stem cell lines HI, H7, and H9 were obtained from WiCell Research Institute, Inc., (Madison, Wl) 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 MATRIGEL™ (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 MATRIGEL™ were routinely passaged as clusters using collagcnasc IV (Invitrogen/GIBCO; Cat # 17104-019), Dispase (Invitrogen; Cat # 17105-041), or Liberase Cl 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. C'lusters 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.
Example 2
Bioassav for the Formation of Cells Expressing Markers Characteristic of the
Definitive Endoderm Lineage
Activin A is an important mediator of differentiation in a broad range of cell types, including differentiation of embryonic stem cells to definitive endoderm. When human embryonic stem cells arc treated with a combination of activin A and Wnt3a. various genes representative of definitive endoderm arc up-rcgulated. 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 SOX 17, considered to be a representative marker of definitive endoderm.
Live Cel/ Assay: Briefly, clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATR1 GEL1M (Inyitfogen; 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 MATRJGELl v,-coated 96-wcli black plates (Packard ViewPlates; Perkin Elmer; Cat #6005182). Cells were allowed to attach as clusters and then recover log phase growth over a I to 3 day period, feeding daily with 100μ1 per well mouse embryonic fibroblast (MEF) conditioned medium supplemented with 8ng/ml bFGF (R&D Systems; Cat # 233-FB).
The assay was initiated by washing the wells of each plate twice in PBS (Invitrogen; Cat # 14190), followed by adding an aliquot (ΙΟΟμΙ) of test sample in DMEM:F12 basal medium (Invitrogen; Cat # 11330-032) to each well. Test conditions were performed in triplicate, feeding on alternate days by aspirating and replacing the medium from each well with test samples over a total four-day assay period. On the first and second day of assay, test samples added to the assay wells were diluted in DMEM:F!2 with 0.5% FCS (HyClonc; Cat # SH30070.03) and 20ng/m! Wnt3a (R&D Systems; Cat # 1324-WN). On the third and fourth day of assay, test samples added to the assay wells were diluted in DMEM:F12 with 2% FCS, without any Wnt3a. Positive control samples consisted of recombinant human activin A (PeproTech; Cat #120-14) added at a concentration of lOOng/ml throughout assay plus Wnt3a (20ng/ml) on days 1 and 2. Negative control samples omitted treatment with both activin A and Wnt3a.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-01 !) at room temperature for 20 minutes, then washed three times with PBS and permcabiiizcd with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat # 16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. Alcxa 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. To counter stain nuclei, 4pg/ml Hocchst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 SOX 17 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.
Figure I shows validation of the screening assay, testing a two-fold dilution curve of a commercial source of activin A (PcproTech) and measuring both cell number (Figure 1A) and SOX1 7 intensity (Figure IB). Optimal activin A effects for induction of SOX 17 expression were generally observed in the 100-200ng/ml range with an ECjo of 30-50ng/ml. Omitting Wnt3a from treatment on days 1 and 2 of assay failed to produce measurable SOX17 expression (Figure IB, white bars). Absence of activin A also failed to yield SOX17 expression (Figure IB).
Example 3
Primary Screening: Effects of the Compounds of the Present Invention on the
Differentiation of Human Embryonic Stem Cells into Cells Expressing Markers
Characteristic of the Definitive Endoderm Lineage in the Absence of Activin A
Differentiation of pluripotcnt stem cells into cells expressing markers characteristic of the definitive endoderm lineage is mediated through a series of rcccptor-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. In other cases, redundant pathways involving alternative members of a larger kinase family may substitute in part for one or more signaling molecules. In other cases, 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. Alternatively, 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. In this case, a library of small molecules comprising aniline-pyridinotriazines, cyclic aniline-pyridinotriazines and intermediate structures in their synthesis was tested for properties important during definitive endodenn 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.
Screening Assay
CelI assay seeding: Briefly, clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATRIGELIM (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 MATRIGEL IM-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of 100 μΙ/well. Cells were allowed to attach as clusters and then recover log phase growth over a I to 3 day period, feeding daily with MEF conditioned medium supplemented with 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates w'crc maintained at 37°C, 5% CO; in a humidified box throughout the duration of assay.
Preparation of compounds and 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 50mM 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. On the first day of assay, test volumes of 200μΙ per well were added back containing DMEM:F12 base medium (Invitrogen; Cat # 11330-032) supplemented with 0.5% FCS (HyClone; Cat # SH30070.03) and 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN) plus 2.5μΜ test compound. On the third day of assay, test volumes of 200μ1 per well were added back containing DMEM:F12 base medium supplemented with 2% FCS plus 2.5μΜ test compound, without Wnt3a. Positive control samples contained the same base medium supplemented with FCS, substituting lOOng/ml recombinant human activin A (PeproTcch; Cat #120-14) for the test compound throughout the four-day assay along with Wnt3a (20ng/ml) added only on days 1 and 2. Negative control samples contained DMEM:F12 base medium supplemented with FCS, adding Wnt3a on days I and 2 but omitting activin A.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken scrum (Invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # AFI924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat lgG; Molecular Probes; Cat # AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS. To counter stain nuclei, 4pg/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 S0X17 intensity were obtained from each wei! using IN Cel! 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 SOX 17 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 SOX 17 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 SOX! 7 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 w as 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.
In some cases, SOX 17 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.
Example 4
Secondary Screening: Effects of the Compounds of the Present Invention on the Differentiation of Human Embryonic Stem Cells into Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage with EGF/FGF4 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 coll 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.
Cell assay seeding: Clusters of ΗI human embryonic stem cells were grown on reduced growth factor MATRIGELiai( Invitrogen; Cat # 356231) -coated tissue culture plastic. Cells were passaged using collagenasc (Invitrogen; Cat # 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 MATRlGELlv,-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of ΙΟΟμΙ/well. 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 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% CO2 in a humidified box throughout the duration of assay.
Preparation of compounds and growth factors: Stock concentrations for EGF (R&D Systems; Cat # 236-EG) and FGF4 (R&D Systems; Cat #235-F4) wrere 250 ng/ml, each solubilized in PBS with 0.1% BSA (Sigma; Cat # A7888). Compounds were available as 5 mM stocks in 96-wclI plate format, solubilized in 100% DMSO (Sigma; Cat # D2650) and stored at -80°C. The compounds were further diluted to an intermediate concentration of 0.2 mM in 50mM HEPES (Invitrogen; Cat # 15630-080), 20% DMSO and stored at 4°C. All growth factors and inhibitors were prepared in a deep well, 96-weIi polypropylene plate, diluted to 5x intermediate stocks in DMEM:F i 2 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 scrum. Test volumes of 80μΙ per well were added back containing DMEM:F12 base medium (Invitrogen; Cat # 11330-032) supplemented with 0.625% FCS (HyClone; Cat# SH30070.03), 25ng/ml Wnt3a (R&D Systems), and 3,125μΜ compound plus 20μΙ 5x stock of growth factors to yield a final concentration of 0.5% FCS, 20ng/ml Wnt3a, and 2.5μΜ compound plus 50ng/ml EGF and 50ng/ml FGF4 in the assay. Positive control wells (ΙΟΟμΙ/well) contained the same base medium supplemented with 0.5% FCS, 20ng/ml Wnt3a and lOOng/ml activin A. Negative control wells (ΙΟΟμΙ/well) contained the same base medium with 0.5% FCS and 20ng/ml Wnt3a, omitting activin A.
On day 3, wells were aspirated and fed with 80μ! DM EM :F 12 base medium supplemented with 2.5% FCS (HyClone) and 3.125 μΜ compound plus 20μ1 5x stock of growth factors per well to yield a final concentration of 2% FCS and 2.5μΜ compound (omitting Wnt3a) plus 50ng/m! EGF and FGF4 in the assay. Positive control wells {ΙΟΟμΙ/well) contained the same base medium supplemented with 2% FCS and lOOng/ml activin A, omitting Wnt3a. Negative control wells (ΙΟΟμί/ well) contained the same base medium with 2% FCS, omitting both activin A and Wnt3a.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS, fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and pcrmcabilizcd with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. Alcxa 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. To counterstain nuclei, 4pg/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 SOX 17 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 3A shows the results for two growth factors, EGF and FGF 4 (50ng/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 arc ranked in descending order for best effects on SOX 17 expression. Although the effects of these compounds on SOX 17 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. In addition, these compounds appear to synergize with EGF and FGF4 to promote modest definitive endoderm differentiation, as measured by SOX 17 expression. The most potent compounds are listed in Table 3B. Other compounds tested in combination with EGF and FGF4 in this assay were ineffective at inducing SOX17 expression but could retain cell numbers in assay (e.g. Compound 90: 85% cell number; 2% SOX 17 expression).
Example 5
Effects of Compounds of the Present Invention in Combination with Other Factors 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 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.
Cell assay seeding: Clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATR1GEL1M (Invitrogen; Cat # 356231) -coated tissue culture plastic. Ceils were passaged using collagcnasc (Invitrogen; Cat # 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 MATRIGEL1 ^-coated 96-wcll black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of ΙΟΟμΙ/well. Cells were allowed to attach as clusters and then recover log phase grcwth over a 1 to 3 day period, feeding daily with MEF conditioned medium supplemented with 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% C02 in a humidified box throughout the duration of assay.
Preparation of compounds and growth factors: Stocks of growth factors purchased from R&D Systems were EGF (Cat # 236-EG), 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 (Cat #293-VE), BMP-1 (Cat #1927-ZNJBMP-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 ). Other agents tested were purchased as follows: 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 5mM stocks in 96-wcll plate format, solubilized in 100% DMSO and stored at -80°C. The compounds of the present invention were further diluted to an intermediate concentration of0.2 mM in 50mM HEPES (Invitrogcn; 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 5x 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 80μΙ per well were added back containing DMEM:F12 base medium (Invitrogcn; Cat # 11330-032) supplemented with 0,625% FCS (HyClonc; Cat # SH30070.03), 25ng/ml Wnt3a (R.&D Systems), and 3.125μΜ compound plus 20μΙ 5x stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20ng/ml Wnt3a, and 2.5μΜ compound. All remaining growth factors were tested at a final assay concentration of SOngrinl (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 (20μΜ), PD98059 (I uM), LY294002 (2.5μΜ), U0124 (!μΜ), U0126 (I μΜ), sodium butyrate (0.5mM). Positive control wells (ΙΟΟμΙ/wcll) contained the same base medium supplemented with 0.5% FCS, 20ng/ml Wnt3a and lOOng/m! activin A. Negative control wells (ΙΟΟμΙ/wcll) contained the same base medium with 0.5% FCS and 20ng/ml Wnt3a, omitting activin A.
On day 3, wells were aspirated and fed with 80μ1 DMEM:F12 base medium supplemented with 2.5% FCS (HyClone) and 3.125 μΜ cyclic aniline-pyridinotriazinc compound plus 20μ1 5x stock of growth factors or small molecules per well to yield a final concentration of 2% FCS and 2.5μΜ compound (omitting Wnt3a) and as denoted on day one for all remaining growth factors or small molecules. Positive control wells (1 ΟΟμΙ/welI) contained the same base medium supplemented with 2% FCS and lOOng/ml activin A, omitting Wnt3a. Negative control wells (ΙΟΟμΙ/welI) contained the same base medium with 2% FCS, omitting both activin A and Wnt3a.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS, fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #16110082) in PBS for 30 minutes a! room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. 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. To counterstain nuclei, 4pg/ml Hoechst 33342 (Invitrogen; Cat # 143570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in lOOplAvell PBS for imaging.
Imaging was performed using an IN Cel! Analyzer 1000 (GE Healthcare) utilizing the 51008bs dlehroic for cells stained with Hoechst 33342 and Aiexa 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 SOX 17 intensity were obtained from each well using ΓΝ 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 SOX 17 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. In general, members of the BMP family (BMP-1, BMP-2, BMP-4, BMP-6, BMP-7, BMP-2/7) inhibited or had negligible effects on SOX 17 expression. The same was true for most of the small molecule enzyme inhibitors tested in this assay (LY294002, PD98059, U0126, U0I24, sodium butyrate). However, some members of the PDGF family (PDGF-A, -AB, -C, and -D) provided an increase in SOX 17 expression (10-25% of the activin A/Wnt3a control). Other growth factors showing similar increases in SOX 17 expression included EGF (34%), VEGF (18%), and FGF4 (17%), although FGF4 was not able to support retention of cel! numbers. The small molecule muscimol (GABAa receptor agonist) tested in combination with Compound 35 also provided a modest increase in SOX 17 expression; the GABAa antagonist bicuculline had no effect on SOX 17 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.
Example 6
Effects of the Compounds of the Present Invention in Combination with Other Factors on the Differentiation of Human Embryonic Stem Cells into Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage in the
Absence of Activin A 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. In this screen, a broader panel of compounds was evaluated in with these agents, either in single pair-wise comparisons or pooled combinations.
Cell assay seeding'. Clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATRIGEL1V1 (Invitrogcn; Cat # 356231) -coated tissue culture plastic. Cells were passaged using collagenase (lnvitrogen; 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 MATRIGELlv,-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of ΙΟΟμΙ/well. 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 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% C02 in a humidified box throughout the duration of the assay.
Preparation of compounds and growth factors'. Stocks of growth factors purchased from R&D Systems were EGF (Cat # 236-EG), FGF4 (Cat #235-F4), PDGF-A (Cat #221 -AA), PDGF-D (Cat #1159-SB), PDGF-A/B (Cat #222-AB), and VEGF (Cat #293-VE). 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, Compounds were available as 5 mM stocks in 96-well plate format, solubilized in 100% DMSO and stored at -80°C. Compounds were further diluted to an intermediate concentration of 0.2 mM in 50mM HEPES (lnvitrogen; Cat # 15630-080), 20% DMSO and stored at 4°C. All growth factors and inhibitors were prepared in a deep well, 96-wcl! polypropylene plate, diluted to 5x intermediate stocks in DMEM:Fi2 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 scrum. Test volumes of 80μΙ per well were added back containing DMEM:F12 base medium (lnvitrogen; Cat # 11330-032) supplemented with 0.625% FCS (HyClone; Cat # SH30070.03), 25ng/ml Wnt3a (R&D Systems), and 3.125μΜ compound plus 20μΙ 5x stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20ng/ml Wnt3a, and 2.5μΜ. All remaining growth factors were tested at a final assay concentration of 50ng/ml (EGF, FGF4, PDGF-A, PDGF-A/B, VEGF). Final assay concentration of muscimol was 20μΜ, Positive control wells (ΙΟΟμΙ/well) contained the same base medium supplemented with 0.5% FCS, 20rig/ml Wnt3a and lOOng/ml activin A. Negative control wells (ΙΟΟμΙ/well) contained the same base medium with 0.5% FCS and 20ng/ml Wnt3a, omitting activin A.
On day 3, wells were aspirated and fed with 80μ1 DMEM:F12 base medium supplemented with 2.5% FCS (FlyClone) and 3.125 μΜ compound plus 20μΙ 5x stock of growth factors or small molecules per well to yield a final concentration of 2% FCS and 2.5μΜ compound (omitting Wnt3a) and as denoted on day one for all remaining growth factors or small molecules. Positive control wells (ΙΟΟμΙ/well) contained the same base medium supplemented with 2% FCS and lOOng/ml activin A, omitting Wnt3a. Negative control wells (ΙΟΟμΙ/well) contained the same base medium with 2% FCS, omitting both activin A and Wnt3a.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS, fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Ceils were washed again three times with PBS and blocked with 4% chicken serum (lnvitrogen; Cat #161 10082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. 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. To counterstain nuclei, 4pg/ml Hocchst 33342 (lnvitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 SOX 17 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 5shows 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 SOX 17 expression with all growth factor combinations tested. However, some compounds were able to induce significant SOX 17 expression with some but not all growth factor combinations. One compound in particular. Compound 34, had significant synergistic responses with all growth factors tested and mediated increases in both cell numbers as well as SOX 17 expression in this assay: Compound 39 with 1) EGF+FGF4 = 77% of positive control response; or 2) EGF+FGF4+PDGF-AB = 68% of positive control response; or 3) EGF+FGF4+PDGF-A+VEGF = 31% of positive control response.
Example 7
Effects of Compound 34 in Combination with Other Factors on the Differentiation of Human Embryonic Stem Cells into Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage in the Absence of Activin A
In this example, an effort was made to analyze the minimum number of growth factors required in combination with the best cyclic aniline-pyridinotriazinc compound, Compound 34 to yield a robust SOX17 response in the absence of activin A. Also in this example, a new growth factor. GDF-8, was added for evaluation. GDF-8, also known as myostatin, is a member of the TGF-β family and has been shown to use the activin type 11 and TGF-β type I receptors (ALK4/5) to induce SMAD 2/3 phosphorylation.
Cell assay seeding: Clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATRIGEL1 λ1 (lnvitrogen; Cat # 356231) -coated tissue culture plastic. Cells were passaged using collagenase (lnvitrogen; 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 MATRIGELlvl-coatcd 96-well black plates (Packard ViewPlatcs; PerkinElmer; Cat #6005182) using volumes of ΙΟΟμΙ/wcll. 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 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% CO2 in a humidified box throughout the duration of assay.
Preparation of compounds and growth factors: Stocks of growth factors purchased from R&D Systems were EGF (Cat # 236-EG), FGF4 (Cat #235-F4), PDGF-A (Cat #221-AA), PDGF-D (Cat #1159-SB), PDGF-A/B (Cat #222-AB), VEGF (Cat #293-VE), and GDF-8 (Cat # 788-G8). 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-pyridinotriazinc 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 50mM HEPES (lnvitrogen; 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 5x intermediate stocks in DMEM:F 12 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 scrum. Test volumes of 80μΙ per well were added back containing DM EM:F 12 base medium (lnvitrogen; Cat # 11330-032) supplemented with 0.625% FCS (HyClone; Cat # SH30070.03), 25ng/ml Wnt3a (R&D Systems), and 3.Ι25μΜ Compound 27 plus 20μ1 5x stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20ng/ml Wnt3a, and 2.5μΜ Compound 34. All remaining growth factors were tested at a final assay concentration of 50ng/ml (EGF, FGF4, PDGF-A, PDGF-A/B, VEGF) with the exception of GDF-8 tested at 25ng/ml. Final assay concentration of muscimol was 20μΜ. Positive control wells (1 ΟΟμΙ/well) contained the same base medium supplemented with 0.5% FCS, 20ug/m! Wnt3a and 10Ong/ml activin A. Negative control wells (ΙΟΟμΙ/well) contained the same base medium with 0.5% FCS and 20ng/ml Wnt3a, omitting activin A.
On day 3. wells were aspirated and fed with 80f.il DMEM:F12 base medium supplemented with 2.5% FCS (HyClone) and 3.125 μΜ Compound 34 plus 20μΙ 5x stock of growth factors or small molecules per well to yield a final concentration of 2% FCS and 2.5μΜ Compound 34 (omitting Wnt3a) and as denoted on day one for all remaining growth factors or small molecules. Positive control wells (ΙΟΟμΙ/well) contained the same base medium supplemented with 2% FCS and lOOng/ml activin A, omitting Wnt3a. Negative control wells (ΙΟΟμΙ/well) contained the same base medium with 2% FCS, omitting both activin A and Wnt3a.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS, fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and pcmncabilizcd with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #161 10082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. 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. To counterstain nuclei, 4gg/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 dining the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX 17 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 SOX 17 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 SOX 17 expression was observed. Furthermore, GDF-8 and WntSa with Compound 34 were sufficient to yield SOX 17 expression {88% of control) in a range similar to that seen with lOOng/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.
Example 8
Additional Screening for Compounds Capable of Differentiating Pluripotent Stem Cells into Cells Expressing Markers Characteristic of the Definitive
Endoderm Lineage
Based on the compound structures for hits identified thus far, an analog search was conducted to find additional related compounds to test in the definitive endoderm bioassay. The substructure search yielded compounds for screening. Screening parameters for this assay were designed with the combination of factors that had yielded optimal results in previous assays, specifically combining EGF, FGF, PDGF-A,VEGF, PDGF-D, muscimol, and GDF-8 with the small molecule compound.
Cell assay seeding: Briefly, clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATR1GEL1M (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 MATRlGELlvl-coatcd 96-well black plates (Packard ViewPlates; PcrkinElmer; Cat #6005182) using volumes of 100 μΐ/wcll. 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 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% CO? in a humidified box throughout the duration of assay.
Preparation of compounds and assay: Growth factors purchased from R&D Systems were EGF (Cat # 236-EG), FGF4 (Cat #235-F4), PDGF-A (Cat #221-AA), PDGF-D (Cat #1159-SB), PDGF-A/B (Cat #222-AB), VEGF (Cat #293-VE), and GDF-8 (Cat # 788-G8). Muscimol was purchased from Tocris (Cat # 0289). Screening was conducted using a library of compounds that were made available as 5 mM stocks in 96-well plate format, solubilized in 100% DMSO (Sigma; Cat # D2650) and stored at -80l’C. The compounds were further diluted to an intermediate concentration of 0.2 mM in 50mM 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. On the first day of assay, test volumes of 200ul per well were added back containing DMEM:F12 base medium (Invitrogen;
Cat # 11330-032) supplemented with 0.5% FCS (HyClone; Cat # SH30070.03) and 20ng/ml Wnt3a (R&D Systems; Cat # I324-WN) plus 2.5μΜ compound. All remaining growth factors were tested at a final assay concentration of 50ng/ml (EGF, FGF4, PDGF-A, PDGF-A/B, VEGF) with the exception of GDF-8 tested at 25ng/ml. Final assay concentration of muscimol was 20 μΜ. 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 (PcproTcch; Cat #120-14). Negative control samples contained DMEM:F12 base medium supplemented with 0.5% FCS and 20ng/ml Wnt3a. On the third day of assay, test volumes of 200μ! per well were added back containing DMEM:F12 base medium supplemented with 2% FCS plus 2.5μΜ compound, without Wnt3a. All remaining growth factors were tested at a final assay concentration of 50ng/ml (EGF, FGF4, PDGF-A, PDGF-A/B, VEGF) with the exception of GDF-8 tested at 25ng/ml. Final assay concentration of muscimol was 20 μΜ. Positive control samples contained the same base medium supplemented with 2% FCS and fOOng/ml recombinant human activin A (PeproTeeh; Cat #120-14). Negative control samples contained DMEM:F12 base medium supplemented with 2% FCS. Positive control samples contained the same base medium supplemented with FCS, substituting lOOng/ml recombinant human activin A (PeproTeeh; Cat #120-14) for the aniline-pyridinotriazine compound throughout the four-day assay along with Wnt3a (20ng/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.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #161 10082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat lgG; Molecular Probes; Cat # AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS. To counterstain nuclei, 4pg/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/wcll PBS for imaging.
Imaging was performed using an IN Ceil 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 cel! loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total S0X17 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 SOX 17 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.
In Table 7, GDF-8 and a combination of growth factors/agonists (EGF, FGF, PDGF-A, VEGF, PDGF-D, muscimol) were tested with a new set of aniline-pyridinotriazine compounds. Results from two assay plates in this single experiment arc ranked with respect to SOX! 7 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.
Of note, four hits from the initial primary screening (Table 2 ) were duplicated in the analog library. Two of these compounds repeated as hits with the analog screening (Compound 34 and Compound 35; shown boxed in Table 8); one was a weak hit in the analog screening, and one compound did not repeat.
Example 9
Effects of the Compounds of the Present Invention on the Differentiation of Human Embry onic Stem Cells to Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage in the Presence of Low Concentrations of Activin
A
It was important to determine if the compounds that had been identified as hits in the definitive endoderm bioassays above could also show synergistic activity with very low doses of activin A. An initial evaluation was performed using the short hit list of cyclic aniline-pyridinotriazine compounds denoted in Table 3B.
Cell assay seeding: Clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATRIGELIV1 (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; I (surface area) on reduced growth factor MATRIGEL1 M-coatcd 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of ΙΟΟμΙ/well. Cells were allowed to attach as clusters and then recover log phase growth over a I to 3 day period, feeding daily with MEF conditioned medium supplemented with 8ng/ml bFGF (R&D Systems; Cat# 233-FB). Plates were maintained at 37°C, 5% C02 in a humidified box throughout the duration of assay.
Preparation of compounds and growth factors: Stocks of growth factors purchased from R&D Systems were EGF (Cat # 236-EG), FGF4 (Cat #235-F4), PDGF-A (Cat #22 l-AA), PDGF-D (Cat #1159-SB), PDGF-A/B (Cat #222-AB), VEGF (Cat #293-VE), and GDF-8 (Cat #788-G8). Activin A was purchased from PcproTech (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-wcll 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 50mM HEPES (Invitrogen; Cat # 15630-080), 20% DMSO and stored at 4°C. All growth factors and inhibitors were prepared in a deep well, 96-wcll polypropylene plate, diluted to 5x intermediate stocks in DM EM :F 12 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 80μ1 per well were added back containing DMEM:F12 base medium (Invitrogen; Cat # 11330-032) supplemented with 0.625% FCS (HyClone; Cat # SH30070.03), 25ng/ml Wnt3a (R&D Systems), 12.5ng/ml activin A. and 3.125μΜ compound plus 20μ1 5x stock of growth factor or small molecule to yield a final concentration of 0.5% FCS, 20ng/ml Wnt3a, lOng/ml activin A, and 2.5μΜ compound. All remaining growth factors were tested at a final assay concentration of 50ng/ml (EGF, FGF4, PDGF-A, PDGF-A/B, VEGF), with the exception of GDF-8 used at 25ng/ml. Final assay concentration of muscimol was 20uM. Positive control wells (ΙΟΟμΙ/well) contained the same base medium supplemented with 0.5% FCS, 20ng/ml Wnt3a and lOng/ml (low dose) or lOOng/ml (high dose) activin A. Negative control wells (ΙΟΟμΙ/well) contained the same base medium with 0.5% FCS and 20ng/ml Wnt3a, omitting activin A.
On day 3, wells were aspirated and fed with 80μΙ DMEM:F12 base medium supplemented with 2.5% FCS (HyClone), 12.5ng/ml activin A, and 3.125 μΜ compound plus 20μ1 5x stock of growth factors or small molecules per well to yield a final concentration of 2% FCS, lOng/ml activin A, and 2.5μΜ compound (omitting Wnt3a) and as denoted on day one for all remaining growth factors or small molecules. Positive control wells (ΙΟΟμΙ/well) contained the same base medium supplemented with 2% FCS and lOng/ml or lOOng/ml activin A, omitting Wnt3a. Negative control wells (ΙΟΟμΙ/ well) contained the same base medium with 2% FCS, omitting both activin A and Wnt3a.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS, fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and pcrmcabilizcd with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken scrum (Invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat lgG; Molecular Probes; Cat # AZI467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS. To counterstain nuclei, 4μg/ml Hocchst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/well PBS for imaging.
Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51 008bs 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 ceil number and total SOX i 7 intensity were obtained from each well using IN Cell Developer Toolbox 1.7 (GE Healthcare) software. Segmentation for the nuclei w'as determined based on grayscale levels (baseline range 100-300) and nuclear size. Averages and standard deviations were calculated for each replicate data set. Total SOX 17 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 show'ed 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.
Example 10
Effects of the Compounds of the Present Invention on the Differentiation of Single Human Embryonic Stem Cells to Cells Expressing Markers of the Definitive Endotlerm Lineage in the Absence of Activin A
Cyclic aniline-pyridinotriazine compounds w'ere 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 w'as shortened from four days to three days to provide a more narrow timeframe to measure results. Finally, the assay included two growth factors, EGF and FGF4 that had previously shown significant but sub-optimal effects on definitive cndoderm differentiation in the absence of activin A.
Screening Assay
Cell assay seeding: Briefly, clusters of Η1 human embryonic stem cells were grown on reduced growth factor MATRIGELim (lnvitrogcn; Cat # 356231) -coated tissue culture plastic. Cultures were treated with Accutase (Sigma; Cat # A6964), using equivalent volumes of 10ml per lOcm" 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 cclls/cirf on reduced growth factor MATRIGELlv,-coatcd 96-wcll black plates (Packard ViewPlates; Cat #6005182) using volumes of 1 ΟΟμΙ/wcll. 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/m! bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% C02 in a humidified box throughout the duration of assay.
Preparation of compounds and assay: Stocks of EGF and FGF4 were prepared in a 96-well polypropylene plate (Coming, Inc.; Cat # 3960). Compound 22 was available as a 5 mM stock solubilized in 100% DMSO (Sigma; Cat # D2650) and stored at-80l'C. Assays were initiated by aspirating culture medium from each well followed by three washes in PBS to remove residual growdi factors and serum. Test volumes of 80μ1 per well were added back containing RPMI 1640 base medium (lnvitrogcn; Cat # 22400-089) supplemented with 2.5% fatty acid free BSA (MP Biomcdicals LLC; Cat# 152401), lOng/mlbFGF(PeproTcch Inc; Cat# 100-18B), 25ng/ml Wnt3a (R&D Systems; Cat # 1324-WN) and 3.125μΜ Compound 22 plus 20μΙ 5x stock of growth factors to yield a final concentration of 2% fatty acid free BSA, 8ng/ml bFGF(PeproTech Inc; Cat # 100-I8B), 20ng/ml Wnt3a, and 2.5μΜ Compound 22 in assay. Positive control wells contained the same base medium supplemented with 2% fatty acid free BSA, 8ng/ml bFGF, 20ng/ml Wnt3a, and lOOng/ml recombinant human activin A (PeproTech; Cat #120-14). Negative control wells contained the same base medium supplemented with 2% fatty acid free BSA, 8ng/ml bFGF, 20ng/ml Wnt3a but omitted treatment with activin A.
On the second day of assay, wells were again aspirated and fed with 80μΙ per well were added back containing RPMI 1640 base medium supplemented with 2.5% fatty acid free BSA, lOng/ml bFGF, and 3.125μΜ Compound 22 plus 20j.il 5x stock of growth factors to yield a final concentration of 2% fatty acid free BSA, 8ng/ml bFGF and 2.5μΜ Compound 22 in assay. Positive control wells contained the same base medium supplemented with 2% fatty acid free BSA, 8ng/ml bFGF and lOOng/ml recombinant human activin A. Negative control samples contained the same base medium supplemented with 2% fatty acid free BSA and 8ng/ml bFGF but omitted treatment with activin A.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS, fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-01 I) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells wrere washed again three times with PBS and blocked with 4% chicken serum (lnvitrogen: Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-luiman SOX17; R&D Systems; cat # AFi924) was diluted 1; 100 in 4% chicken serum and added to each well for one hour at room temperature. Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Molecular Probes; Cat # AZI467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS. To counterstain nuclei, 4pg/ml Hoechst 33342 (lnvitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 ceil loss during the bioassay and subsequent staining procedures. Measurements for total cell number and total SOX 17 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 SOX 17 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 SOX 17 expression. Addition of Compound 34 added significant enhancement of SOX 17 expression.
Example 11 A Comparison of the Ability of Activin A and GDF-8 to Differentiate Human Embryonic Stem Cells to Cells Expressing Markers Characteristic of the
Definitive Endoderm Lineage A previous example showed that 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 (FI I human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRJGEL-coatcd 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 8ng/ml bFGF and seeded onto reduced growth factor MATRIGEL™-coatcd 96-well Packard VIEWPLATES (PerkinElmer; Cat # 6005182) in volumes of 100f.il/wcll. MEF conditioned medium supplemented with 8ng/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 w^ere maintained at 37°C, 5% CO2 in a humidified box throughout the duration of assay.
Assay: The assay was initiated by aspirating the culture medium from each well and adding back an aliquot (100μ1) 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 1ml assay medium composed of RPMI-1640 medium (Invitrogcn; Cat#: 22400) supplemented with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA)(MP Biomedicals, Inc; Cat # 152401) and 8ng/ml bFGF (PeproTech Inc.; Cat #: 100-I8B), and with 20ng/m! 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 fl esh 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 scries of two-fold test dilutions was created, containing Activin A or GDF-8 at concentrations ranging from 1 .bngdnl to 1600 ng/ml, for addition to the respective assay wells.
High Content Analysis: At the conclusion of three days of culture, assay plates were washed once with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-01 !) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #161 10082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-hum an SOX 17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken scrum and added to each well for two hours at room temperature. After washing three times with PBS, Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A2J467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5ug/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/well PBS for imaging.
Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 5 !008bs dichroic for ceils stained with Hoechst 33342 and Alexa Fluor 488. Images were acquired from 25 fields per well. Measurements for total SOX 17 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 SOX 17 intensity data were calculated using GraphPad Prism 4.02 (GraphPad Software, Inc., Lo Jolla, CA). Data were normalized to define the smallest and largest values in each data set as 0% and 100%, respectively. Table II shows the normalized values for each of the activin A and GDF-8 data sets. Two sigmoidal dose-response curves arc shown in Figure 2 as generated using the normalized values shown in Table 11. The R~ 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, EC50 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. These data indicate that 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 SOX 17 expression.
Example 12
Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage that were Formed According to the Methods of the Present Invention are able to Further Differentiate into Cells Expressing Markers Characteristic of the
Pancreatic Endocrine Lineage
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 (FI 1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor M ATRJ G ELIM-coated dishes in MEF conditioned medium with passage on average every four days. Passage was performed by exposing cell cultures to a solution of I mg/ml dispasc (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 dispasc. Cell clusters w'ere 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 8ng/ml bFGF and seeded onto reduced growth factor MATRIGELIM-coated 24-well, black wall culture plates (Arctic White; Cat # AWLS-303012) in volumes of 0.5ml/we!l. 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% CO2 throughout the duration of assay.
Assay: The assay was initiated by aspirating the culture medium from each well and adding back an aliquot (0.5ml) 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, lOOng/ml activin A (PeproTech; Cat #120-14) or 200ng/ml GDF-8 (R&D Systems, Cat # 788-G8) was added to respective assay wells where each growth factor was diluted into RPM1-1640 medium (Invitrogen; Cat # 22400) with 1% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat # 15240 i), 1 % Probumin (Milliporc; Cat # 81-068-3) and 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF), On the second day of assay, 1 OOng/ml activin A or 200ng/ml GDF-8 was diluted into RPMI-1640 medium supplemented with 2% FAF BSA without Wnt3a. In some test samples using GDF-8. Wnt3a was replaced with a either Compound 34 or Compound 56 at a concentration of 2.5μΜ, and either Compound 34 or Compound 56 was added daily during all three days of definitive endoderm differentiation. At the conclusion of the first step of differentiation, cells from some wells were harvested for flow cytometry analysis to evaluate levels of CXCR4, a marker of definitive endoderm formation. Additional wells were harvested for RT-PCR analysis to measure other markers of differentiation.
At the conclusion of the first step of differentiation, replicate sets of parallel wells from each treatment group were subjected to further step-wise differentiation. It is important to note that after the first differentiation step, all wells undergoing continuing culture and differentiation received the same treatment. The protocol for this continuing differentiation is described below.
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 (0.5ml) of DMEM:F12 medium (Invitrogen; Cat # 11330-032) containing 2%
Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomcdicals, Inc; Cat # 152401), 50ng/ml FGF7 (PeproTech; Cat # i00-19), and 250nM cyciopamine (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 (0.5ml) of DMEM-high glucose (lnvitrogen; Cat # 10569) supplemented with 1% B27 (Invitrogen; Cat # 17504-044), 50 ng/ml FGF7, 100 ng/mi Noggin (R&D Systems; Cat # 3344-NG), 250 nM KAAD-cyclopaminc (Calbiochem; Cat # 239804), and 2 μΜ all-trans retinoic acid (RA) (Sigma-Aldrich; Cat # R2625). At the conclusion of the third step of differentiation, cells from some wells were harvested for analysis by RT-PC'R to measure markers of differentiation. Other culture wells were subjected to high content image analysis for protein expression levels of Pdx 1, a transcription factor associated with pancreatic endoderm, and Cdx2. a transcription factor associated with intestinal endoderm.
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 (0.5ml) of DMEM-high glucose supplemented with 1% B27, 100 ng/ml Noggin, lOOng/ml Netrin-4, ΙμΜ DAPT (EMD Biosciences; Cat #565770), and ΙμΜ Aik 5 inhibitor (Axxora; Cat # ALX-270-445). At the conclusion of the fourth step of differentiation, cells from some wells were harvested for analysis by RT-PC'R to measure markers of differentiation. Other culture wells were subjected to high content image analysis for protein expression levels of PDX 1.
Step 5 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 1% B27, and ΙμΜ Aik 5 inhibitor. Medium in each well was aspirated and replaced with a fresh aliquot (0.5ml) on all days. At the conclusion of the fifth step of differentiation, 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 (0.5ml) 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 C’D9 PE (BD; Cat # 555372), C'D99 PE (Caltag; Cat # MHCD9904) and CXCR4 APC (R&D Systems; Cat# FAB 173A) for 30 minutes at 4°C. 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 IgGl K Isotype control antibody for both PE and APC was used to gate percent positive cells. RT-PCR Analysis: RNA samples were purified by binding to a silica-gel membrane (Rneasy Mini Kit, Qiagcn, CA) 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, rNC), 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.
Unless otherwise stated, all reagents were purchased from Applied Biosystems. Realtime 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 μΙ. Each cDNA sample was run in duplicate to correct for pipetting errors. Primers and FAM-labeled TAQM AN® probes were used at concentrations of 200 nM. The level of expression for each target gene was normalized using a human glyccraldehyde-3-phosphatc dehydrogenase (GAPDH) endogenous control previously developed by Applied Biosystems. Primer and probe sets arc 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 denatured on step at 95°C for 15 sec followed by an anneal in g/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. Briefly, for each cDNA sample, the endogenous control Ct value was subtracted from the gene of interest Ct to give the delta Ct value (ACt). The normalized amount of target was calculated as 2-ACt, assuming amplification to be 100% efficiency. Final data were expressed relative to a calibrator sample.
High Content Analysis: At the conclusion of culture, assay plates were washed once with PBS (Invitrogcn; Cat # 14390), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken scrum and added to each well for two hours at room temperature. After washing three times with PBS, Alcxa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5ug/mi Hoechst 33342 (Invitrogen; Cat # H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/well PBS for imaging. Other primary antibodies used for analysis included 1:100 dilution mouse anti-human CDX2 (Invitrogen; Cat # 397800), 1:100 dilution goat anti-human Pdxl (Santa Cruz Biotechnology; Cat # SC-14664), 1:200 dilution rabbit anti-human insulin (Cell Signaling; Cat # C27C9), and 1:1500 dilution mouse anti-human glucagon (Sigma-Aldrich; Cat # G2654). Secondary antibodies used for analysis included 1:400 dilution Alcxa Fluor 647 chicken anti-mouse IgG (Invitrogen; Cat # A-21463), 1:200 dilution Alcxa Fluor 488 donkey anti-goat IgG (Invitrogen; Cat # A11055), 1:1000 dilution Alcxa 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 grayscale 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 cndodcrmal and endocrine differentiation.
Figure 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. Similarly, treatment of human embryonic stem cells with GDF-8 and a small molecule (Compound 34 or Compound 56) also yielded an equivalent or slightly higher percentage of CXC4 positive cells. Figure 4 shows high content image analysis for normalized SOX 17 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.
Figure 5 shows high content image analysis for normalized Pdxl and Cdx2 protein expression in human embryonic stem cells after the third step of differentiation to pancreatic endodeim. 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. In some treatment groups the cell number retained after differentiation decreased thereby increasing the ratio of PDXI expressing cells. Similar results were obtained showing equivalent normalized PDX1 expression in all treatment groups after the fourth step of differentiation as shown in
Figure 6. In Figure 7, normalized protein levels of insulin and glucagon are shown, demonstrating equivalent expression between the Activin A and GDF-8 treatment groups.
These collective results demonstrate that 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.
Example 13
Formation of Cells Expressing Markers Characteristic of the Definitive
Endoderm Lineage with Other Members of the GDF Family of Proteins
It was important to determine if treating human embryonic stem cells with other GDF family members could the formation of cells expressing markers characteristic of the definitive endoderm lineage. Wnt3a in combination with cither Compound 34 or Compound 56 were tested on human embryonic stem cells in combination with six different GDF growth factors [GDF-3, GDF-5, GDF-8, GDF-10, GDF-1 1, and GDF-15] to determine the ability of members of the GDF family of proteins to differentiate human embryonic stem cells toward cells expressing markers characteristic of the definitive endoderm lineage. A parallel control of cells treated with activin A and Wnt3a was maintained for comparison purposes.
Preparation of cells for assay: Stock cultures of human embryonic stem cells (Η 1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATFUGEL1M (BD Bioscienccs; Cat # 356231 )-coatcd 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 (Invitrogem 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 8ng/m! bFGF and seeded onto reduced growth factor MATRIGELlv,-coated 96-well Packard V1EWPLATES (PcrkinElmer; 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? throughout the duration of assay. A$$ay: The assay was initiated by aspirating the culture medium from each well and adding back aliquots (ΙΟΟμΙ) 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. Various members of the GDF family of proteins were obtained for testing as follow's: 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). On the first day of assay, all wells received an aliquot (80μ!) of basal medium DMEM:FI2 medium (Invitrogen; Cat# 11330-032) supplemented with 0.5% fetal bovine serum (Hyclone; Cat # SH30070.03). A series of five different control or experimental test samples w'as created to evaluate activin A or various GDFs in combination w ith Wnt3a or Compound 34 or Compound 56. These test samples were added in 20μ1 aliquots (5x concentrated) to appropriately matched assay wells to yield a final assay volume of ΙΟΟμΙ 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.c. no supplementary growth factor or small molecule); 2) lOOng/ml activin A (PeproTech; Cat #120-14) in combination with 20ng/mi Wnt3a (R&D Systems; Cat # 1324-WN/CF); 3) 20ng/ml Wnt3a alone; 4) Compound 34 alone (2.5μΜ) without any growth factor or small molecule; 5) Compound 56 alone (2.5μΜ) without any grow'th factor or small molecule. In the second set of test samples, the following conditions were tested in combination with 100ng/ml GDF3; 1) no additive (i.e. GDF-3 alone); 2) 20ng/ml Wnt3a; 3) 20ng/m! Wnt3a with Compound 34 (2.5μΜ); 4) Compound 34 (2.5μΜ); 5) Compound 56 (2.5μΜ); and 6) 20ng/ml Wnt3a with Compound 56 (2.5μΜ). In the third set of test samples, each of the six conditions was combined with lOOng/ml GDF-5. In the fourth set of test samples, each of the six conditions was combined with 1OOng/ml GDF-8. In the fifth set of test samples, each of the six conditions was combined with ]00ng/ml GDF-10. In the sixth set of test samples, each of the six conditions was combined with lOOng/ml GDF-11. In the seventh set of test samples, each of the six conditions was combined with 1 OOng/ml GDF-15. On the third day of assay, ail wells for all test samples, received lOOng/mi Activin A or 1 OOng/ml respective GDF growth factor, without Wnt3a or Compound 34 or Compound 56, diluted into DMEM:F!2 medium supplemented with 2% FBS.
High Content Analysis: At the conclusion of culture, assay plates were washed once with PBS (Invitrogen; Cat # 14190). fixed with 4% paraformaldehyde (Alexis Biochemical: Cat# ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat # 161 10082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX1 7; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for two hours at room temperature. After washing three times with PBS, Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5gg/m! Hoechst 33342 (Invitrogen; Cat # H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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-scalc 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 grayscale 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.
Figure 8 shows high content image analysis for SOX 17 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 Figure 8A, only the positive control treatment yielded significant expression of SOX 17; treatment with Wnt3a alone or either Compound 34 or Compound 56 alone failed to induce SOX 17 expression. In Figure 8, panels B through G, normalized SOX 17 expression levels arc shown for each GDF growth factor substituting for activin A in the respective treatments. GDF-3 (Figure 8B) and GDF-5 (Figure 8C) induced weak expression of SOX 17 and only in test samples where one of the compounds of the present invention was present. GDFI0 (Figure 8D), GDFI 1 (Figure 8E) and GDF 15 (Figure 8G) induced significant levels of SOX 17 expression, more than observed with GDF3 or 5 treatments but less than observed that observed with activin A and Wnt3a treatment. In general, SOX 17 expression was negligible when GDF-10. GDF-11, or GDF-15 was combined with Wnt3a, but improved in combination with one of the compounds of the present invention; in particular when combined with Compound 34. Figure 8D shows results for treatment groups using GDF-8 where GDF-8 in combination with either Compound 34 or Compound 56caused a robust induction of SOX 17, exceeding results seen with the activin A/Wnt3a positive control. In some of these examples, the presence of Compound 34 or Compound 56 combined with a GDF growth factor also caused an increase in cell number during differentiation.
These collective results demonstrate that GDF-8 was superior to all other GDF family members tested when used in combination with Compound 34 or Compound 56, and could substitute for activin A during definitive endoderm differentiation.
Example 14
Formation of Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage with Other Members of the TGF Superfamily of Proteins
It was important to determine if treating human embryonic stem cells with other TGF superfamily members could facilitate the formation of cells expressing markers characteristic of the definitive endoderm lineage. Compound 34 and Wnt3a were tested on human embryonic stem cells in combination with either TGFP-l, BMP2, BMP3, or BMP4 to determine the ability of members of the TGF superfamily members to differentiate human embryonic stem cells toward cells expressing markers characteristic of the definitive endoderm lineage. In parallel, two different commercial sources of GDF-8 were tested with Wnt3a for their ability to differentiate human embryonic stem cells toward cells expressing markers characteristic of the definitive endoderm lineage. A positive control using activin A with Wnt3a was maintained for comparison purposes.
Preparation of cells for assay: Stock cultures of human embryonic stem cells (Η 1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRIGELim-(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 I mg/ml dispasc (lnvitrogen; 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 dispasc. Ceil 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 8ng/ml bFGF and seeded onto reduced growth factor MATRlGEL1M-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat # 6005182) in volumes of 0. Iml/wcll. 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% CO2 throughout assay.
Assay: The assay was initiated by aspirating the culture medium from each well and adding back aliquots (100μΙ) 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. Various growth factor proteins were obtained for testing as follows: BMP-2 (R&D Systems; Cat # 355-BM): BMP-3 (R&D Systems; Cat # 113-BP); BMP-4 (R&D Systems; Cat # 314-BP); TGFP-l (R&D Systems; Cat # 240-B); GDF-8 (PeproTech; Cat # 120-00); GDF-8 (Shenandoah; Cat #100-22); and activin A (PeproTech; Cat #120-14). On the first day of assay, each well was treated w ith 80μΙ of growth medium [RPMI-1640 (Jnvitrogen; Cat #; 22400) containing 2.5% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomedicals, Inc; Cat # 152401), and lOng/ml bFGF]. In some wells, 25ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF) was added to the growth medium to yield a final assay concentration of 20ng/mi. In some wells, activin A w;as added to the growth medium to yield a final assay concentration of lOOng/ml. In some wells, 3.125μΜ Compound 34was added to the growth medium to yield a final assay concentration of 2.5μΜ. A dose titration of additional growth factors (5x concentrated, diluted in RPMI-1640) was also added to respective test wells to yield a final assay volume of 100μ1 in each well for all treatment conditions. On the second day of assay, Wnt3a and Compound 34wcrc omitted from assay. All wells received 80μ1 of growth medium [RPMI-1640 containing 2.5% FAF BSA, and iOng/ml bFGF] and 20μΙ of respective growth factor dilution (5x concentrated, diluted in RPMI-1640). Comparative controls for this assay included: 1) no added growth factors; 2) Wnt3a alone; and 3) activin A with Wni3a. Each commercial source of GDF-8 W'as tested in combination with Wnt3a. Each of the BMP growth factors, as well as TGFP-l, was tested in combination with Wnt3a, with Compound 34, and with both Wnt3a in combination with Compound 34.
High Content Analysis: At the conclusion of culture, assay plates were washed once with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #161 10082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken scrum and added to each well for two hours at room temperature. After washing three times with PBS, Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A2J467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5ug/ml Hoechst 33342 (invitrogen; Cat # H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 Alcxa Fluor 488. Images were acquired from 25 fields per well. Measurements for total intensity were obtained from each well using ΓΝ 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 grayscale 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.
Figure 9 shows high content image analysis for SOX 17 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 Figure 9A, show that treatment with growth medium alone, or Wni3a alone failed to induce SOX 17 expression; only the addition of activin A caused a robust expression of SOX17. In Figure 9, panels B and C, results for each of tiie 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 SOX 17 expression in cells treated with GDF-8 in combination with Wnt3a. In Figure 9. panels D, E, F and G, results are shown for definitive endoderm differentiation using BMP2, BMP3, BMP4, and TGFP-l, incorporating a dose titration for each growth factor in combination with Wnt3a, or Compound 34, or both Wnt3a with Compound 34. Although some treatments had a significant effect on cell numbers at the conclusion of assay (e.g. BMP2 and BMP4), induction of SOX 17 expression resulting from any of these growth factors and treatment combinations was weak or negligible compared to the Wnt3a treatment alone.
Example 15
Dose Ranging Studies for Formation of Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage with a Selection of the Compounds of the
Present Invention
It was important to know the optimal working concentrations for Compound 181, Compound 180, Compound 19, Compound 202, Compound 40, and Compound 34 that would mediate the formation of cells expressing markers characteristic of the definitive endoderm lineage. In conjunction, side-by-side comparisons were performed for titrations of each compound in combination with activin A or GDF-8 in the definitive endoderm assay. Finally, the duration of exposure for each compound was tested in assay, also in combination with activin A or GDF-8, adding compound only on the first day of assay or throughout all three days of definitive endoderm formation.
Preparation of cells for assay: Stock cultures of human embryonic stem cells (Η 1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotcnt state on reduced growth factor MATRIGEL1M (BD Bioscicnccs: Cat # 356231 )-coated dishes in MEF conditioned medium supplemented with 8ng/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 I mg/ml dispase (lnvitrogen; 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 karyotype and absence of mycoplasma.
Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8ng/ml bFGF and seeded onto reduced growth factor MATRlGELl vl-coated 96-well Packard V1EWPLATES (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? throughout the duration of assay.
Assay: Assay was initiated by aspirating the culture medium from each well and adding back aliquots (ΙΟΟμΙ) 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. Each well was treated with 80μΙ of growth medium [RPM1-1640 (Invitrogen; Cat #: 22400) containing 2.5% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (MP Biomcdicals, Inc; Cat # 152401), lOng/ml bFGF, and additional growth factors {1,25x concentrated)] and 20μ1 of test compound (5x concentrated diluted in RPMI-1640) to yield a final assay volume of I OOul in each well. 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). On the first day of assay, wells were treated with various control or experimental conditions. Control conditions, with final assay concentrations as indicated, were as follows: 1) growth medium alone; 2) 20ng/ml Wnt3a only R&D Systems; Cat # 1324-WM/CF); 3) lOOng/ml activin A (PeproTech; Cat #120-14); 4) lOOng/ml activin A and 20ng/ml Wnt3a; 5) lOOng/ml GDF-8 (R&D Systems, Cat # 788-G8); 6) lOOng/ml GDF-8 and 20ng/ml Wnt3a.
Test compounds were diluted two-fold in scries to yield a concentration range from 78nM to ΙΟμΜ in the final assay. Experimental test samples combined each individual compound dilution series with lOOng/ml activin A or lOOng/ml GDF-8, both treatment sets in the absence of Wnt3a. On the second and third day of assay, some wells continued to be treated with 20ng/ml Wnt3a or diluted test compound in combination with either activin A or GDF-8. In other wells, activin A or GDF-8 treatment continued on the second and third day of assay, but Wnt3a or diluted test compound was removed.
High Content Analysis: At the conclusion of culture, assay plates were washed once with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked w'ith 4% chicken serum (Invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for two hours at room temperature. After washing three times with PBS. Alcxa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat #A21467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5gg/ml Hocchst 33342 (Invitrogen; Cat # H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/wcll PBS for imaging.
Imaging was performed using an IN Cell Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hocchst 33342 and Alcxa 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 grayscale 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 Figures 10 - 14 and resulting ceil number at the conclusion of assay in Figures 15-19. In Figure 10. results are shown for SOX 17 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 SOX 17 expression than was observed with GDF-8 treatment. Similarly, as seen in Figure 15, 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. Adding any of Compound 181, Compound 180, Compound 19, Compound 202, Compound 40, or Compound 34 with activin A treatment did not enhance SOX 17 expression (Figures 11 -12) or increase cell numbers (Figures 17-18), regardless of whether the compound was present for one day at the initiation of assay or three days throughout the duration of assay. However, treatment with either Compound 181, Compound 180, Compound 19, Compound 202, Compound 40, or Compound 34 in combination with GDF-8 significantly improved SOX17 expression (Figures 13-14) and also enhanced cell numbers at the end of assay (Figures 18-19). When either Compound 181. Compound 180, Compound 19, Compound 202, Compound 40, or Compound 34and GDF-8 were used in combination, the improvements to SOX 17 expression and cel! number in many cases were equivalent to results observed with activin A treatment, improved differentiation in combination with GDF-8was apparent in a dose titration effect for many of the compounds, although toxicity was sometimes observed at the highest concentrations. In most cases, optimal beneficial effects from treatment with the compound and GDF-8 were apparent with only one day of compound exposure at the initiation of assay. In some cases, presence of the compound throughout the duration of assay had no detrimental effect or had a slight beneficial effect. From these collective results an optimal working concentration range for each compound in combination with GDF8 treatment was determined. Results were compound specific, generally in the 1-10μΜ range as tested in this assay.
Example 16
Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage that were Formed Without According to the Methods of the Present Invention are able to Further Differentiate into Cells Expressing Markers Characteristic of the Pancreatic Endocrine Lineage
Additional small molecules were tested in combination with GDF-8 for definitive cndodeim differentiation. These 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 (Η 1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATR1GELIVI (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 dispasc (Invitrogcn, 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 dispasc. 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 nonrial karyotype and absence of mycoplasma.
Cell clusters were evenly resuspended in MEF conditioned medium supplemented with 8ng/ml bFGF and plated onto reduced growth factor MATRiGEL-coated 24-wcll, black wall culture plates (Arctic White; Cat # AWLS-303012) in volumes of 0.5ml/wcll. Daily feeding was conducted by aspirating spent culture medium from each well and replacing with an equal volume of fresh medium. Piates were maintained at 37°C, 5% CO? throughout the duration of assay.
Assay: The assay was initiated by aspirating the culture medium from each well and adding back an aliquot (0.5ml) of test medium. Test conditions for the first step of differentiation w'ere 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, lOOng/ml activin A (PeproTech; Cat #120-14) or lOOng/ml GDF-8 (R&D Systems, Cat # 788-G8) was added to respective assay wells where each growth factor was diluted into RPM1-1640 medium (Invitrogcn; Cat #: 22400) with 2% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA) (Proliant Inc. Cat #: SKU 68700), and 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF). On the second day of assay, lOOng/ml activin A or lOOng/ml GDF-8 was diluted into RPMI-1640 medium supplemented with 2% FAF BSA without Wnt3a. In some test samples using GDF-8, Wnt3a was replaced with a small molecule compound, added only on the first day of definitive endoderm differentiation. These small molecules included Compound 19 (2.5liM in assay), Compound 202 (2.5μΜ in assay), Compound 40 (2.5μΜ in assay), or a commercially available GSK3 inhibitor BIO (0.5μΜ in assay) (EMD Chemicals. Inc.; Cat # 361550 ). At the conclusion of the first step of differentiation, cells from some wells were harvested for flow cytometry analysis to evaluate levels of CXCR4, a marker of definitive endoderm formation. Additional wells were harvested for RT-PC’R analysis to measure other markers of differentiation.
At the conclusion of the first step of definitive endoderm differentiation, replicate sets of parallel wells from each treatment group were subjected to further step-wise differentiation. It is important to note that after the first differentiation step, all wells undergoing subsequent culture and differentiation received the same treatment. The protocol for this continuing differentiation is described below.
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 (0.5ml) of DMEM:F12 medium (Invitrogen; Cat # 11330-032) containing 2% FAF BSA, 50ng/ml FGF7 (PcproTech; Cat# 100-19), and 250nM cyclopamine-KAAD (Calbiochem; Cat # 239804).
Step 3 of the differentiation protocol was earned out over seven days. Cells were fed daily by aspirating medium from each well and replacing with a fresh aliquot (0.5ml) of DMEM-high glucose (Invitrogen; Cat # 10569) supplemented with 0.1% Aibumax (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 μΜ all-trans retinoic acid (RA) (Sigma-Aldrich; Cat # R2625). At the conclusion of the third step of differentiation, 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 Pdx I, and Cdx2.
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 (0.5ml) of DMEM-high glucose supplemented with 0.1% Aibumax, 0.5x Insulin-Transferrin-Sclcnium, 100 ng/ml Noggin, and ΙμΜ Aik 5 inhibitor (Axxora; Cat # ALX-270-445). At the conclusion of the fourth step of differentiation, 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 Pdx 1.
Step 5 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 0.1% Albumax, 0.5x Insulin-Transferrin-Selenium, and ΙμΜ Aik 5 inhibitor. Medium in each well was aspirated and replaced with a fresh aliquot (0.5ml) on all days. At the conclusion of the fifth step of differentiation, ceils 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. 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 IgG 1K Isotype control antibody for both PE and APC was used to gate percent positive cells. RT-PCR Analysis: RNA samples were purified by binding to a silica-gel membrane (Rneasy Mini Kit, Qiagen, CA) 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 AB1 (AB1, CA) high capacity cDNA archive kit.
Unless otherwise stated, all reagents were purchased from Applied Biosystcms. Realtime 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 μΙ. Each cDNA sample was run in duplicate to correct for pipetting errors. Primers and FAM-Iabeled TAQMAN(R)probes w'ere 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. 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 dcnaturation 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. Briefly, for each cDNA sample, the endogenous control Ct value was subtracted from the gene of interest Ct to give the delta Ct value (ACt). The normalized amount of target was calculated as 2-ACt, assuming amplification to be 100% efficiency. Final data were expressed relative to a calibrator sample.
High Content Analysis: At the conclusion of culture, assay plates were washed once with PBS (lnvitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-0 i 1) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (lnvitrogen; Cat # 16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems: Cat # AF i 924) was diluted 1:100 in 4% chicken serum and added to each well for two hours at room temperature. After washing three times with PBS, Alcxa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; lnvitrogen; Cat # A21467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5pg/ml Hocchst 33342 (lnvitrogen; Cat # H3570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in I00ul/wcll 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 antihuman glucagon (Sigma-Aldrich; Cat # G2654). Secondary antibodies used for analysis included 1:1000 dilution Alcxa Fluor 647 chicken anti-rabbit IgG (lnvitrogen; Cat # A21443), and 1:1000 dilution Alexa Fluor 488 chicken anti-mouse IgG (lnvitrogen; Cat # A21200).
Imaging was performed using an IN Ceil Analyzer 1000 (GE Healthcare) utilizing the 51008bs dichroic for cells stained with Hoechst 33342 and Alexa Fluor 4X8. 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 grayscale 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. PC'R 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.
Figure 20, panel A shows FACS analysis for the definitive endodemn marker, CXCR4, after the first step of differentiation. Treatment of human embryonic stem cells with GDF-8 and Wni3a 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 IX BIO) also yielded an equivalent or slightly higher percentage of CXC4 positive cells. Figure 20, panel B shows high content image analysis for normalized SOX 17 protein expression in human embryonic stem cells after three days differentiation to definitive endoderm. In some cases, treatment with GDF-8 resulted in a lower cell number at the conclusion of the first step of differentiation. However, GDF-8 treatment in combination with Wnt3a or with the small molecule inhibitors clearly induced expression of SOX i 7, a marker of definitive endoderm. In one instance, treatment with GDF-8 and Compound 40 yielded cell numbers in culture and SOX 17 expression equivalent to treatment with activin A and Wnt3a.
Figure 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. In Figure 20, panels D-F. normalized protein levels of insulin and glucagon arc shown, along with their respective ratio for each treatment group. Similar levels of insulin and glucagon could be obtained with each of the GDF-8 treatments relative to treatment with activin A and Wnt3a, demonstrating that GDF-8, in combination with Wnt3a or a small molecule, can substitute for activin A during definitive endoderm differentiation and subsequent pancreatic endoderm and endocrine differentiation.
Example 17
Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage that were Formed Using GDF-8 and a Compound of the Present Invention are able to Further Differentiate into Cells Expressing Markers Characteristic of the
Pancreatic Endocrine Lineage
Additional small molecules were tested in combination with GDF-8 and activin A for definitive endoderm differentiation. These included a commercial inhibitor of GSK.3 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.
Preparation of cells for assay: Stock cultures of human embryonic stem cells (Η I human embryonic stem cell line) were maintained in an undifferentiated, pluripotent state on reduced growth factor MATRJGEL™ (BD Biosciences; Cat # 356231 )-coated dishes in MEF conditioned medium supplemented with 8ng/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 I mg/ml dispasc (Invitrogcn; 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 8ng/ml bFGF and plated onto reduced growth factor MATRIGELlvl-coatcd 24-well, black wall culture plates (Arctic White: Cat # AWLS-303012) in volumes of 0.5ml/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% C02 throughout assay.
Assay: The assay was initiated by aspirating the culture medium from each well and adding back an aliquot (0.5ml) 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, lOOng/ml activin A (PeproTcch; Cat #120-14) or lOOng/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). In some samples, 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF) was also included. On the second day of assay, lOOng/ml activin A or lOOng/ml GDF-8 was diluted into RPMI-1640 medium supplemented with 2% FAF BSA, omitting Wnt3a from all samples. In some test samples using GDF-8, Wnt3a was replaced with a given concentration of small molecule compound, added only on the first day of definitive endoderm differentiation. These small molecules included: Compound 181 (1.25μΜ in assay). Compound 180 (2.5μΜ in assay), Compound 19 (!0μΜ in assay), Compound 202 (2.5μΜ in assay), Compound 40 (5μΜ in assay). Compound 34 (2.5μΜ in assay ). Compound 206 (2.5μΜ in assay), and a commercially available GSK.3 inhibitor IX BIO (10μΜ in assay) (EMD Chemicals, Inc.; Cat # 361550). At the conclusion of the first step of differentiation, cells from some wells were harvested for flow cytometry analysis to evaluate levels of CXCR4, a marker of definitive endoderm formation. Additional wells were harvested for RT-PCR analysis to measure other markers of differentiation.
At the conclusion of the first step of definitive endoderm differentiation, replicate sets of parallel wells from each treatment group were subjected to further step-wise differentiation. It is important to note that after the first differentiation step, all wells undergoing subsequent culture and differentiation received the same treatment. The protocol for this continuing differentiation is described below.
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 (0.5ml) of DMEM:F12 medium (Invitrogen; Cat # 11330-032) containing 2% FAF BSA, 50ng/ml FGF7 (PcproTech; Cat# 100-19), and 250nM 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 (0.5ml) 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 μΜ all-trans retinoic acid (RA) (Sigma-Aldrich; Cat # R2625). At the conclusion of the third step of differentiation, cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation.
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 (0.5ml) of DMEM-high glucose supplemented with 0.1% Albumax, 0.5x Insulin-Transferrin-Sclenium, 100 ng/ml Noggin, and ΙμΜ Aik 5 inhibitor (Axxora; Cat # ALX-270-445). At the conclusion of the fourth step of differentiation, cells from some wells were harvested for analysis by RT-PCR to measure markers of differentiation.
Step 5 of the differentiation protocol was carried out over seven days in DMEM-high glucose with 0.1% Albumax, 0.5x Insulin-Transferrin-Selenium, and ΙμΜ Aik 5 inhibitor. Medium in each well was aspirated and replaced with a fresh aliquot (0.5ml) on all days. At the conclusion of the fifth step of differentiation, cells from some wells were harvested for analysis by RT-PC'R 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# FAB i 73A) for 30 minutes at 4°C. After a scries 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 IgG 1K Isotype control antibody for both PE and APC was used to gate percent positive cells. RT-PC'R Analysis: RNA samples were purified by binding to a silica-gel membrane (Rncasy Mini Kit, Qiagcn, CA) 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-frcc 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.
Unless otherwise stated, all reagents were purchased from Applied Biosystems. Realtime 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 μ!. Each cDNA sample was run in duplicate to correct for pipetting errors. Primers and FAM-labcled TAQMAN(R)probcs were used at concentrations of 200 nM. The level of expression for each target gene was normalized using a human glyccraldchyde-3-phosphate dehydrogenase (GAPDH) endogenous control previously developed by Applied Biosystems. 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(R>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. Briefly, for each cDNA sample, the endogenous control Ct value was subtracted from the gene of interest Ct to give the delta Ct value (ACt). The normalized amount of target was calculated as 2-ACt, assuming amplification to be 100% efficiency. Final data were expressed relative to a calibrator sample.
High Content Analysis: At the conclusion of culture, assay plates were washed once with PBS (Invitrogcn; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogcn; Cat # 16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # API924) was diluted 1:100 in 4% chicken serum and added to each well for two hours at room temperature. After washing three times with PBS, Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat IgG; Invitrogen; Cat # A21467) diluted 1:200 in PBS was added to each well. To counterstain nuclei, 5gg/ml Hoechst 33342 (Invitrogen; Cat # F13570) was added for fifteen minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/well PBS for imaging. Other primary antibodies used for analysis included 1:100 dilution mouse anti-human CDX2 (Invitrogen; Cat # 397800), 1:100 dilution goat anti-human Pdxl (Santa Cruz Biotechnology; Cat # SC-14664), 1:200 dilution rabbit anti-human insulin (Cell Signaling; Cat # C27C9), and 1:1500 dilution mouse anti-human glucagon (Sigma-Aldrich; Cat # G2654). 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 grayscale 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.
Resit I is: Results for representative differentiation markers are shown in Figure 21 and Table 15 for cells harvested from each step of differentiation. In Figure 21A and B. flow cytometric results for CXCR4 are shown for various treatments during the first step of definitive endoderm differentiation. Figure 21A shows the effects on CXCR4 expression from treatment with various compounds in combination with activin A . Figure 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.
In Figures 21 C and 21D, normalized RT-PC’R 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 (Figure 210 or in combination with GDF-8 (Figure 21D). Similar normalized RT-PC'R values were evaluated at the conclusion of step three of the differentiation protocol (Figures 21E and 21F) and at the end of step four of the differentiation protocol (Figures 21G and 21H) and at the end of step 5 of the differentiation protocol (Figures 211 and 21J). Treatments during differentiation step 1, which combined a compound of the present invention with GDF-8, had improved expression of various endoderm and pancreatic markers relative to GDF-8 treatment alone (Figures 21 F and 21H and 21J ). Treatments combining compounds of the present invention with activin A had minimal or no improvement in expression markers relative to treatment with activin A alone or activin A with Wnt3a (Figures 21E, and 21G and 211). Table 15 summarizes comparative CT values for additional gene markers at the end of each differentiation step, comparing treatments during step one that combined activin A or GDF-8 with or without a compound of the present invention. At the conclusion of step five of differentiation, high content analysis was performed to measure cell numbers (Figure 21K and 21M) and protein expression of insulin and glucagon (Figures 21L and 21N ). Treatment with GDF-8 during the first step of differentiation, alone or in combination with a compound of the present invention, resulted in insulin and glucagon expression at the conclusion of step five of differentiation, demonstrating that GDF-8 was able to substitute for activin A during the initiation of definitive endoderm formation and subsequently led to pancreatic hormonal cells. Collectively, these data show that addition of any of the respective small molecules had minimal effects on differentiation markers for treatments in combination with activin A. However, addition of a small molecule in combination with GDF8 treatment had significant improved effects on immediate definitive endoderm differentiation at the conclusion of step 1 differentiation and also on downstream differentiation markers at the conclusion of steps 3, 4, and 5. Variability was observed within the panel of small molecules, perhaps attributable to the concentration of compound used in assay and/or mechanism of action.
Example 18
Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage that were Formed Using GDF-8 and a Compound of the Present Invention are able to Release C-Peptide Following Transplantation into a Rodent
It was important to determine whether cells expressing markers characteristic of the pancreatic endoderm lineage generated in vitro by treatment with GDF-8 and a small molecule could produce functional endocrine cells in vivo. An in vivo transplant study was done to compare cells differentiated by treatment with activin A and Wnt3a versus treatment with GDF-8 and small molecule compounds.
Preparation of cells: Clusters of Fll human embryonic stem cells were grown on reduced growth factor MATRIGELI M(Invitrogen; Cat # 356231) -coated tissue culture plastic with passage on average every four days. MEF conditioned medium supplemented with 8ng/ml bFGF was used for initial seeding and expansion. All human ES cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma contamination.
Cell passage was performed by exposing cell cultures to a solution of 1 mg/ml dispasc (Invitrogcn; 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 8ng/ml bFGF (PcproTcch Inc.; Cat # I00-18B) for seeding on reduced growth factor MATRJGEL (BD Biosciences; Cat # 356231 )-coated 6-well plates (Nunc; Cat# 140685) at a 1:3 ratio using volumes of2.5ml/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% CO2 throughout the time in culture.
Cell differentiation: The differentiation process was started three days after the cells were seeded onto 6-well plates coated with reduced growth factor MATRIGEL1 M. A four-step protocol was used for the in vitro differentiation of HI human embryonic stem cells to cells expressing markers characteristic of the pancreatic endoderm lineage. Step I was conducted over three days to generate definitive endoderm cells. On the first day of step 1, 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 8ng/m! bFGF. In one treatment group, cells were exposed to lOOng/ml activin A (PeproTech; Cat #120-14) with 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF). In a second treatment group, cells were exposed to 100 ng/ml GDF-8 (R&D Systems; Cat # 788-G8) with 2.5 μΜ Compound 40. In a third treatment group, cells were exposed to 100 ng/ml GDF-8 (R&D Systems; Cat # 788-G8) with 2.5 μΜ Compound 202. On the second and third day of step I of differentiation, cells in all treatment groups were fed with RPMI-1640 containing 2% FAF BSA, 8ng/ml bFGF and cither lOOng/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. At the end of the third day of culture, one well from each treatment group was collected for FACS analysis.
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/mi FGF7 (PcproTcch; 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 μΜ 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 ΙμΜ ALK.5 inhibitor (Axxora; Cat # ALX-270-445 ). On the third day, cells were lifted from the substratum by using a 20 μΐ tip (Ramin; Cat # RT-LI0F) and a cell scraper (Coming; 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 ΙμΜ ALK5 inhibitor, then cultured overnight in six-well Costar Ultra Low Attachment Microplates (Coming Inc., Cat # 3471). On the following day, cells in suspension culture were collected and counted. Aliquots of 10x10'’ cells/ mouse were used for transplantation. Aliquots of 0.5x 106 cells were collected for RT-PCR analysis.
Figure 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.
Results for RT-PCR analysis for cells from each treatment group at the conclusion of step 4 of the differentiation protocol arc shown in Figure 22, panel B. Cells differentiated to pancreatic endoderm (PE) using Activin A and Wnt3a or using GDF-S and Compound 40 or using GDF-8 and Compound 202 expressed equivalent levels of PE markers: CDX2, MAFA, NGN3, NKX6.1, PDX1 and Ptfl alpha. These results suggest that the differentiation protocol utilizing GDF-8 and a small molecule was equally effective in creating a pancreatic endoderm precursor population of cells.
Transplantation of Human Embryonic Stem Cells Treated According to the Methods of the Present Invention into Mice: Five to six-week-old male scid-beige mice (C.B-!gh-! bGbmsTac-Prkdccu,-L\wth' 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 w ith a mixture of isolfluranc 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 x %” 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. During preoperative preparation of the mice, cells for transplant were centrifuged in a 1.5 mL microfugc 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 w'as 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 scaled 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.
Following transplantation, 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-pcptide assay was performed. Human C-pcptide levels were determined using the Mercodia/ALPCO Diagnoses Ultrasensitive C-peptide ELISA according to the manufacturer’s instructions. ELISA results for human C-peptide are shown in Figure 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.
Example 19
Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage that were Formed Using GDF-8 are able to Release C-Peptide Following Transplantation into a Rodent
It was important to demonstrate that cells differentiated with GDF-8 in the absence of activin A could also be further differentiated to an endocrine cell population capable of secreting human C-peptide in an in vivo rodent transplant model.
Preparation of cells: Clusters of HI human embryonic stem cells were grown on reduced growth factor MATRIGEL™ (lnvitrogen; Cat # 356231) -coated tissue culture plastic with passage on average every four days. MEF conditioned medium supplemented with 8ng/ml bFGF was used for initial seeding and expansion. All human ES cell lines were maintained at passage numbers less than 50 and routinely evaluated for normal karyotype and absence of mycoplasma contamination.
Cell passage was performed by exposing cell cultures to a solution of 1 mg/ml dispase (lnvitrogen; 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 8ng/ml bFGF (PeproTech Inc.; Cat # 100-18B) for seeding on reduced growth factor MATR1GELIM (BD Biosciences; Cat # 356231 )-coated 6-well plates (Nunc; Cat# 140685) at a 1:3 ratio using volumes of 2.5ml/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% COv throughout culture.
Cell differentiation: The differentiation process was started three days after the cells were seeded into 6-well plates. A four-step protocol was used for the in vitro differentiation of Η1 human embryonic stem cells to cells expressing markers characteristic of the pancreatic endoderm lineage. Step 1 was conducted over three days to generate cells expressing markers characteristic of the definitive endoderm lineage. On the first day of step 1, differentiation was initiated by aspirating spent culture medium and adding an equal volume of RPMI-1640 basal medium (lnvitrogen* Cat # 22400) with 2% Albumin Bovine Fraction V. Fatty Acid Free (FAF BSA) (Proliant Biologicals; Cat # SKU 68700) and 8ng/ml bFGF. In one treatment group, duplicate sets of cells were treated with 100 ng/ml GDF-8 (R&D Systems; Cat # 788-G8) and 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF). In a second treatment group, duplicate sets of cells were treated with 100 ng/ml GDF-8 and 2.5 μΜ Compound 40. On the second and third day of step 1 differentiation, cells in all treatment groups were fed with RPMI-1640 containing 2% FAF BSA, 8ng/ml bFGF and 100 ng/ml GDF-8 but without the addition of Wnt3a or Compound 40. At the end of the third day of culture, 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 (PeproTcch: 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 μΜ 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 ΙμΜ ALK5 inhibitor (Axxora; Cat # ALX-270-445), and 100 ng/ml GDF-8 (R&D Systems; Cat # 788-G8) during the first two days. On the third day of step 4, ceils were harvested from the 6-well plates using a 20 μΐ tip (Rainin; Cat # RT-L1 OF) and a cell scraper (Coming; Cat # 3008) and transferred to a 50 ml tube. 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 ΙμΜ ALK5 inhibitor, then cultured overnight in six-well Costar Ultra Low Attachment Microplatcs (Corning Inc., Cat # 3471). On the following day, cells in suspension culture were collected and counted. Aliquots of 10x10' cells/mouse were used for transplantation. Aliquots of 0.5x10'cells were collected for RT-PCR analysis.
Figure 24A shows flow cytometric results for definitive endodeim 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 arc shown in
Figure 24B. Cells differentiated to pancreatic endoderm (PE) using GDF-8 and Wnt3a or GDF-8 and Compound 40 expressed equivalent levels of markers characteristic of the pancreatic endoderm lineage, such as:CDX2, MafA, Ngn3, NKX6.1, Pdx-1 and PtflA. These results demonstrate that the differentiation protocol utilizing GDF-8 and Wnt3a or GDF-8 and a compound of the present invention was effective in creating a pancreatic endoderm precursor population of cells. The differentiation protocol was conducted in two independent but identical treatment sets. Results from duplicate treatment sets were in strong agreement as shown by RT-PCR analysis.
Human embryonic stem cel/ Transplantation into Mice: Five to six-week-old male scid-beige mice {C.B-Igh-1 b/GbmsT'dc-Prkc/c11*1 -Lyst1* 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 w'as determined using a hand held glucometcr (LifeScan; One Touch ). On the day of surgery, mice were anesthetized with a mixture of isolfluranc and oxygen, and the surgical site was shaved with small animal clippers. Mice were dosed with 0.1 mg.kg Buprenex subcutaneously pre-operativeiy. 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 x 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. During preoperative preparation of the mice, cells for transplant were centrifuged in a 1.5 mL microfugc 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, 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-opcratively.
Following transplantation, 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 Diagnoses Ultrasensitive C-peptide ELISA according to the manufacturer’s instructions. ELISA results for human C-peptide are shown in Figure 29C and D for 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.
Example 20
Evaluation of the Potential of Inhibitors of CDK, GSK3, and TRK to Differentiate Human Embryonic Stem Cells into Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage A subset of 14 proprietary small molecules, known to have specificity tor the CDK, GSK3, and/or TRK signaling pathways were evaluated for their potential to differentiate human embryonic stem cells to ceils expressing markers characteristic of the definitive endoderm lineage.
Cell assay seeding: Briefly, clusters of Η1 human embryonic stem cel is were grown on reduced growth factor Matrigel1V1 (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 MATRJGELIM (BD Biosciences; Cat # 356231 )-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat # 6005182) using volumes of 100 μΐ/well. 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 Hng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C, 5% C02 in a humidified box throughout the duration of assay.
Preparation of compounds and assay: Screening was conducted using the compounds described in Table 16. In addition Compound 34 was included as a positive control, as demonstrated in previous examples. Compounds were made available as 5 mM stocks in 96-wcll 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 50mM F1EPES (lnvitrogen; Cat # 15630-080), 20% DMSO and stored at 4°C. Test conditions were performed in triplicate, feeding on alternating days over a four-day assay period. Assay was initiated by aspirating culture medium from each well followed by three washes in PBS (lnvitrogen; Cat # 14190) to remove residual growth factors. On the first day of assay, test volumes of 200μΙ per well were added to each well using DMEM:F!2 base medium (lnvitrogen; 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 2.5μΜ compound. A parallel set of test samples were treated in an identical manner but omitting GDF-8 from the medium. On the third day of assay, test volumes of 100μ1 per well were added to each well using DM EM :F 12 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 andlOOng/m! recombinant human aetivin A (PeproTech; Cat #120-14) throughout the four day assay along with Wnt3a (20ng/ml) addition on days 1 and 2. Negative control samples contained DMEM:F12 base medium supplemented with FCS.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS (lnvitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken serum (Invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX 17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken scrum and added to each well for one hour at room temperature. 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. To counterstain nuclei, 4pg/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 SOX 17 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 SOX 17 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 arc shown in Table 17. None of the small molecules induced significant SOX 17 expression in the absence of GDF-8 during the four day differentiation process. Compound 34 served as an experimental control and induced significant SOX 17 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.
Example 21
Screening for Analogues of the Compounds of the Present Invention that are Capable of Mediating the Formation of Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage
Based on the structures for the compounds of the present invention, 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.
Cell assay seeding: Briefly, clusters of Η1 human embryonic stem cells were grown on reduced growth factor MatrigelLV1 (lnvitrogen; Cat # 356231) -coated tissue culture plastic. Cells were passaged using collagenase (lnvitrogen; 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 MATRIGELIM (BD Biosciences; Cat # 35623l)-coated 96-well black plates (Packard ViewPlates; PerkinElmer; Cat #6005182) using volumes of 100 μΙ/well. 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 8ng/ml bFGF (R&D Systems; Cat # 233-FB). Plates were maintained at 37°C\ 5% CO2 in a humidified box throughout the duration of assay.
Preparation of compounds and assay: Screening was conducted using a library of the analogue compounds. Compounds from this library wrerc made available as 5 mM stocks in 96-wcll 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 50mM HEPES (lnvitrogen; Cat # 15630-080), 20% DMSO and stored at 4°C. Test conditions were performed in triplicate, feeding on alternating days over a four-day assay period. Assays were 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 200μΙ per well were added to each well using DMEM:F12 base medium (Invitrogen; Cat # 11330-032) supplemented with 0.5% FCS (HyClonc; Cat # SH30070.03) and 200 ng/ml GDF-8 (R&D Systems, Cat # 788-G8) plus 2.5μΜ compound. On the third day of assay, test volumes of ΙΟΟμΙ 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 andlOOng/ml recombinant human activin A (PcproTcch; Cat #120-14) throughout the four-day assay along with Wnt3a (20ng/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.
High Content Analysis: At the conclusion of four-days of culture, assay plates were washed twice with PBS (Invitrogen; Cat # 14190), fixed with 4% paraformaldehyde (Alexis Biochemical; Cat # ALX-350-011) at room temperature for 20 minutes, then washed three times with PBS and permeabilized with 0.5% Triton X-100 (Sigma; Cat # T8760-2) for 20 minutes at room temperature. Cells were washed again three times with PBS and blocked with 4% chicken scrum (invitrogen; Cat #16110082) in PBS for 30 minutes at room temperature. Primary antibody (goat anti-human SOX17; R&D Systems; Cat # AF1924) was diluted 1:100 in 4% chicken serum and added to each well for one hour at room temperature. Alexa Fluor 488 conjugated secondary antibody (chicken anti-goat lgG; Molecular Probes; Cat # AZ1467) was diluted 1:200 in PBS and added to each sample well after washing three times with PBS. To counterstain nuclei, 4pg/ml Hoechst 33342 (Invitrogen; Cat # H3570) was added for ten minutes at room temperature. Plates were washed once with PBS and left in ΙΟΟμΙ/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 w'dls 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 S0X17 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 SOX 17 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.
Screening results are shown in Table 18 from four assay plates in this single experiment. Compounds arc ranked with respect to SOX 17 expression as a percentage of the positive control treatment with activin A and Wnt3a. This assay identified a list of 12 new analogue hits as shown in Table 19.
Example 22
Human Embryonic Stem Cells Grown on Microcarriers can be Differentiated into Cells Expressing Markers Characteristic of the Definitive Endoderm Lineage According to the Methods of the Present Invention
For purposes of differentiation and production of large numbers of endocrine cells under scalable conditions, it was important to show that human embryonic stem cells could be grown and differentiated to definitive endoderm on microcarricr beads using the methods of the present invention.
Preparation of cells for assay and differentiation: HI p49C3 cells were routinely grown on C’ytodex3 beads (GE Healthcare Life Sciences, NJ) in a 125ml spinner flask, according to the methods described in US 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 30cm2 bead surface area per well, and the plate was placed on a rocking platform. Cells on beads in the positive control treatment well (designated AA/Wnt3a) were differentiated with addition of lOOng/ml activin A (PcproTech; Cat #120-14) and 20t»g/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF) for two days followed by lOGng/ml activin A and 8ng/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 2ml/well. Compound 34, at a Final concentration of 2.5μΜ was added to a negative control treatment well (designated CMP alone) in RPMI-1640 with 2% Fatty Acid Free BSA (2ml/wcll) for three days in the absence of any other growth factor treatment. A third treatment well (designated CMP+8) received Compound 34at 2.5μΜ plus 50ng/ml GDF-8 (R&D Systems, Cat # 788-G8) in RPMI-1640 with 2% Fatty Acid Free BSA (2mi/well) for three days. A fourth treatment well (designated CMP+ 8+D) received Compound 34at 2.5μΜ with 50ng/ml GDF-8 and 50ng/ml PDGF-D in RPMI-1640 with 2%
Fatty Acid Free BSA (2ml/wcll) for three days. A fifth treatment well (designated CMP+ 8+D+V) received Compound 34at 2.5μΜ with 50ng/ml GDF-8, 50ng/ml PDGF-D, and 50ng/ml VEGF in RPMI-1640 with 2% Fatty Acid Free BSA (2ml/wcll) for three days. A sixth treatment well (designated CMP+ 8+D+V+M) received Compound 34at 2.5μΜ with 50ng/ml GDF-8, 50ng/ml PDGF-D, 50ng/ml VEGF, and 20ng/ml Muscimol in RPMI-1640 with 2% Fatty Acid Free BSA (2ml/wcll) for three days. All media and treatments were exchanged daily.
At the conclusion of treatment and culture, cells were harvested from the beads, according to the methods described in US Patent Application No. 61/116,447. The harvested cells were counted and analyzed by flow cytometry, according to the methods described above.
Results arc shown in Figure 25. As show n in panel A, similar numbers of cells were recovered tor all treatment groups undergoing differentiation. As shown in panel B, cells treated with the Compound 34alone 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 34added with the various growth factor combinations induced CXCR4 expression in 50% of the cells, on average. Of note, 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.
Example 23
The Compounds of the Present Invention, Together with GDF-8 Enhance Cell
Proliferation A previous example showed that 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.
Preparation of cells for assay: Stock cultures of human embryonic stem cells (Η 1 human embryonic stem cell line) were maintained in an undifferentiated, pluripotcnt state on reduced growth factor MATRIGEL1 M (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 I 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 8ng/ml bFGF and seeded onto reduced growth factor MATRIGEL™-coated 96-well Packard VIEWPLATES (PerkinElmer; Cat # 6005182) in volumes of ΙΟΟμΙ/well. MEF conditioned medium supplemented with 8ng/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? in a humidified box throughout the duration of assay.
Assay: The assay was initiated by aspirating the culture medium from each well and adding back a final aliquot (1 ΟΟμΙ) 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.
On the first day of assay, all wells containing cells received an aliquot (80μΙ) of RPM1-1640 medium (Invitrogen; Cat #: 22400) supplemented with 2.5% Albumin Bovine Fraction V, Fatty Acid Free (FAF BSA; 2% in final assay) (Proliant Inc. Cat #: SKU 68700). Various control and test samples were created at 5x concentration to be added to appropriate wells (20μ1 per well ). Control conditions included the follow ing, with final growth factor concentrations as indicated: 1) basal medium with 2% FAF BSA; 2) lOOng/ml activin A (PeproTech; Cat #120-14) with 8ng/ml bFGF (PcproTcch; Cat # 100-I8B); 3) lOOng/ml activin A with 8ng/ml bFGF and 20ng/ml Wnt3a (R&D Systems; Cat # I324-WN/CF); 4) lOOng/ml GDF-8 (R&D Systems, Cat # 788-G8) with 8ng/ml bFGF; 5) GDF-8 with 8ng/ml bFGF and 20ng/ml Wnt3a.
Cells in an additional set of control wells were treated with MEF conditioned medium throughout the assay. In some control samples using GDF-8, Wnt3a was replaced with a compound of the present invention. For experimental test samples, eight different compounds were diluted two-fold in series to create three different dose concentrations then combined with lOOng/ml GDF-8 and 8ng/ml bFGF. These small molecules included proprietary compounds Compound 181, Compound 180, Compound 19, Compound 202, Compound 40, Compound 34. Compound 56, and a commercially available GSK.3 inhibitor BIO (EMD Chemicals, Inc.; Cat # 361550). On the second and third day of assay, all wells for control and experimental samples were aspirated and fed again using identical treatment conditions except that Wnt3a was removed from some control wells. 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, 20μ1 of MTS was added to each well, and assay plates were incubated at 37°C, 5% C02 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. At a single time point, 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. Figure 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. In contrast, cells cultured in basal medium without growth factors (no treatment), showed a steady decline in OD490 correlated with a loss in cell number over time. Activin A treatments during the differentiation process, with and without Wnt3a, showed incremental increases in OD490, indicating significant expansion of the cell population over time. GDF-8 treatment in the absence of Wnt3a resulted in a decrease in OD490 relative to activin A treatment; this w as noticeable on the first day and sustained throughout all three days of culture. Addition of Wnt3a to the GDF-8 treatment group resulted in a recovery and increase in OD490 by the third day of culture.
Figures 26, panel B through Figure 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. In all cases, 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.
Example 24
Human Embryonic Stem Cells Grown on Microcarriers can be Differentiated into Endocrine Progenitor Cells According to the Methods of the Present
Invention
For purposes of differentiation and production of large numbers of endocrine cells under industrial conditions, it was important to show that human embryonic stem cells could be grown and differentiated to endocrine progenitor cells on microcarrier beads using a protocol without activin A.
Preparation of cells for assay and differentiation: Η 1 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 I 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 (designated AA+Wnt) were differentiated with addition of lOOng/ml activin A (PcproTech; Cat #120-14), 8ng/ml bFGF (PeproTcch Inc.; Cat #: 100-1 SB), and 20ng/ml Wnt3a (R&D Systems; Cat # 1324-WN/CF) for one day followed by lOOng/ml activin A and 8ng/ml bFGF (PeproTcch Inc.; Cat#: 100-18B) for two days in RPM1-1640 (Invitrogen; Cat#: 22400) with 2% Fatty Acid Free BSA (Proliant Biomcdicals, Inc; SKU # 68700) using volumes of 2ml/wcll. A second treatment well (designated GDF-8+MCX) received Compound 202 at 2.5μΜ plus 200ng/ml GDF-8 (R&D Systems, Cat # 788-G8) and 8ng/ml bFGF for one day followed by two days with 200ng/ml GDF-8 and 8ng/ml bFGF in RPMI-1640 with 2% Fatty Acid Free BSA (2ml/wcll) media. A third treatment well (designated GDF-8+Wnt) received 200ng/ml GDF-8 with 20ng/ml Wnt3a and 8ng/ml bFGF for one day followed by two days with 200ng/ml GDF-8 and 8ng/ml bFGF in RPMI-1640 with 2% Fatty Acid Free BSA (2ml/wcll) media. All media and treatments were exchanged daily.
At the conclusion of treatment and culture, cells were harvested and counted to determine cell recovery and undergo flow cytometric analysis. High levels of CXCR4 and CD99 was seen following all three treatment regiments (Figure 27A). Cell number varied between samples (Figure 27B). A lower cell number was observed in samples treated with GDF-8 and at the definitive endoderm and fourth stage than the other treatment groups. This suggests that the compounds of the present invention may increase proliferation of the cells during differentiation.
At the end of stage 3 the cndodcrmal genes PDX1, HNF4 alpha, and CDX2 arc expressed in the cells (Figure 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 Pdx 1 than the control differentiation treatment. At the end of stage 4, endodermal genes were up regulated further (Figure 27E, F). These results conclude that GDF-8 plus Compound 202 can replace activin A and Wnt3a for definitive endoderm differentiation resulting in pancreatic endoderm formation.
Publications cited throughout this document are hereby incorporated by reference in their entirety. Although the various aspects of the invention have been illustrated above by reference to examples and preferred embodiments, it will be appreciated that the scope of the invention is defined not by the foregoing description but by the following claims properly construed under the principles of patent law.
ΒΙΕ Η

Claims (21)

  1. What is claimed is:
    1. A method to differentiate pluripotent stem cells into definitive endoderm cells comprising culturing the pluripotent stem cells with a medium supplemented with activin A and a cyclic aniline-pyridinotriazine.
  2. 2. The method of claim 1, wherein the cyclic aniline-pyridinotriazine is selected from the group consisting of 14-Prop-2-en-l-yl-3,5,7,14,17,23,27- heptaazatetracyclo[ 19.3.1.1-2,6-.1-8,12~]heptacosa-1 (25),2(27),3,5,8(26),9,11,21,23-nonaen-16-one, 14-Methyl-3,5,7,14,18,24,28- heptaazatetracyclo[20.3.1.1-2,6-. l~8,12~]octacosa-l(26),2(28),3,5,8(27),9,11,22,24-nonaen-17-one, and 5-Chloro-l,8,10,12,16,22,26,32-octaazapentacyclo[24.2.2.1-3,7-. 1-9,13-. 1-14,18~]tritriaconta-3(33),4,6,9(32), 10,12,14(31), 15,17-nonaen-23-one.
  3. 3. The method of claim 1 or claim 2, wherein the medium is further supplemented with wnt3A.
  4. 4. The method of claim 3, wherein the medium is serum-free.
  5. 5. The method of claim 2, the cyclic aniline-pyridinotriazine is 14-Prop-2-en-l-yl- 3,5,7,14,17,23,27-heptaazatetracyclo[19.3.1.1~2,6~.l~8,12~]heptacosa- 1 (25),2(27),3,5,8(26),9,11,21,23-nonaen-16-one.
  6. 6. The method of claim 2, the cyclic aniline-pyridinotriazine is Id-Methyls',7,14,18,24,28-heptaazatetracyclo[20.3.1.1-2,6~. 1-8,12~]octacosa- 1 (26),2(28),3,5,8(27),9,11,22,24-nonaen-17-one.
  7. 7. The method of claim 2, wherein the cyclic aniline-pyridinotriazine is 5-Chloro- l,8,10,12,16,22,26,32-octaazapentacyclo[24.2.2.1~3,7~.l~9,13~.l~14,18~]tritriaconta-3(33),4,6,9(32), 10,12,14(31),15,17-nonaen-23-one.
  8. 8. The method of claim 7, wherein the medium is supplemented with activin A, Wnt3a, Chloro-1,8,10,12,16,22,26,32- octaazapentacyclo[24.2.2.1-3,7-. 1-9,13~.l~14,18~]tritriaconta-3(33),4,6,9(32),10,12,14(31),15,17-nonaen-23-one, and EGF.
  9. 9. The method of claim 7, wherein the medium is supplemented with activin A, Wnt3a, Chloro-1,8,10,12,16,22,26,32- octaazapentacyclo[24.2.2.1-3,7~. 1-9,13-. 1-14,18~]tritriaconta-3(33),4,6,9(32),10,12,14(31),15,17-nonaen-23-one, and FGF-4.
  10. 10. The method of claim 7 wherein the medium is supplemented with activin A, Wnt3a, Chloro-1,8,10,12,16,22,26,32- octaazapentacyclo[24.2.2.1~3,7~.l~9,13~.l~14,18~]tritriaconta-3(33),4,6,9(32),10,12,14(31),15,17-nonaen-23-one, EGF, and FGF-4.
  11. 11. The method of claim 10, wherein the medium is further supplemented with PDGF-AB.
  12. 12. The method of claim 11, wherein the medium is further supplemented with VEGF.
  13. 13. The method of claim 11, wherein the medium is further supplemented with PDGF-8 and muscimol.
  14. 14. The method of claim 7, wherein the medium is supplemented with activin A, Wnt3a, Chloro-1,8,10,12,16,22,26,32- octaazapentacyclo[24.2.2.1~3,7~.l~9,13~.l~14,18~]tritriaconta- 3(33),4,6,9(32),10,12,14(31),15,17-nonaen-23-one, EGF, FGF-4, PDFG-A, VEGF, PDGF, muscimol, and GDF-8.
  15. 15. The method of claim 1 or claim 2, wherein the pluripotent stem cells are human pluripotent stem cells.
  16. 16. The method of claim 15, wherein the human pluripotent stem cells are human embryonic stem cells.
  17. 17. The method of claim 3, further comprising differentiating the definitive endoderm cells into pancreatic endoderm cells.
  18. 18. The method of claim 17, further comprising differentiating the pancreatic endoderm cells into pancreatic endocrine cells.
  19. 19. The method of claim 3, further comprising differentiating the definitive endoderm cells into cells expressing markers characteristic of the pancreatic endoderm lineage, wherein the markers characteristic of the pancreatic endoderm lineage are selected from the group consisting of PDX1, HNF-1 beta, PTF1 alpha, HNF6, HB9 and PROX1.
  20. 20. The method of claim 19, further comprising differentiating the cells expressing markers characteristic of the pancreatic endoderm lineage into cells expressing markers characteristic of the pancreatic endocrine lineage, wherein the markers characteristic of the pancreatic endocrine lineage are selected from the group consisting ofNGN3, NEUROD, ISL1, PDX1, NKX6.1, PAX4, and PTF-1 alpha.
  21. 21. Definitive endoderm cells when prepared according to the method of any one of claims 1 to 20.
AU2018201993A 2008-06-30 2018-03-21 Differentiation of pluripotent stem cells Active AU2018201993B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2018201993A AU2018201993B2 (en) 2008-06-30 2018-03-21 Differentiation of pluripotent stem cells

Applications Claiming Priority (10)

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
US61/076,908 2008-06-30
US61/076,900 2008-06-30
US61/076,915 2008-06-30
PCT/US2009/049183 WO2010002846A1 (en) 2008-06-30 2009-06-30 Differentiation of pluripotent stem cells
AU2009267137A AU2009267137A1 (en) 2008-06-30 2009-06-30 Differentiation of pluripotent stem cells
AU2015261644A AU2015261644B2 (en) 2008-06-30 2015-11-26 Differentiation of pluripotent stem cells
AU2018201993A AU2018201993B2 (en) 2008-06-30 2018-03-21 Differentiation of pluripotent stem cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2015261644A Division AU2015261644B2 (en) 2008-06-30 2015-11-26 Differentiation of pluripotent stem cells

Publications (2)

Publication Number Publication Date
AU2018201993A1 true AU2018201993A1 (en) 2018-04-12
AU2018201993B2 AU2018201993B2 (en) 2020-02-06

Family

ID=54848970

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2015261644A Active AU2015261644B2 (en) 2008-06-30 2015-11-26 Differentiation of pluripotent stem cells
AU2018201993A Active AU2018201993B2 (en) 2008-06-30 2018-03-21 Differentiation of pluripotent stem cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2015261644A Active AU2015261644B2 (en) 2008-06-30 2015-11-26 Differentiation of pluripotent stem cells

Country Status (1)

Country Link
AU (2) AU2015261644B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN118272378A (en) * 2024-04-24 2024-07-02 呈诺再生医学科技(北京)有限公司 Method for maintaining stem property of pluripotent stem cells and removing residues thereof in differentiated cells

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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

Also Published As

Publication number Publication date
AU2015261644A1 (en) 2015-12-17
AU2015261644B2 (en) 2017-12-21
AU2018201993B2 (en) 2020-02-06

Similar Documents

Publication Publication Date Title
US10233421B2 (en) Differentiation of pluripotent stem cells
AU2016202260B2 (en) Differentiation of human embryonic stem cells
JP2017012178A (en) Differentiation of human embryonic stem cells
AU2018201993B2 (en) Differentiation of pluripotent stem cells

Legal Events

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