US20210254017A1 - Culture Medium for Expanding Breast Epithelial Stem Cells - Google Patents

Culture Medium for Expanding Breast Epithelial Stem Cells Download PDF

Info

Publication number
US20210254017A1
US20210254017A1 US17/182,457 US202117182457A US2021254017A1 US 20210254017 A1 US20210254017 A1 US 20210254017A1 US 202117182457 A US202117182457 A US 202117182457A US 2021254017 A1 US2021254017 A1 US 2021254017A1
Authority
US
United States
Prior art keywords
breast
cells
organoid
culture medium
stem cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/182,457
Inventor
Lars Norman SACHS
Ana Gracanin
Jarno DROST
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.)
Koninklijke Nederlandse Akademie van Wetenschappen
Original Assignee
Koninklijke Nederlandse Akademie van Wetenschappen
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Koninklijke Nederlandse Akademie van Wetenschappen filed Critical Koninklijke Nederlandse Akademie van Wetenschappen
Priority to US17/182,457 priority Critical patent/US20210254017A1/en
Assigned to KONINKLIJKE NEDERLANDSE AKADEMIE VAN WETENSCHAPPEN reassignment KONINKLIJKE NEDERLANDSE AKADEMIE VAN WETENSCHAPPEN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DROST, Jarno, GRACANIN, Ana, SACHS, Lars Norman
Publication of US20210254017A1 publication Critical patent/US20210254017A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0693Tumour cells; Cancer cells
    • C12N5/0695Stem cells; Progenitor cells; Precursor cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0625Epidermal cells, skin cells; Cells of the oral mucosa
    • C12N5/0631Mammary cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor

Definitions

  • the present application is filed with a Sequence Listing in electronic format.
  • the Sequence Listing is provided as a file entitled “2021-02-22_01169-0008-01US_Sequence_Listing_ST25.txt” created on Feb. 22, 2021, which is 126,976 bytes in size.
  • the information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
  • the invention is in the field of breast epithelial stem cell culture media and methods, in particular culture media and methods for expanding populations of breast epithelial stem cells, e.g. human breast epithelial stem cells.
  • stem cells have many uses. For example, stem cells and their differentiated progeny can be used in cellular assays, drug screening, and toxicity assays. Stem cells also show promise for cell-based therapies, such as in regenerative medicine for the treatment of damaged tissue. Furthermore, efficient cell culture media are important for providing and maintaining populations of cells for research purposes.
  • a culture system for culturing epithelial stem cells derived from normal breast tissue is described in Pasic et al. (2011) Genes & Development 25:1641-1653. However, this system is unable to support long-term culture of breast organoids. Cultures using this system have been observed to stop expanding after 2-3 passages. Accordingly, it is not possible to obtain large numbers of stem cells using this technique.
  • a culture system for culturing circulating tumour cells from breast cancer patients is described in Yu et al. (2014) Science 345(6193):216-220. However, the authors of this paper provided no guidance with respect to culturing breast epithelial stem cells obtained from normal breast tissue, primary tumour or metastases. Furthermore, circulating breast tumour cells cultured using the Yu et al. system form aggregates of cells.
  • the invention provides a method for expanding breast epithelial stem cells comprising:
  • the invention further provides a culture medium comprising an ErbB3/4 ligand and one or more FGFR2b ligands and a Wnt agonist.
  • the invention further provides a culture medium comprising an ErbB3/4 ligand suitable for expanding a population of breast epithelial stem cells for at least 4 passages.
  • the invention further provides a breast organoid which comprises a population of breast epithelial stem cells.
  • said breast organoid is obtainable or obtained by a method of the invention.
  • the invention further provides a breast organoid of the invention in a culture medium of the invention.
  • the invention further provides the use of a breast organoid of the invention, or a cell derived from said organoid, in a drug discovery screen; toxicity assay; research of tissue embryology, cell lineages, or differentiation pathways; gene expression studies including recombinant gene expression; research of mechanisms involved in tissue injury or repair; research of inflammatory and infectious diseases; studies of pathogenetic mechanisms; or studies of mechanisms of cell transformation or aetiology of cancer.
  • the invention further provides an organoid of the invention, or a cell derived from said organoid, for use in medicine.
  • the invention further provides a culture medium comprising a p53 stabilising agent.
  • FIG. 1A-1B (A) Breast tumour tissues obtained from patients displayed according to subtype (estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor-2 (HER2) status); (B) established or promising organoid lines displayed according to subtype (ER, PR and HER2 status).
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2 human epidermal growth factor receptor-2
  • FIG. 2 Phase contrast images of established and promising mammary (A) tumour and (B) normal organoid lines. Scale bars equal 150 ⁇ m.
  • FIG. 3A-3B Example of media optimization. Normal and tumour mammary cells of patient W855 were suspended in Cultrex® Basement Membrane Extract (Trevigen, Inc.) and supplemented with the indicated media. Phase contrast images are displayed for: (A) Passage 0 day 4 and (B) Passage 0 day 14. The most beneficial culture conditions are marked by bold grey boxes. Image widths: 1.5 mm top panels, 300 ⁇ m bottom panels.
  • FIG. 4 Aneuploid karyotypes of established mammary tumour organoid lines.
  • FIG. 5 Histological and immunohistochemical comparison of original mammary gland tissue (W854 tumour (W854T), W855 normal and tumour (W855N and W855T), W859 tumour (W859T)) with the respectively derived organoid lines. Scale bars equal 100 ⁇ m (overview) and 20 ⁇ m (detailed view).
  • HE haematoxylin and eosin stain
  • PR progesterone receptor (clone PgR 636, M356929 Dako)
  • ER estrogen receptor alpha (clone SP1, ab6660 Abcam)
  • HER2 human epidermal growth factor receptor 2 (c-erbB2) (clone SP3, #RM-9103, Thermoscientific).
  • FIG. 6 Immunofluorescent analysis of whole mounted normal (W855) and tumour (W854, W855 and W859) mammary organoid lines. Nuclear counterstain is DRAQ5 (DR50050 Biostatus limited). Scale bar equals 100 ⁇ m.
  • FIG. 7 Quantitative PCR on organoids for basal (KRT5/14), luminal (KRT8), epithelial (CDH1) and mesenchymal (VIM, ZEB1) markers normalized to normal mammary organoid line W855N.
  • FIG. 8 Phase-contrast images of mammary tumour organoid lines W854T, W855T and W859T during a pilot drug screen with increasing doses of the EGFR inhibitor Iressa or the p53 stabilizer Nutlin-3. Visibly affected organoids are marked by bold black boxes. Image widths are 500 ⁇ m.
  • FIG. 9 Viability of mammary tumour organoid lines W854T, W855T and W859T treated with increasing does of the EGFR inhibitor Iressa or the p53 stabilizer Nutlin-3 for 7 days, as measured by a luminescent ATP assay (CellTiter-Glo 2.0, G9241 Promega).
  • FIG. 10 Normal human mammary organoids in the culture medium described in Pasic et al. compared with organoids from the same patient grown in base-F7/10E medium.
  • FIG. 11 Mouse mammary tumour organoids. Phase-contrast images of organoids established from KB2P mammary tumors lacking Brca2 and p53, Passage 4, Image widths are 7 mm, 1.5 mm, and 600 ⁇ m.
  • FIG. 12 Human metastatic mammary tumor organoids. Phase-contrast images of organoids established from a human breast cancer metastasis to the skin, Passage 5, Image widths are 7 mm, 1.5 mm, and 600 ⁇ m.
  • FIG. 13 Overview of mammary organoid cultures.
  • FIG. 14 Organoid culture media.
  • the ability to keep the cells and resulting organoids alive for longer and increase the passage number advantageously allows more cells to be obtained from a collection of starting cells than was possible using previous methods. This enables a large number of cells to be available for various applications, for example, drug screening, in which a large amount of material is required to test various different drugs.
  • the ability to generate the cells from a single starting source is advantageous for such applications where it is necessary to compare results between experiments.
  • the enhanced ability to obtain more cells from a small collection of starting cells e.g. a collection of approximately 100-300 cells
  • Culturing the cells in a medium of the invention allows the cells to multiply whilst retaining their stem or progenitor cell phenotype, which is referred to herein as expansion.
  • Organoids are formed comprising these stem or progenitor cells.
  • Use of the medium of the invention is therefore advantageous for providing increased numbers of these useful stem or progenitor cells and for obtaining organoids containing these cells.
  • a method for culturing breast epithelial stem cells comprising culturing one or more breast epithelial stem cells in contact with an extracellular matrix in the presence of a culture medium, the culture medium comprising a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands.
  • the culture medium used in the method of the invention is preferably a culture medium of the invention as described herein.
  • a culture medium comprising a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands (e.g. 1, 2, 3, 4, or more than 4), and one or more ligands of FGFR2b (e.g. 1, 2, 3, 4 or more than 4), preferably wherein the one or more ligands of FGFR2b is FGF7 and/or FGF10, for example, for use in the above method.
  • ErbB3/4 ligands e.g. 1, 2, 3, 4, or more than 4
  • FGFR2b e.g. 1, 2, 3, 4 or more than 4
  • a culture medium comprising a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands (e.g. 1, 2, 3, 4, or more than 4), and one or more Wnt agonists (e.g. 1, 2, 3, 4 or more than 4), for example, for use in the above method.
  • ErbB3/4 ligands e.g. 1, 2, 3, 4, or more than 4
  • Wnt agonists e.g. 1, 2, 3, 4 or more than 4
  • the one or more Wnt agonists is a Lgr5 agonist, Lgr4 agonist, Lgr6 agonist or Wnt3a.
  • the Lgr5 agonist is an Rspondin, e.g. any one of Rspondin 1-4.
  • the culture medium of the invention comprises a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands (e.g. 1, 2, 3, 4, or more than 4), one or more ligands of FGFR2b (e.g. 1, 2, 3, 4 or more than 4), preferably wherein the one or more ligands of FGFR2b is FGF7 and/or FGF10, and one or more Wnt agonists (e.g. 1, 2, 3, 4 or more than 4), preferably wherein the one or more Wnt agonists is a Lgr5 agonist, preferably wherein the Lgr5 agonist is an Rspondin, e.g. any one of Rspondin 1-4.
  • one or more ErbB3/4 ligands e.g. 1, 2, 3, 4, or more than 4
  • FGFR2b e.g. 1, 2, 3, 4 or more than 4
  • Wnt agonists e.g. 1, 2, 3, 4 or more than 4
  • the culture medium of the invention or the culture medium used in the method of the invention comprises one or more additional components selected from: a ligand of FGFR2b (e.g. FGF7 and/or FGF10), a Lgr5 agonist (e.g. Rspondin, e.g. any one of Rspondin 1-4), a BMP inhibitor (for example, Noggin), B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor (for example, an ALK 4, 5, 7 inhibitor, e.g. A83-01) and a p38 MAP kinase inhibitor (for example, SB 202190).
  • a ligand of FGFR2b e.g. FGF7 and/or FGF10
  • a Lgr5 agonist e.g. Rspondin, e.g. any one of Rspondin 1-4
  • BMP inhibitor for example, Noggin
  • B27 for example, N-ace
  • the culture medium further comprises at least two, at least three, at least four, at least five, at least six, at least seven, (e.g. 2, 3, 4, 5, 6, 7) of these additional components. In some embodiments, the culture medium further comprises all of these additional components.
  • the culture medium further comprises a further receptor tyrosine kinase ligand, e.g. EGF.
  • a further receptor tyrosine kinase ligand e.g. EGF.
  • the culture medium further comprises a Wnt agonist, e.g. Wnt3a.
  • the culture medium further comprises one or more of amphiregulin, TGF-alpha and PDGF-CC.
  • the culture medium further comprises PDGF-CC.
  • the culture medium further comprises amphiregulin, TGF-alpha and PDGF-CC.
  • the invention also provides a method for culturing breast epithelial stem cells which uses an expansion medium as described in WO2012/168930, WO2010/090513 or WO2012/014076 to which at least one (e.g. 1, 2, 3, 4, or more than 4) ErbB3/4 ligand and at least one (e.g. 1, 2, 3, 4, or more than 4) FGFR2b ligands are added.
  • an expansion medium as described in WO2012/168930, WO2010/090513 or WO2012/014076 to which at least one (e.g. 1, 2, 3, 4, or more than 4) ErbB3/4 ligand and at least one (e.g. 1, 2, 3, 4, or more than 4) FGFR2b ligands are added.
  • p53 stabilizing agents increase the cellular concentration of p53 (e.g. by blocking the interaction between p53 and Mdm2), stimulate p53-dependent expression and induce cellular senescence.
  • p53 mutations commonly occur in tumour cells, which may result in the mutant p53 protein being unable to be stabilised by a p53 stabilizing agent.
  • a subset of tumour cells with the mutant p53 protein would escape the senescence induced by a p53 stabilizing agent and outgrow normal cells in a mixed population.
  • the invention therefore further provides the use of a p53 stabilizing agent for culturing tumour cells.
  • the invention also provides a method for culturing tumour stem cells which uses an expansion medium as described herein or in WO2012/168930, WO2010/090513 or WO2012/014076 to which at least one (e.g. 1, 2, 3, 4, or more than 4) p53 stabilizing agent is added.
  • the invention also provides a culture medium comprising a p53 stabilizing agent.
  • the culture medium is a culture medium as described herein or in W2012/168930, WO2010/090513 or WO2012/014076.
  • the culture medium is suitable for culturing epithelial stem cells.
  • a preferred p53 stabilizing agent is a member of the Nutlin family, e.g. Nutlin-1, Nutlin-2 or Nutlin-3.
  • the p53 stabilizing agent is Nutlin-3.
  • Other p53 stabilising agents are known in the art (e.g. CP-31398) and the skilled person will be able to use any of these accordingly.
  • the ErbB receptor tyrosine kinase family consists of four cell surface receptors: i) ErbB1/EGFR/HER1, ii) ErbB2/HER2, iii) ErbB3/HER3, and iv) ErbB4/HER4.
  • An ErbB3/4 ligand is herein defined as a ligand that is capable of binding to ErbB3 and/or ErbB4. Accordingly, EGF is not an ErbB3/4 ligand because it does not bind to ErbB3/4. In some embodiments, the Erb3/4 ligand binds to ErbB3 and does not bind to ErbB4.
  • the Erb3/4 ligand binds to Erb4 and does not bind to Erb3. In some embodiments, binding of the ErbB3/4 ligand to ErbB3 or ErbB4 induces the heterodimerization of said ErbB3 or said ErbB4 with ErbB2. In some embodiments, the induction of heterodimerization of ErbB3 or ErbB4 with ErbB2 stimulates intrinsic kinase activity, which leads to tyrosine phosphorylation. In the context of a culture medium of the invention, an ErbB3/4 ligand is known as “N”.
  • the one or more ErbB3/4 ligands of the culture medium are members of the neuregulin/heregulin family.
  • the neuregulin/heregulin family is referred to herein as the neuregulin family.
  • the neuregulin family is a family of structurally related polypeptide growth factors that are gene products of alternatively spliced genes NRG1, NRG2, NRG3 and NRG4.
  • the one or more ErbB3/4 ligands of the culture medium are polypeptides that are gene products of one or more of NRG1, NRG2, NRG3 and NRG4 (i.e., a neuregulin polypeptide).
  • the ErbB3/4 ligand is a polypeptide that is a gene product of NRG.
  • the polypeptide that is a gene product of NRG1, NRG2, NRG3 and NRG4 may be any of the isoforms that result from the alternative splicing of NRG1, NRG2, NRG3 or NRG4 mRNA.
  • the polypeptide that is a gene product of NRG1 may be any of the following isoforms: Type I NRG1 (also known as heregulin, NEU differentiation factor (NDF) or acetylcholine receptor inducing activity (ARIA)), Type II NRG1 (also known as Glial Growth Factor-2 (GGF2)), Type III NRG1 (also known as Sensory and motor neuron-derived factor (SMDF)), Type IV NRG1, Type V NRG1 or Type VI NRG1.
  • Type I NRG1 also known as heregulin, NEU differentiation factor (NDF) or acetylcholine receptor inducing activity (ARIA)
  • Type II NRG1 also known as Glial Growth Factor-2 (GGF2)
  • Type III NRG1 also known as Sensory and motor neuron-derived factor (SMDF)
  • Type IV NRG1, Type V NRG1 or Type VI NRG1 Type I NRG1
  • Type V NRG1 also known as heregulin, NEU differentiation factor (NDF)
  • the neuregulin polypeptide is pro-neuregulin-1, membrane-bound isoform isoform HRG-beta1 (NP_039250.2), pro-neuregulin-1, membrane-bound isoform isoform HRG-beta1b (NP_001153471.1), pro-neuregulin-1, membrane-bound isoform isoform HRG-beta1c (NP_001153467.1), pro-neuregulin-1 or membrane-bound isoform isoform HRG-beta1d (NP_001153473.1).
  • the ErbB3/4 ligand is of human origin. Accordingly, in some embodiments the ErbB3/4 ligand is a human gene product of one or more of NRG1, NRG2, NRG3 and NRG4 (i.e. a human neuregulin polypeptide).
  • the NRG1 gene has the sequence of Gene ID: 3084 or NG_012005.1. In some embodiments, the NRG2 gene has the sequence of Gene ID: 9542. In some embodiments, the NRG3 gene has the sequence of Gene ID: 10718 or NG_013373.1. In some embodiments, the NRG4 gene has the sequence shown of Gene ID: 145957.
  • a neuregulin polypeptide is a gene product of NRG1 and has the sequence shown in NP_001153467.1 (SEQ ID NO: 1), NP_001153468.1 (SEQ ID NO: 2), NP_001153471.1 (SEQ ID NO: 3), NP_001153473.1 (SEQ ID NO: 4), NP_001153474.1 (SEQ ID NO: 5), NP_001153476.1 (SEQ ID NO: 6), NP_001153477.1 (SEQ ID NO: 7), NP_001153479.1 (SEQ ID NO: 8), NP_001153480.1 (SEQ ID NO: 9), NP_004486.2 (SEQ ID NO: 10), NP_039250.2 (SEQ ID NO: 11), NP_039251.2 (SEQ ID NO: 12), NP_039252.2 (SEQ ID NO: 13), NP_039253.1 (SEQ ID NO: 14), NP_039254.1 (SEQ ID NO: 15), NP_03925
  • a neuregulin polypeptide is a gene product of NRG2 and has the sequence shown in NP_001171864.1 (SEQ ID NO: 18), NP_004874.1 (SEQ ID NO: 19), NP_053584.1 (SEQ ID NO: 20), NP_053585.1 (SEQ ID NO: 21), or NP_053586.1 (SEQ ID NO: 22).
  • a neuregulin polypeptide is a gene product of NRG3 and has the sequence shown in NP_001010848.2 (SEQ ID NO: 23), NP_001159444.1 (SEQ ID NO: 24), or NP_001159445.1 (SEQ ID NO: 25).
  • a neuregulin polypeptide is a gene product of NRG4 and has the sequence shown in NP_612640.1 (SEQ ID NO: 26).
  • the at least one ErbB3/4 ligand is a biologically active variant of one or more naturally occurring ErB3/4 ligands, for example, of one or more members of the neuregulin family.
  • Neuregulin variants which may be naturally occurring (e.g. allelic variants that occur at the NRG1 locus) or recombinantly produced, have amino acid sequences that are the same as, similar to or substantially similar to a neuregulin polypeptide.
  • a neuregulin variant has a sequence that is at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a neuregulin polypeptide, for example the neuregulin variant has a sequence that is at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to any one of SEQ ID NOs: 1-26.
  • the at least one ErbB3/4 ligand is at least one biologically active fragment of at least one naturally occurring ErbB3/4 ligand, for example a biologically active fragment of of one or more neuregulin polypeptides.
  • Biologically active is defined herein as meaning that the ErbB3/4 ligand, for example, the variant or fragment, is capable of binding to ErbB3 and/or ErbB4, optionally wherein binding to ErbB3 and/or ErbB4 induces the heterodimerization of said ErbB3 or said ErbB4 with ErbB2, and optionally wherein said induction of heterodimerization of ErbB3 or ErbB4 with ErbB2 stimulates intrinsic kinase activity, which leads to tyrosine phosphorylation.
  • the ErbB3/4 ligand is a fragment of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26 wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95 or at least 100 amino acids of the sequence recited in any one of SEQ ID NOs: 1-26.
  • the ErbB3/4 ligand is a variant of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26.
  • references to a percentage sequence identity between two amino acid sequences means that, when aligned, that percentage of amino acids are the same in comparing the two sequences.
  • This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987) Supplement 30.
  • a preferred alignment is determined by the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62.
  • the Smith-Waterman homology search algorithm is disclosed in Smith & Waterman (1981) Adv. Appl. Math. 2:482-489.
  • the ErbB3/4 ligand is a variant of a fragment of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95 or at least 100 amino acids of the sequence recited in any one of SEQ ID NOs: 1-26, and wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the fragment.
  • the ErbB3/4 ligand is a fragment of a biologically active variant of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, and wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95 or at least 100 amino acids of the variant.
  • the ErbB3/4 ligand is a non-naturally occurring ligand, for example a synthetic ligand or an anti-ErbB3/4 antibody.
  • Methods of generating antibodies against a target of interest are well known in the art.
  • the anti-ErbB3/4 antibody is an agonistic anti-ErbB3/4 antibody.
  • the antibody is biologically active. Any suitable antibodies may be used, for example, as described herein.
  • ErbB3/4 ligands can be identified using methods known in the art, for example, binding of GST-tagged candidate ligands to insect-cell-expressed ErbB receptors causing tyrosine phosphorylation (Carraway 3 rd et al., (1997) Nature 387(6632):512-6).
  • the ErbB3/4 ligand comprises an EGF domain or EGF-like domain.
  • EGF and EGF-like domains are evolutionary conserved protein domains that are recognised in the art (see, for example, Wouters et al. (2005) Protein Sci. 14(4): 1091-1103). Accordingly, in some embodiments in which the ErbB3/4 ligand is a biologically active fragment of at least one naturally occurring ErbB3/4 ligand, the fragment comprises an EGF domain or an EGF-like domain.
  • the ErbB3/4 ligand is human neuregulin ⁇ -1.
  • the human neuregulin ⁇ -1 has the amino acid sequence recited in SEQ ID NO: 27.
  • Human neuregulin ⁇ -1 is a fragment of a neuregulin polypeptide, wherein the fragment comprises an EGF-like domain.
  • the ErbB3/4 ligand comprises or consists of the EGF-like domain of human neuregulin ⁇ -1.
  • the ErbB3/4 ligand has similar biological activity to recombinant human neuregulin ⁇ -1.
  • the ErbB3/4 ligand is a fragment of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 62, or at least 64 amino acids of the sequence recited in SEQ ID NO: 27.
  • the ErbB3/4 ligand is a variant of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in SEQ ID NO: 27.
  • the ErbB3/4 ligand is a variant of a fragment of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 62 or at least 64 amino acids of the sequence recited in SEQ ID NO: 27, and wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the fragment.
  • the ErbB3/4 ligand is a fragment of a biologically active variant of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, and wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 62 or at least 64 amino acids of the variant.
  • the ErbB3/4 ligand may be present at any suitable concentration.
  • the ErbB3/4 ligand may be present at a concentration of between 0.05 and 500 nM, between 0.05 and 100 nM, between 0.05 and 50 nM, between 0.05 and 10 nM, between 0.05 and 5 nM, between 0.05 and 1 nM, between 0.5 and 500 nM, between 0.5 and 100 nM, between 0.5 and 50 nM, between 0.5 and 10 nM, between 0.5 nM and 1 nM, between 1 and 10 nM, between 3 and 10 nM, between 3 and 8 nM, between 4 and 6 nM.
  • the ErbB3/4 ligand may be present at a concentration of approximately 5 nM.
  • the ErbB3/4 ligand is used at a concentration of at least 0.05 nM, at least 0.5 nM, at least M, at least 3 nM, at least 4 nM, at least 5 nM, at least 10 nM, at least 50 nM, or at least 100 nM.
  • cell culture media generally contain a number of components which are necessary to support maintenance and/or expansion of the cultured cells.
  • a cell culture medium of the invention will therefore normally contain many other components in addition to an ErbB3/4 ligand. Suitable combinations of components can readily be formulated by the skilled person, taking into account the disclosure herein.
  • a culture medium according to the invention will generally be a nutrient solution comprising standard cell culture components, such as amino acids, vitamins, inorganic salts, a carbon energy source, and a buffer as described in more detail below.
  • Other standard cell culture components that may be included in the culture include hormones, such as progesterone, proteins, such as albumin, catalase, insulin and transferrin. These other standard cell culture components make up the “basal” culture medium.
  • a culture medium according to the invention may be generated by modification of an existing cell culture medium.
  • Suitable cell culture media include, but are not limited to, Dulbecco's Modified Eagle Media (DMEM), Minimal Essential Medium (MEM), Knockout-DMEM (KO-DMEM), Glasgow Minimal Essential Medium (G-MEM), Basal Medium Eagle (BME), DMEM/Ham's F12, Advanced DMEM/Ham's F12, Iscove's Modified Dulbecco's Media and Minimal Essential Media (MEM), Ham's F-10, Ham's F-12, Medium 199, and RPMI 1640 Media. Examples of suitable “basal” culture are also provided in WO2012/168930, WO2010/090513, and WO 2012/014076.
  • the basal medium is AdDF+++.
  • the AdDF+++ comprises Advanced DMEM/F12 (Dulbecco's Modified Eagle Medium/Ham's F-12), GlutaMax, HEPES, penicillin/streptomycin, primocin.
  • the basal medium is D10F.
  • the D10F comprises 31966-DMEM (Dulbecco's Modified Eagle Medium), fetal bovine serum (FBS) and penicillin/streptomycin.
  • the culture medium of the invention additionally comprises one or more ligands of FGFR2b (for example, FGF7 and/or FGF10) and/or one or more Wnt agonists (preferably an agonistic Lgr5 ligand, preferably Rspondin).
  • the culture medium of the invention comprises the following additional components: (i) FGF7 and/or FGF10 and (ii) Rspondin.
  • the culture medium of the invention comprises the following additional components: (i) FGF7 and/or FGF10, (ii) Rspondin and (iii) one or more further receptor tyrosine kinase ligands.
  • the culture medium of the invention further comprises one or more components selected from: a BMP inhibitor (for example, Noggin), B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor (for example, A83-01), a p38 MAP kinase inhibitor (for example, SB 202190).
  • a BMP inhibitor for example, Noggin
  • B27 for example, N-acetylcysteine
  • Nicotinamide a ROCK inhibitor
  • TGF-beta inhibitor for example, A83-01
  • a p38 MAP kinase inhibitor for example, SB 202190.
  • the culture medium of the invention comprises an ErbB3/4 ligand, Rspondin, a BMP inhibitor (for example, Noggin), B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor (for example, A83-01), a p38 MAP kinase inhibitor (for example, SB 202190), FGF7 and FGF10, and optionally one or more additional components selected from: one or more further receptor tyrosine kinase ligands (for example, EGF, amphiregulin, TGF-alpha, PDGF), a p53 stabilizing agent and a Wnt agonist (for example, Wnt3a).
  • BMP inhibitor for example, Noggin
  • B27 for example, N-acetylcysteine
  • Nicotinamide for example, a ROCK inhibitor
  • TGF-beta inhibitor for example, A83-01
  • Feeder cell layers are often used to support the culture of stem cells, and to inhibit their differentiation.
  • a feeder cell layer is generally a monolayer of cells that is co-cultured with, and which provides a surface suitable for growth of, the cells of interest.
  • the feeder cell layer provides an environment in which the cells of interest can grow.
  • Feeder cells are often mitotically inactivated (e.g. by irradiation or treatment with mitomycin C) to prevent their proliferation.
  • the use of feeder cells is undesirable, because it complicates passaging of the cells (the cells must be separated from the feeder cells at each passage, and new feeder cells are required at each passage).
  • the use of feeder cells can also lead to contamination of the desired cells with the feeder cells. This is clearly problematic for any medical applications, and even in a research context, complicates analysis of the results of any experiments performed on the cells.
  • the culture media of the invention are particularly advantageous because they can be used to culture cells without feeder cell contact, i.e. the methods of the invention do not require a layer of feeder cells to support the cells whose growth is being sponsored.
  • compositions of the invention may be feeder cell-free compositions.
  • a composition is conventionally considered to be feeder cell-free if the cells in the composition have been cultured for at least one passage in the absence of a feeder cell layer.
  • a feeder cell-free composition of the invention will normally contain less than about 5%, less than about 4%, less than about 3%, less than about 2%, less than about 1% feeder cells (expressed as a % of the total number of cells in the composition) or preferably no feeder cells at all.
  • the culture media of the invention may comprise one or more bone morphogenetic protein (BMP) inhibitor.
  • BMP bone morphogenetic protein
  • the culture medium may comprise any suitable BMP inhibitor.
  • BMPs bind as a dimeric ligand to a receptor complex consisting of two different receptor serine/threonine kinases, type I and type II receptors.
  • the type II receptor phosphorylates the type I receptor, resulting in the activation of this receptor kinase.
  • the type I receptor subsequently phosphorylates specific receptor substrates (SMAD), resulting in a signal transduction pathway leading to transcriptional activity.
  • BMP inhibitors promote expression of Lgr5, and so the presence of a BMP inhibitor in a culture medium of the invention will likely result in more proliferative organoids than if the BMP inhibitor is absent.
  • cells cultured with a BMP inhibitor have upregulated expression of Lgr5 compared to cells cultured without a BMP inhibitor. Therefore, addition of a BMP inhibitor typically results in more proliferative organoids.
  • a BMP inhibitor is defined as an agent that binds to a BMP molecule to form a complex wherein the BMP activity is neutralized, for example by preventing or inhibiting the binding of the BMP molecule to a BMP receptor.
  • said inhibitor is an agent that acts as an antagonist or reverse agonist.
  • This type of inhibitor binds with a BMP receptor and prevents binding of a BMP to said receptor.
  • An example of a latter agent is an antibody that binds a BMP receptor and prevents binding of BMP to the antibody-bound receptor.
  • a BMP inhibitor may be added to the media in an amount effective to inhibit a BMP-dependent activity in a cell to at most 90%, more preferred at most 80%, more preferred at most 70%, more preferred at most 50%, more preferred at most 30%, more preferred at most 10%, more preferred 0%, relative to a level of a BMP activity in the absence of said inhibitor, as assessed in the same cell type.
  • a BMP activity can be determined by measuring the transcriptional activity of BMP, for example as exemplified in Zilberberg et al., 2007. BMC Cell Biol. 8:41.
  • BMP-binding proteins include Noggin (Peprotech), Chordin and chordin-like proteins (R&D systems) comprising chordin domains, Follistatin and follistatin-related proteins (R&D systems) comprising a follistatin domain, DAN and DAN-like proteins (R&D systems) comprising a DAN cysteine-knot domain, sclerostin/SOST (R&D systems), decorin (R&D systems), and alpha-2 macroglobulin (R&D systems).
  • Noggin Pheprotech
  • Chordin and chordin-like proteins comprising chordin domains
  • Follistatin and follistatin-related proteins comprising a follistatin domain
  • DAN and DAN-like proteins comprising a DAN cysteine-knot domain
  • sclerostin/SOST R&D systems
  • decorin R&D systems
  • alpha-2 macroglobulin R&D systems
  • BMP inhibitors for use in a method of the invention are Noggin, DAN, and DAN-like proteins including Cerberus and Gremlin (R&D systems). These diffusible proteins are able to bind a BMP ligand with varying degrees of affinity and inhibit their access to signalling receptors. The addition of any of these BMP inhibitors to the basal culture medium prevents the loss of stem cells.
  • a preferred BMP inhibitor is Noggin.
  • the culture medium as described herein comprises Noggin.
  • Noggin is also referred to herein as “No”.
  • Noggin is preferably added to the basal culture medium at a concentration of at least 10 ng/ml, for example, at least 20 ng/ml, more preferred at least 25 ng/ml. A still more preferred concentration is about 25 ng/ml.
  • said BMP inhibitor may be added to the culture medium when required, for example, daily or every other day.
  • the BMP inhibitor is preferably added to the culture medium every second day.
  • the culture medium may be refreshed when required, for example, daily or every other day.
  • the culture media of the invention may comprise one or more Wnt agonist.
  • the Wnt signalling pathway is defined by a series of events that occur when the cell-surface Wnt receptor complex, comprising a Frizzled receptor, LRP and LGR is activated, usually be an extracellular signalling molecule, such as a member of the Wnt family. This results in the activation of Dishevelled family proteins which inhibit a complex of proteins that includes axin, GSK-3, and the protein APC to degrade intracellular ⁇ -catenin. The resulting enriched nuclear ⁇ -catenin enhances transcription by TCF/LEF family transcription factors.
  • a Wnt agonist is defined as an agent that activates TCF/LEF-mediated transcription in a cell.
  • Wnt agonists are therefore selected from true Wnt agonists that bind and activate the Wnt receptor complex including any and all of the Wnt family proteins, an inhibitor of intracellular ⁇ -catenin degradation, a GSK inhibitor (such as CHIR9901) and activators of TCF/LEF.
  • a Wnt agonist is a secreted glycoprotein including Wnt-1/Int-1, Wnt-2/Irp (1 nM-related Protein), Wnt-2b/13, Wnt-3/Int-4, Wnt-3a (R&D systems), Wnt-4, Wnt-5a, Wnt-5b, Wnt-6 (Kirikoshi H et al 2001 Biochem Biophys Res Com 283 798-805), Wnt-7a (R&D systems), Wnt-7b, Wnt-8a/8d, Wnt-8b, Wnt-9a/14, Wnt-9b/14b/15, Wnt-10a, Wnt-10b/12, WnM 1, and Wnt-16.
  • the Wnt agonist is an inhibitor of RNF43 or ZNRF3. It has been shown that RNF43 and ZNRF3 reside in the cell membrane and negatively regulate levels of the Wnt receptor complex in the membrane, probably by ubiquitination of Frizzled. Therefore, the inventors hypothesise that inhibition of RNF43 or ZNRF3 with antagonistic antibodies, RNAi or small molecule inhibitors would indirectly stimulate the Wnt pathway. RNF43 and ZNRF3 have a catalytic ring domain (with ubiquitination activity), which can be targeted in small molecule inhibitor design.
  • anti-RNF43 antibodies and several anti-ZNRF3 antibodies are available commercially. In some embodiments, such antibodies are suitable Wnt agonists in the context of the invention.
  • the Wnt agonist in the culture medium is any agonist able to stimulate the Wnt pathway via the Lgr5 cell surface receptor, i.e. in a preferred embodiment, the Wnt agonist in the culture medium is an Lgr5 agonist.
  • Lgr5 agonists include Rspondin, fragments and derivatives thereof, and anti-Lgr5 antibodies (e.g. see WO 2012/140274 and De Lau, W. et al. Nature, 2011 Jul. 4; 476(7360):293-7).
  • a preferred Lgr5 agonist is Rspondin.
  • any suitable Rspondin may be used, for example, it may be selected from one or more of Rspondin 1, Rspondin 2, Rspondin 3 and Rspondin 4 or derivatives thereof.
  • Rspondin 1 NU206, Nuvelo, San Carlos, Calif.
  • Rspondin 2 ((R&D systems), Rspondin 3, and Rspondin 4)
  • Rspondin 1, 2, 3, and 4 are also referred to herein as “Rspondin 1-4”.
  • Fragments of Rspondin may be used as the Wnt agonist.
  • the Wnt agonist is a fragment of Rspondin comprising or consisting of the furin domain.
  • the Lgr5 agonist is an anti-Lgr5 antibody, more preferably an agonistic anti-Lgr5 antibody.
  • An example of an agonistic anti-Lgr5 antibody is 1D9 (available commercially from BD Biosciences, BDB562733, No.:562733). Therefore, in one embodiment, the agonist is the antibody 1D9.
  • the VL of antibody 1D9 is represented by SEQ ID NO: 32 and the VH is represented by SEQ ID NO: 33. Therefore, in one embodiment, the agonist is an antibody comprising or consisting of SEQ ID NO: 32 and/or SEQ ID NO: 33.
  • the Wnt agonist in the culture medium is any agonist able to stimulate the Wnt pathway via the Lgr4 cell surface receptor, i.e. in some embodiments, the Wnt agonist in the culture medium is an Lgr4 agonist.
  • the Wnt agonist in the culture medium is any agonist able to stimulate the Wnt pathway via the Lgr6 cell surface receptor, i.e. in some embodiments, the Wnt agonist in the culture medium is an Lgr6 agonist.
  • the Wnt agonist may be added to the media in an amount effective to stimulate a Wnt activity in a cell by at least 10%, more preferred at least 20%, more preferred at least 30%, more preferred at least 50%, more preferred at least 70%, more preferred at least 90%, more preferred at least 100%, relative to a level of said Wnt activity in the absence of said molecule, as assessed in the same cell type.
  • Wnt activity can be determined by measuring the transcriptional activity of Wnt, for example by pTOPFLASH and pFOPFLASH Tcf luciferase reporter constructs (Korinek et al., 1997. Science 275:1784-1787).
  • a soluble Wnt agonist such as Wnt-3a, may be provided in the form of Wnt conditioned media.
  • Wnt conditioned media For example, about 10% to about 30%, e.g. about 10 ng/ml to about 10 ⁇ g/ml, preferably about 1 ⁇ g/ml, Wnt conditioned media may be used.
  • Rspondin 1-4 may be provided in the form of Rspo conditioned media.
  • Rspo conditioned media For example, about 10% to about 30%, e.g. about 10 ng/ml to about 10 ⁇ g/ml, preferably about 1 ⁇ g/ml, Rspo conditioned media may be used.
  • the culture medium comprises an Lgr5 agonist, for example Rspondin, and additionally comprises a further Wnt agonist.
  • the further Wnt agonist may, for example, be selected from the group consisting of Wnt-3a, a GSK-inhibitor (such as CHIR99021), Wnt-5, Wnt-6a and Norrin.
  • the culture medium comprises Rspondin and additionally comprises a soluble Wnt ligand, such as Wnt3a. Addition of a soluble Wnt ligand has been shown to be particularly advantageous for expansion of human epithelial stem cells (as described in WO2012/168930).
  • Any suitable concentration of Wnt agonist e.g. Rspondin
  • a still more preferred concentration of Rspondin is at least 500 ng/ml or about 1 ⁇ g/ml.
  • said Wnt agonist may be added to the culture medium when required, for example, daily or every other day.
  • the Wnt agonist is preferably added to the culture medium every second day.
  • Antibodies such as anti-ErbB3/4 antibodies, agonistic anti-Lgr5 antibodies or antagonistic TGF-beta inhibitors (see below), used in the invention may be any antibodies, fragments, etc.
  • a conventional antibody is comprised of two identical heavy chains and two identical light chains that are joined by disulfide bonds. Each heavy and light chain contains a constant region and a variable region. Each variable region contains three CDRs which are primarily responsible for binding an epitope of an antigen. They are referred to as CDR1, CDR2, and CDR3, numbered sequentially from the N-terminus, of which the CDR3 region comprises the most variable region and normally provides a substantial part of the contact residues to a target. The more highly conserved portions of the variable regions are called the “framework regions”.
  • antibody is used herein in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies) of any isotype such as IgG, IgM, IgA, IgD and IgE, polyclonal antibodies including recombinant polyclonal antibodies, Oligoclonics, multispecific antibodies, chimeric antibodies, nanobodies, diabodies, BiTE's, Tandabs, mimetobodies, bispecific antibodies, humanized antibodies, human antibodies, deimmunised antibodies and antibody fragments.
  • scaffolds will be covered under this term, such as Anticalins, Ankarins, etc.
  • An antibody reactive with a the specific epitopes of the Lgr proteins discussed above can be generated by recombinant methods such as selection of libraries of recombinant antibodies in phage or similar vectors, or by immunizing an animal with the Lgr epitopes of nucleic acid encoding them.
  • an antibody according to the invention comprises a single domain antibody, a F(ab′)2, Fab, Fab′, or single chain Fv (scFv) fragment.
  • An Fc fragment which for example activates complement and may bind to Fc receptors, can be present but is not required for an antibody and variants or derivatives thereof.
  • a scFv fragment is an epitope-binding fragment that contains at least one fragment of an antibody heavy chain variable region (VH) linked to at least one fragment of an antibody light chain variable region (VL).
  • the linker may be a short, flexible peptide selected to assure that the proper three-dimensional folding of the VL and VH regions occurs once they are linked so as to maintain the target molecule binding-specificity of the whole antibody from which the single-chain antibody fragment is derived.
  • the carboxyl terminus of the VL or VH sequence may be covalently linked by a linker to the amino acid terminus of a complementary VL or VH sequence.
  • the antibody may be a diabody, mimetibody, nanobody, and/or a bispecific antibody.
  • a nanobody is a single domain antibody that occurs naturally in camelids.
  • nanobodies are relatively simple proteins comprising only a heavy chain-like variable region.
  • Bispecific antibodies are artificially engineered monoclonal antibodies that consist of two distinct binding sites and are capable of binding two different epitopes. Examples of bispecific antibodies are discussed in more detail below in the section on dual-targeting and multi-targeting agonists.
  • the antibody may be a chimeric antibody comprising a binding portion, for example the variable region or part thereof of the heavy and light chains, of a non-human antibody, while the remainder portion, for example the constant region of the heavy and light chains, is of a human antibody.
  • a chimeric antibody may be produced by recombinant processes well known in the art, and has an animal variable region and a human constant region.
  • the antibody may be a human antibody or a humanized antibody.
  • human antibody means an antibody in which the variable and constant domain sequences are derived from human sequences. In a humanized antibody, only the complementarity determining regions (CDRs), which are responsible for antigen binding and specificity are animal derived and have an amino acid sequence corresponding to the animal antibody, and substantially all of the remaining portions of the molecule (except, in some cases, small portions of the framework regions within the variable region) are human derived and correspond in amino acid sequence to a human antibody.
  • CDRs complementarity determining regions
  • antibodies such as rat antibodies can be humanized by grafting their CDRs onto the variable light (VL) and variable heavy (VH) frameworks of human Ig molecules, while retaining those rat framework residues deemed essential for specificity and affinity.
  • VL variable light
  • VH variable heavy
  • CDR grafted antibodies consist of more than 80% human amino acid sequences.
  • the antibody is a deimmunised antibody in which the T and B cell epitopes have been eliminated. They have reduced immunogenicity when applied in vivo.
  • the culture media of the invention may comprise a TGF-beta inhibitor.
  • TGF-beta inhibitor in the media is advantageous because it increases human organoid formation efficiency.
  • TGF-beta signalling is involved in many cellular functions, including cell growth, cell fate and apoptosis. Signalling typically begins with binding of a TGF-beta superfamily ligand to a type II receptor which recruits and phosphorylates a type I receptor. The type I receptor then phosphorylates SMADs, which act as transcription factors in the nucleus and regulate target gene expression.
  • the TGF-beta superfamily ligands comprise bone morphogenic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, nodal and TGF-betas.
  • BMPs bone morphogenic proteins
  • GDFs growth and differentiation factors
  • AMH anti-mullerian hormone
  • Smad2 and Smad3 are phosphorylated by the ALK4, 5 and 7 receptors in the TGF-beta/activin pathway.
  • Smad1, Smad5 and Smad8 are phosphorylated as part of the bone morphogenetic protein (BMP) pathway.
  • a “TGF-beta inhibitor” or an “inhibitor of TGF-beta signalling” is preferably an inhibitor of the TGF-beta pathway which acts via Smad2 and Smad3. Therefore, in some embodiments the TGF-beta inhibitor is not a BMP inhibitor, e.g. the TGF-beta inhibitor is not Noggin. In some embodiments, a BMP inhibitor is added to the culture medium in addition to the TGF-beta inhibitor (see above).
  • the TGF-beta inhibitor may be any agent that reduces the activity of the TGF-beta signalling pathway, preferably the signalling pathway that acts via Smad2 and/or Smad3, more preferably the signalling pathway that acts via ALK4, ALK5 or ALK7.
  • the signalling pathway that acts via Smad2 and/or Smad3 preferably the signalling pathway that acts via ALK4, ALK5 or ALK7.
  • the TGF-beta signaling may be disrupted by: inhibition of TGF-beta expression by a small-interfering RNA strategy; inhibition of furin (a TGF-beta activating protease); inhibition of the pathway by physiological inhibitors; neutralisation of TGF-beta with a monoclonal antibody; inhibition with small-molecule inhibitors of TGF-beta receptor kinase 1 (also known as activin receptor-like kinase, ALK5), ALK4, ALK6, ALK7 or other TGF-beta-related receptor kinases; inhibition of Smad 2 and Smad 3 signaling e.g.
  • a cellular assay may be used in which cells are stably transfected with a reporter construct comprising the human PAI-1 promoter or Smad binding sites, driving a luciferase reporter gene. Inhibition of luciferase activity relative to control groups can be used as a measure of compound activity (De Gouville et al., Br J Pharmacol. 2005 May; 145(2): 166-177).
  • a TGF-beta inhibitor according to the present invention may be a protein, peptide, small-molecules, small-interfering RNA, antisense oligonucleotide, aptamer or antibody.
  • the inhibitor may be naturally occurring or synthetic.
  • the TGF-beta inhibitor is an inhibitor of ALK4, ALK5 and/or ALK7.
  • the TGF-beta inhibitor may bind to and directly inhibit ALK4, ALK5 and/or ALK7.
  • Examples of preferred small-molecule TGF-beta inhibitors that can be used in the context of this invention include the small molecule inhibitors listed in Table 1:
  • the TGF-beta inhibitor is a small molecule inhibitor optionally selected from the group consisting of: A83-01, SB-431542, SB-505124, SB-525334, LY 364947, SD-208 and SJN 2511.
  • a medium of the invention comprises one or more of any of the inhibitors listed in table 1.
  • a medium may comprise any combination of one inhibitor with another inhibitor listed.
  • a medium may comprise SB-525334 or SD-208 or A83-01; or SD-208 and A83-01.
  • SB-203580 is a p38 MAP kinase inhibitor that, at high concentrations (for example, approximate 10 ⁇ M or more) is thought to inhibit ALK5. Any such inhibitor that inhibits the TGF-beta signalling pathway can also be used in the context of this invention.
  • the TGF beta inhibitor is present at at least 5 nM, for example, at least 50 nM, at least 100 nM, at least 300 nM, at least 450 nM, at least 475 nM, for example 5 nM-500 mM, 10 nM-100 mM, 50 nM-700 uM, 50 nM-10 uM, 100 nM-1000 nM, 350-650 nM or more preferably about 500 nM.
  • A83-01 may be added to the medium at a concentration of between 10 nM and 10 uM, or between 1 uM and 8 uM, or between 4 uM and 6 uM.
  • A83-01 may be added to the medium at about 5 uM.
  • the skilled person would know how to determine the concentration of other TGF beta inhibitors for use in the invention.
  • ErbB3/4 ligands are receptor tyrosine kinase ligands.
  • the culture media of the invention may further comprise one or more additional receptor tyrosine kinase ligands that are not themselves ErbB3/4 ligands.
  • An example of a receptor tyrosine kinase ligand for use in the invention is EGF, which is the ligand for the receptor tyrosine kinase EGFR.
  • Many receptor tyrosine kinase ligands are also mitogenic growth factors.
  • the culture media of the invention may comprise one or more mitogenic growth factor.
  • the one or more mitogenic growth factor may be selected from a family of growth factors comprising epidermal growth factor (EGF, Peprotech), Transforming Growth Factor-alpha (TGF-alpha, Peprotech), basic Fibroblast Growth Factor (bFGF, Peprotech), brain-derived neurotrophic factor (BDNF, R&D Systems), Platelet Derived Growth Factor (PDGF, Peprotech), amphiregulin (R&D Systems).
  • a preferred PDGF is PDGF-CC.
  • EGF is also referred to herein as “E”
  • FGF is also referred to herein as “F”.
  • the receptor tyrosine kinase ligand for use in the invention is one or more mitogenic growth factors selected from human EGF, human PDGF (e.g. human PDGF-CC), human amphiregulin and human TGF-alpha.
  • EGF is a potent mitogenic factor for a variety of cultured ectodermal and mesodermal cells and has a profound effect on the differentiation of specific cells in vivo and in vitro and of some fibroblasts in cell culture.
  • the EGF precursor exists as a membrane-bound molecule which is proteolytically cleaved to generate the 53-amino acid peptide hormone that stimulates cells.
  • the culture medium of the invention does not contain EGF.
  • the inventors found that the addition of EGF may be beneficial for growth of breast organoids. Therefore, in some embodiments, the culture media of the invention further comprises EGF.
  • EGF is preferably added to the basal culture medium at a concentration of between 1 and 500 ng/ml or of at least 1 and not higher than 500 ng/ml.
  • a preferred concentration is at least 1, 5, 10, 20, 25, 30, 40, 45, or 50 ng/ml and not higher than 500, 450, 400, 350, 300, 250, 200, 150, or 100 ng/ml.
  • a more preferred concentration is at least 1 and not higher than 100 ng/ml.
  • the same concentrations could be used for a different mitogenic growth factor, such as a PDGF or a FGF. If more than one FGF is used, for example FGF7 and FGF10, the concentration of a FGF is as defined above and refers to the total concentration of FGF used.
  • FGF7 and FGF10 are proteins that belong to the fibroblast growth factor (FGF) family of proteins. FGF family members possess broad mitogenic and cell survival activities, and are involved in a variety of biological processes, including embryonic development, cell growth, morphogenesis, tissue repair, tumor growth and invasion. FGFs stimulate cells by interacting with cell surface tyrosine kinase receptors (FGFR). Four closely related receptors (FGFR1-FGFR4) have been identified. FGFR1-FGFR3 genes have been shown to encode multiple isoforms, and these isoforms can be critical in determining ligand specificity. Most FGFs bind more than one receptor (Ornitz J Biol Chem. 1998 Feb. 27; 273 (9):5349-57).
  • FGF10 and FGF7 are unique among FGFs in that they interact only with a specific isoform of FGFR2, designated FGFR2b which is expressed exclusively by epithelial cells (Igarashi, J Biol Chem. 1998 273(21):13230-5).
  • the culture medium of the invention comprises a ligand of FGFR2b (e.g. FGF1, FGF3, FGF7, FGF10 or FGF22).
  • the ligand of FGFR2b is a high-affinity ligand of FGFR2b.
  • the ligand of FGFR2b binds to FGFR2b but does not bind to any other FGFR isoforms.
  • the FGFR2b ligand is a member of the FGF7 subfamily (e.g. FGF7, FGF10 or FGF22).
  • the FGFR2b ligand is FGF10 and/or FGF7.
  • the culture medium comprises FGF7 and FGF10.
  • biologically active fragments or variants of naturally occurring FGFR2b ligands for example, biologically active fragments or variants of FGF7 and/or FGF10 are used as FGFR2b ligands.
  • biologically active synthetic ligands are used as FGFR2b ligands in the culture medium of the invention.
  • Biologically active as used in this context means that the FGFR2b ligand, for example, the fragment or variant of a naturally occurring FGFR2b ligand, or a synthetic FGFR2b ligand, is capable of binding to FGFR2b and eliciting similar downstream signalling compared to FGF7 and/or FGF10, for example eliciting tyrosine phosphorylation of FRS2a (FGF receptor substrate 2a) or FRS2p (FGF receptor substrate 2p). Tyrosine phosphorylation of FRS2a and/or FRS2p is the earliest cytosolic event in the signalling pathways activated by FGF7 and/or FGF10.
  • the FGFR2b ligand is substituted with a compound that activates the FGFR2 or FGFR4 pathway (a “FGF-pathway activator”).
  • the one or more FGFR2b ligands used in the culture medium of the invention is selected from the following: human FGF7, human FGF10, human FGF22, human FGF1 or human FGF22.
  • a combination of mitogenic growth factors such as, for example, EGF and FGF7, or EGF and FGF10, is added to the basal culture medium.
  • a combination of mitogenic growth factors such as, for example, (i) FGF7 and FGF10, (ii) EGF, FGF7 and FGF10, or (iii) PDGF, FGF7 and FGF10, or (iv) EGF, PDGF, amphiregulin, FGF7 and FGF10 is added to the culture medium.
  • TGF-alpha or amphiregulin are added to the basal culture medium. These mitogenic growth factors may replace EGF. In some embodiments, hepatocyte growth factor (HGF) is added to the culture medium.
  • HGF hepatocyte growth factor
  • a combination of receptor tyrosine kinase ligands (e.g. EGF, FGF10 and FGF7) is preferably added to the culture medium when required, for example, daily or every other day. They may be added singularly or in combination. It is preferable that they are added every second day.
  • ROCK inhibitors such as Y-27632 (10 ⁇ M; Sigma)
  • Y-27632 10 ⁇ M
  • any of the media defined herein may additionally comprise a ROCK inhibitor for the first few days.
  • the culture medium of the invention additionally comprises a ROCK inhibitor, such as Y-27632, for example for the first few days of culture before performing cell sorting experiments.
  • a further embodiment a culture medium of the invention comprises a Rock (Rho-kinase) inhibitor.
  • a Rock inhibitor was found to prevent anoikis, especially when culturing single stem cells.
  • Said Rock inhibitor is preferably selected from R-(+)-trans-4-(1-aminoethyl)-N-(4-Pyridyl)cyclohexanecarboxamide dihydrochloride monohydrate (Y-27632, Sigma-Aldrich), 5-(1,4-diazepan-1-ylsulfonyl)isoquinoline (fasudil or HA1077, Cayman Chemical), and (S)-(+)-2-methyl-1-[(4-methyl-5-isoquinolinyl)sulfonyl]-hexahydro-1H-1,4-diazepine dihydrochloride (H-1 152, Tocris Bioschience).
  • Said Rho-kinase inhibitor for example Y-27632, is preferably added to the culture medium every second day during the first seven days of culturing said stem cells.
  • a Rock inhibitor is preferably included in the medium in the first few days e.g. for the first 1, 2, 3, 4, 5, 6 or 7 days of culture after single cell seeding or after a split. Any suitable concentration of the Rock inhibitor may be used, for example, 1-200 uM, 1-100 uM, 5-50 uM or approximately 10 uM.
  • a preferred concentration for Y27632 is 10 uM.
  • the invention provides a method for culturing stem cells and/or a method for obtaining an organoid wherein a Rock inhibitor is added to the culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day. In some embodiments, the Rock inhibitor is not added to the culture medium after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • the invention provides a method for obtaining an organoid, wherein the method comprises culturing stem cells, optionally single stem cells, wherein a Rock inhibitor is added to the culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day, and optionally not adding the Rock inhibitor to the culture medium after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • the invention provides a method for obtaining an organoid, wherein the method comprises culturing stem cells, optionally a tissue fragment, wherein the Rock inhibitor is not added to the culture medium either at all or after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • a Rock inhibitor may be added to the culture medium in the same way, meaning for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day, after the split, particularly when the split involves taking single stem cells from a first culture and placing these into a second culture. If the split involves taking multiple stem cells from the first culture and placing these into a second culture then addition of a Rock inhibitor is less important, and sometimes not necessary. Therefore, in some embodiments, wherein the method for obtaining organoids or for culturing stem cells involves a split, optionally where a single cell is involved in the split, a Rock inhibitor is added to the new culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day, after the split.
  • the method for obtaining organoids or for culturing stem cells involves a split, optionally where multiple cells are involved in the split, is not added to the culture medium either at all or after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • the culture medium of the invention further comprises a Notch agonist.
  • Notch signaling has been shown to play an important role in cell-fate determination, as well as in cell survival and proliferation.
  • Notch receptor proteins can interact with a number of surface-bound or secreted ligands, including but not limited to Delta 1, Jagged 1 and 2, and Delta-like 1, Delta-like 3, Delta-like 4.
  • Notch receptors Upon ligand binding, Notch receptors are activated by serial cleavage events involving members of the ADAM protease family, as well as an intramembranous cleavage regulated by the gamma secretase presenilin. The result is a translocation of the intracellular domain of Notch to the nucleus where it transcriptionally activates downstream genes.
  • a preferred Notch agonist is selected from Jagged 1 and Delta 1, or an active fragment or derivative thereof.
  • a most preferred Notch agonist is DSL peptide (Dontu et al., 2004. Breast Cancer Res 6. R605-R615) with the sequence CDDYYYGFGCNKFCRPR. Said DSL peptide is preferably used at a concentration between 10 ⁇ M and 100 nM or at least 10 ⁇ M and not higher than 100 nM.
  • the addition of a Notch agonist especially during the first week of culturing, increases the culture efficiency by a factor of 2-3.
  • Said Notch agonist is preferably added to the culture medium every second day during the first seven days of culturing said stem cells.
  • the invention provides a method for culturing stem cells and/or a method for obtaining an organoid wherein a Notch agonist is added to the culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day.
  • the Notch agonist is not added to the culture medium after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • a Notch agonist is defined as a molecule that stimulates a Notch activity in a cell by at least 10%, more preferred at least 20%, more preferred at least 30%, more preferred at least 50%, more preferred at least 70%, more preferred at least 90%, more preferred at least 100%, relative to a level of a Notch activity in the absence of said molecule.
  • a Notch activity can be determined by measuring the transcriptional activity of Notch, for example by a 4xwtCBFl-luciferase reporter construct as described (Hsieh et al, 1996 Mol Cell. Biol. 16, 952-959).
  • the culture medium optionally comprises Nicotinamide and is preferably supplemented with one or more (e.g., 1, 2, 3 or all) of the compounds selected from the group consisting of B27, N-acetylcysteine and N2.
  • the culture medium further comprises one or more components selected from the group consisting of: nicotinamide, B27, N2 and N-Acetylcysteine.
  • the culture medium further comprises B27, N-Acetylcysteine and Nicotinamide.
  • Nicotinamide is also referred to herein as “Nic”.
  • Nicotinamide is present at 7-15 mM, for example about 10 mM.
  • the B27 supplement is ‘B27 Supplement minus Vitamin A’ (available from Invitrogen, Carlsbad, Calif.; www.invitrogen.com; currently catalog no. 12587010; and from PAA Laboratories GmbH, Pasching, Austria; www.paa.com; catalog no.
  • F01-002; Brewer et al., J Neurosci Res., 35(5):567-76, 1993) may be used to formulate a culture medium that comprises biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin.
  • a culture medium that comprises biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin.
  • the B27 Supplement supplied by PAA Laboratories GmbH comes as a liquid 50 ⁇ concentrate, containing amongst other ingredients biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinol, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin.
  • these ingredients at least linolenic acid, retinol, retinyl acetate and tri-iodothyronine (T3) are nuclear hormone receptor agonists.
  • B27 Supplement may be added to a culture medium as a concentrate or diluted before addition to a culture medium. It may be used at a x final concentration or at other final concentrations.
  • Use of B27 Supplement is a convenient way to incorporate biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinol, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin into a culture medium of the invention. It is also envisaged that some or all of these components may be added separately to the culture medium instead of using the B27 Supplement. Thus, the culture medium may comprise some or all of these components.
  • retinoic acid is absent from the B27 Supplement used in the culture medium, and/or is absent from the culture medium.
  • N2 Supplement′ is available from Invitrogen, Carlsbad, Calif.; www.invitrogen.com; catalog no. 17502-048; and from PAA Laboratories GmbH, Pasching, Austria; www.paa.com; catalog no. F005-004; Bottenstein & Sato, PNAS, 76(1):514-517, 1979.
  • the N2 Supplement supplied by PAA Laboratories GmbH comes as a 100 ⁇ liquid concentrate, containing 500 ⁇ g/ml human transferrin, 500 ⁇ g/ml bovine insulin, 0.63 ⁇ g/ml progesterone, 1611 ⁇ g/ml putrescine, and 0.52 ⁇ g/ml sodium selenite.
  • N2 Supplement may be added to a culture medium as a concentrate or diluted before addition to a culture medium. It may be used at a x final concentration or at other final concentrations. Use of N2 Supplement is a convenient way to incorporate transferrin, insulin, progesterone, putrescine and sodium selenite into a culture medium of the invention. It is of course also envisaged that some or all of these components may be added separately to the culture medium instead of using the N2 Supplement. Thus, the culture medium may comprise some or all of these components.
  • the medium comprises B27
  • it does not also comprise N2.
  • the embodiments of the present invention can therefore be adapted to exclude N2 when B27 is present, if desired.
  • N2 is not present in the culture medium.
  • the medium comprises N2
  • it does not also comprise B27.
  • the embodiments of the present invention can therefore be adapted to exclude B27 when N2 is present, if desired.
  • B27 is not present in the culture medium.
  • the culture medium is supplemented with B27 and/or N2.
  • the basal medium is supplemented with 150 ng/ml to 250 ng/ml N-Acetylcysteine; preferably, the basal medium is supplemented with about 200 ng/ml N-Acetylcysteine.
  • the culture medium comprises progestin. In other embodiments the culture medium does not comprise progestin.
  • the culture medium comprises estradiol. In other embodiments the culture medium does not comprise estradiol.
  • the culture medium comprises hepatocyte growth factor. In other embodiments the culture medium does not comprise hepatocyte growth factor.
  • the culture medium comprises receptor activator of nuclear factor kappa-B ligand (RANKL). In other embodiments the culture medium does not comprise RANKL.
  • the culture medium of the invention is supplemented with nicotinamide. Addition of nicotinamide has been found to improve culture efficiency and lifespan of human organoids. Nicotinamide may be added to the culture medium to a final concentration of between 1 and 100 mM, between 5 and 50 mM, or preferably between 5 and 20 mM. For example, nicotinamide may be added to the culture medium to a final concentration of approximately 10 mM.
  • the culture medium of the invention comprises a p53 stabilizing agent.
  • a p53 stabilizing agent may be added to the culture medium in order to ensure that the cell population is predominantly tumour cells.
  • p53 stabilizing agents increase the cellular concentration of p53 (e.g. by blocking the interaction between p53 and Mdm2), stimulate p53-dependent expression and induce cellular senescence.
  • p53 mutations commonly occur in tumour cells, which may result in the mutant p53 protein being unable to be stabilised by a p53 stabilizing agent.
  • a preferred p53 stabilizing agent is a member of the Nutlin family, e.g. Nutlin-1, Nutlin-2 or Nutlin-3.
  • the p53 stabilizing agent is Nutlin-3. Any suitable concentration of the p53 stabilizing agent may be used, for example between 1 nM and 10 mM, between 10 nM and 1 mM, between 100 nM and 100 ⁇ M, between 1 ⁇ M and 10 ⁇ M.
  • the culture medium used in the method advantageously comprises a p53 stabilizing agent.
  • the p53 stabilizing agent may be present in the culture medium when the method is used for culturing normal breast epithelial stem cells, although it is not required.
  • the culture medium of the invention comprises a p38 MAP kinase inhibitor.
  • a p38 MAP kinase inhibitor is an inhibitor that directly or indirectly negatively regulates p38 signalling.
  • the p38 MAP kinase inhibitor is SB202190.
  • other suitable p38 MAP kinase inhibitors may alternatively be used and these are readily available to the skilled person.
  • any suitable pH may be used.
  • the pH of the medium may be in the range from about 7.0 to 7.8, in the range from about 7.2 to 7.6, or about 7.4.
  • the pH may be maintained using a buffer.
  • a suitable buffer can readily be selected by the skilled person. Buffers that may be used include carbonate buffers (e.g. NaHCO 3 ), and phosphates (e.g. NaH 2 PO 4 ). These buffers are generally used at about 50 to about 500 mg/l.
  • a culture medium may comprise a pH indicator, such as phenol red, to enable the pH status of the medium to be easily monitored (e.g. at about 5 to about 50 mg/litre).
  • a pH indicator such as phenol red
  • a culture medium for use in the invention may comprise one or more amino acids.
  • Amino acids which may be present include L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine and combinations thereof.
  • each amino acid when present is present at about 0.001 to about 1 g/L of medium (usually at about 0.01 to about 0.15 g/L), except for L-glutamine which is present at about 0.05 to about 1 g/L (usually about 0.1 to about 0.75 g/L).
  • the amino acids may be of synthetic origin.
  • a culture medium for use in the invention may comprise one or more vitamins.
  • Vitamins which may be present include thiamine (vitamin B1), riboflavin (vitamin B2), niacin (vitamin B3), D-calcium pantothenate (vitamin B5), pyridoxal/pyridoxamine/pyridoxine (vitamin B6), folic acid (vitamin B9), cyanocobalamin (vitamin B12), ascorbic acid (vitamin C), calciferol (vitamin D2), DL-alpha tocopherol (vitamin E), biotin (vitamin H) and menadione (vitamin K).
  • a culture medium for use in the invention may comprise one or more inorganic salts.
  • inorganic salts are typically included in culture media to aid maintenance of the osmotic balance of the cells and to help regulate membrane potential.
  • Inorganic salts which may be present include salts of calcium, copper, iron, magnesium, potassium, sodium, zinc. The salts are normally used in the form of chlorides, phosphates, sulphates, nitrates and bicarbonates.
  • Specific salts that may be used include CaCl 2 , CuSO 4 -5H 2 O, Fe(NO 3 )-9H 2 O, FeSO 4 -7H 2 O, MgCl, MgSO 4 , KCl, NaHCO 3 , NaCl, Na 2 HPO 4 , Na 2 HPO 4 —H 2 O and ZnSO 4 -7H 2 O.
  • the osmolarity of the medium may be in the range from about 200 to about 400 mOsm/kg, in the range from about 290 to about 350 mOsm/kg, or in the range from about 280 to about 310 mOsm/kg.
  • the osmolarity of the medium may be less than about 300 mOsm/kg (e.g. about 280 mOsm/kg).
  • a culture medium for use in the invention may comprise a carbon energy source, in the form of one or more sugars.
  • a carbon energy source in the form of one or more sugars.
  • Sugars which may be present include glucose, galactose, maltose and fructose.
  • the sugar is preferably glucose, particularly D-glucose (dextrose).
  • a carbon energy source will normally be present at between about 1 and about 10 g/L.
  • a culture medium of the invention may contain serum.
  • Serum obtained from any appropriate source may be used, including fetal bovine serum (FBS), goat serum or human serum.
  • FBS fetal bovine serum
  • goat serum human serum.
  • human serum is used.
  • Serum may be used at between about 1% and about 30% by volume of the medium, according to conventional techniques.
  • a culture medium of the invention may contain a serum replacement.
  • a serum replacement Various different serum replacement formulations are commercially available and are known to the skilled person. Where a serum replacement is used, it may be used at between about 1% and about 30% by volume of the medium, according to conventional techniques.
  • a culture medium of the invention may be serum-free and/or serum replacement-free.
  • a serum-free medium is one that contains no animal serum of any type. Serum-free media may be preferred to avoid possible xeno-contamination of the stem cells.
  • a serum replacement-free medium is one that has not been supplemented with any commercial serum replacement formulation.
  • the cell culture medium is supplemented with a purified, natural, semi-synthetic and/or synthetic growth factor and does not comprise an undefined component, such as fetal bovine serum or fetal calf serum.
  • a purified, natural, semi-synthetic and/or synthetic growth factor and does not comprise an undefined component, such as fetal bovine serum or fetal calf serum.
  • supplements such as B27 (Invitrogen), N-Acetylcysteine (Sigma) and N2 (Invitrogen) stimulate proliferation of some cells.
  • the cell culture medium is supplemented with one or more of these supplements, for example one, any two or all three of these supplements.
  • a culture medium for use in the invention may comprise one or more trace elements, such as ions of barium, bromium, cobalt, iodine, manganese, chromium, copper, nickel, selenium, vanadium, titanium, germanium, molybdenum, silicon, iron, fluorine, silver, rubidium, tin, zirconium, cadmium, zinc and/or aluminium.
  • trace elements such as ions of barium, bromium, cobalt, iodine, manganese, chromium, copper, nickel, selenium, vanadium, titanium, germanium, molybdenum, silicon, iron, fluorine, silver, rubidium, tin, zirconium, cadmium, zinc and/or aluminium.
  • the medium may comprise a reducing agent, such as beta-mercaptoethanol at a concentration of about 0.1 mM.
  • a reducing agent such as beta-mercaptoethanol at a concentration of about 0.1 mM.
  • a culture medium of the invention may comprise one or more additional agents, such as nutrients or growth factors previously reported to improve stem cell culture, such as cholesterol/transferrin/albumin/insulin/progesterone, putrescine, selenite/other factors.
  • additional agents such as nutrients or growth factors previously reported to improve stem cell culture, such as cholesterol/transferrin/albumin/insulin/progesterone, putrescine, selenite/other factors.
  • the culture medium of the invention comprises recombinant human neuregulin ⁇ -1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7 and FGF10.
  • an Rspondin e.g. Rspondin 1
  • Noggin Noggin
  • B27 N-Acetylcysteine
  • Nicotinamide a ROCK 1,2 inhibitor
  • ALK 4, 5, 7 inhibitor for example, A83-01
  • a p38 MAP Kinase inhibitor for example, SB 202190
  • the culture medium of the invention comprises recombinant human neuregulin ⁇ -1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10 and PDGF-CC.
  • an Rspondin e.g. Rspondin 1
  • Noggin for example, B27, N-Acetylcysteine
  • Nicotinamide for example, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10 and PDGF-CC.
  • the culture medium of the invention comprises recombinant human neuregulin ⁇ -1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10 and EGF.
  • an Rspondin e.g. Rspondin 1
  • Noggin for example, B27, N-Acetylcysteine
  • Nicotinamide for example, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190)
  • FGF7 for example, FGF10 and EGF.
  • the culture medium of the invention comprises recombinant human neuregulin ⁇ -1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10, amphiregulin, TGF-alpha and EGF.
  • an Rspondin e.g. Rspondin 1
  • Noggin for example, B27, N-Acetylcysteine
  • Nicotinamide for example, Y-27632
  • a ROCK 1,2 inhibitor for example, Y-27632
  • ALK 4, 5, 7 inhibitor for example, A83-01
  • a p38 MAP Kinase inhibitor for example, SB 202190
  • FGF7, FGF10 amphiregulin
  • the method for culturing epithelial stem cells comprises culturing one or more epithelial stem cells in contact with an extracellular matrix. Any suitable extracellular matrix may be used.
  • Isolated epithelial stem cells are preferably cultured in a microenvironment that mimics at least in part a cellular niche in which said stem cells naturally reside. This cellular niche may be mimicked by culturing said stem cells in the presence of biomaterials, such as an extracellular matrix that provides key regulatory signals controlling stem cell fate.
  • a cellular niche is in part determined by the stem cells and surrounding cells, and the extracellular matrix (ECM) that is produced by the cells in said niche.
  • ECM extracellular matrix
  • epithelial stem cells are cultured in contact with an ECM. “In contact” means a physical or mechanical or chemical contact, which means that for separating said resulting organoid or population of epithelial stem cells from said extracellular matrix a force needs to be used.
  • the epithelial stem cells are embedded in the ECM.
  • a culture medium of the invention may be diffused into an extracellular matrix (ECM).
  • ECM extracellular matrix
  • isolated tissue fragments or isolated epithelial stem cells are attached to an ECM.
  • ECM is composed of a variety of polysaccharides, water, elastin, and glycoproteins, wherein the glycoproteins comprise collagen, entactin (nidogen), fibronectin, and laminin.
  • ECM is secreted by epithelial cells, endothelial cells, parietal endoderm-like cells (e.g. Englebreth-Holm-Swarm Parietal Endoderm-Like cells described in Hayashi et al. (2004) Matrix Biology 23:47-62) and connective tissue cells.
  • ECM epithelial cells, endothelial cells, parietal endoderm-like cells or fibroblast cells, in a receptacle, prior to the removal of these cells and the addition of isolated tissue fragments or isolated epithelial stem cells.
  • ECM-producing cells such as for example epithelial cells, endothelial cells, parietal endoderm-like cells or fibroblast cells
  • extracellular matrix-producing cells examples include chondrocytes, producing mainly collagen and proteoglycans, fibroblast cells, producing mainly type IV collagen, laminin, interstitial procollagens, and fibronectin, and colonic myofibroblasts producing mainly collagens (type I, III, and V), chondroitin sulfate proteoglycan, hyaluronic acid, fibronectin, and tenascin-C.
  • said ECM is commercially provided.
  • Examples of commercially available extracellular matrices are extracellular matrix proteins (Invitrogen) and basement membrane preparations from Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells (e.g.
  • a synthetic extracellular matrix material such as ProNectin (Sigma Z378666) may be used. Mixtures of extracellular matrix materials may be used, if desired.
  • ECM extracellular matrix material for culturing stem cells
  • the use of an ECM for culturing stem cells enhanced long-term survival of the stem cells and the continued presence of undifferentiated stem cells. In the absence of an ECM, stem cell cultures could not be cultured for longer periods and no continued presence of undifferentiated stem cells was observed. In addition, the presence of an ECM allowed culturing of three-dimensional tissue organoids, which could not be cultured in the absence of an ECM.
  • the extracellular matrix material will normally be a drop on the bottom of the dish in which cells are suspended.
  • the medium is added and diffuses into the ECM.
  • the cells in the medium stick to the ECM by interaction with its surface structure, for example interaction with integrins.
  • An example of an ECM for use in a method of the invention comprises at least one glycoprotein, such as laminin.
  • a preferred ECM for use in a method of the invention comprises at least two distinct glycoproteins, such as two different types of collagen or a collagen and laminin.
  • the ECM can be a synthetic hydrogel extracellular matrix or a naturally occurring ECM.
  • a further preferred ECM comprises laminin, entactin, and collagen IV.
  • a further preferred ECM is provided by MatrigelTM (BD Biosciences), which comprises laminin, entactin, and collagen IV.
  • the extracellular matrix is a laminin-containing extracellular matrix such as MatrigelTM (BD Biosciences).
  • the ECM comprises laminin, entactin, collagen IV and heparin sulphate proteoglycan (e.g. Cultrex® Basement Membrane Extract Type 2 (Trevigen, Inc.)).
  • the single stem cell, population of cells, or tissue fragment is embedded in matrigel, which is optionally growth factor reduced and/or phenol red-free.
  • the culture medium is placed on top of the ECM.
  • the culture medium can then be removed and replenished as and when required.
  • the culture medium is replenished every 1, 2, 3, 4, 5, 6 or 7 days. If components are “added” or “removed” from the media, then this can in some embodiments mean that the media itself is removed from the ECM and then a new media containing the “added” component or with the “removed” component excluded is placed on the ECM.
  • the culture medium of the invention is in contact with an extracellular matrix or a 3D matrix that mimics the extracellular matrix by its interaction with the cellular membrane proteins, such as integrins.
  • a culture medium of the invention and an extracellular matrix, e.g. supplied as a kit.
  • the culture medium of the invention is suitable for culturing adult breast stem cells. In some embodiments, the culture medium of the invention is suitable for obtaining breast organoids. In some embodiments, the culture medium of the invention is suitable for expanding a population of breast stem cells for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or more than 14 passages.
  • the invention provides a culture medium as described herein that comprises one or more ErbB3/4 ligands that is suitable for expanding a population of breast stem cells for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or more than 14 passages.
  • the culture medium of the invention is suitable for establishing populations of successful breast organoids in at least 50%, at least 60%, at least 70%, at least 80% or at least 90% of inoculations.
  • a successful breast organoid is defined herein as an organoid that is capable of being passaged at least four times.
  • the invention provides a culture medium that comprises one or more ErbB3/4 ligands that is suitable for establishing populations of successful organoids with an efficiency of at least 50%, at least 60%, at least 70%, at least 80% or at least 90%.
  • the invention provides the use of the culture medium of the invention for expanding an epithelial stem cell, population of epithelial stem cells, tissue fragment or organoid.
  • the invention also provides the use of a culture medium of the invention for expanding a breast epithelial stem cell, population of breast epithelial stem cells, breast tissue fragment or breast organoid.
  • the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a normal tissue.
  • the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a non-cancerous tissue.
  • the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a diseased tissue, for example a cancerous tissue, such as from an adenoma, a fibroadenoma, an adenocarcinoma, a carcinoma, a sarcoma, circulating tumour cells or metastasized cancer.
  • the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a benign or malignant breast tumour.
  • the stem cell, population of stem cells, tissue fragment or organoid is or comprises a breast cancer cell(s).
  • the stem cell, population of stem cells, or the tissue fragment is a biopsy from a breast tumour. The tumour is preferably a breast tumour by origin rather than being a metastatic tumour originating from a different tissue.
  • the epithelial stem cells to be cultured in the method of the invention and/or from which the organoids are derived are obtained from adult tissue, i.e. the epithelial stem cells are adult epithelial stem cells.
  • adult means mature tissue, i.e. includes newly-born baby or child but excludes embryonic or foetal.
  • the epithelial stem cells are not derived from embryonic stem cells or embryonic stem cell lines, e.g. which have been differentiated in vitro, for example human embryonic stem cells or human embryonic stem cell lines.
  • the cells and tissues are preferably mammalian cells and tissues. More preferably, the cells and tissues are human cells and tissues. In some embodiments they are from other mammals, for example from a laboratory animal (e.g. mouse, rabbit, rat, guinea pig), a companion animal (e.g. dog, cat, horse) or a farm animal (e.g. cow, pig, sheep, goat).
  • a laboratory animal e.g. mouse, rabbit, rat, guinea pig
  • a companion animal e.g. dog, cat, horse
  • a farm animal e.g. cow, pig, sheep, goat.
  • the epithelial stem cells are primary epithelial stem cells.
  • Primary cells represent the best experimental models for in vivo situations.
  • the epithelial stem cells are (or are derived from) primary epithelial stem cells.
  • Primary cell cultures can be passaged to form secondary cell cultures. With the exception of cancer cells, traditional secondary cell cultures have a limited lifespan. After a certain number of population doublings (e.g. 50-100 generations) cells undergo the process of senescence and stop dividing. Cells from secondary cultures can become immortalized to become continuous cell lines. Immortalization can occur spontaneously, or may be virally- or chemically-induced.
  • Immortalized cell lines are also known as transformed cells.
  • the methods of the present invention allow continuous passaging of epithelial stem cells without immortalisation or transformation.
  • the epithelial stem cells are not immortalised or transformed cells or are not derived from an immortalised cell line or a transformed cell line.
  • An advantage of the present invention is that the epithelial stem cells, undergoing multiple rounds of expansion and passaging, retain the characteristics of primary cells and have minimal or no genotypic or phenotypic changes.
  • the epithelial stem cells may be obtained by any suitable method.
  • cells are isolated by collagenase digestion, for example, as described in the examples and in Dorell et al., 2008 (Hepatology. 2008 October; 48(4):1282-91. Surface markers for the murine oval cell response. Dorrell C, Erker L, Lanxon-Cookson K M, Abraham S L, Victoroff T, Ro S, Canaday P S, Streeter P R, Grompe M).
  • collagenase digestion is performed on a tissue biopsy.
  • collagenase and accutase digestion are used to obtain the epithelial stem cells for use in the invention.
  • breast tissue may be minced, washed (for example in basal culture medium) and incubated in a culture medium of the invention supplemented with a protease (for example, collagenase, dispase or trypsin) (see, for example, Example 1).
  • the protease may be at any suitable concentration, for example, between 0.1 and 10 mg/ml, between 0.1 and 1 mg/ml or between 1 and 10 mg/ml.
  • the protease may be present at between 1 and 2 mg/ml.
  • Incubation in the culture medium supplemented with a protease may be for any suitable length of time.
  • the tissue may be incubated in the medium for between 10 minutes and 10 hours, between 20 minutes and 5 hours, between 30 minutes and 5 hours, between 30 minutes and 3 hours, between 30 minutes and 2 hours, between 1 hour and 5 hours, between 1 hour and 3 hours or between 1 hour and 2 hours.
  • the tissue is incubated in the medium for at least 10 minutes, at least 20 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 3 hours or at least 5 hours.
  • tissue fragments contained in the resultant digested tissue suspension may then be further reduced in size.
  • the suspension may be sheared. Filtration and/or centrifugation steps may be used to isolate tissue fragments or isolated cells.
  • the isolated tissue fragments or isolated cells may then be cultured in a culture medium of the invention.
  • the method comprises culturing a fragment of tissue which comprises epithelium.
  • the epithelial stem cells are isolated from a tissue fragment.
  • the epithelial stem cells are obtained for culturing based on the expression of epithelial cell surface markers, such as EPCAM, MUC-1, KRT-5, KRT8 and KRT18.
  • a method of the invention therefore comprises preparing a cell suspension from said epithelial tissue as described above, contacting said cell suspension with an epithelial cell surface marker binding compound (e.g. EPCAM, MUC-1, KRT-5, KRT8 and/or KRT18 binding compounds), isolating the epithelial marker binding compound, and isolating the cells from said binding compound.
  • an epithelial cell surface marker binding compound e.g. EPCAM, MUC-1, KRT-5, KRT8 and/or KRT18 binding compounds
  • epithelial stem cells are obtained for culturing based on the expression of Lgr5 and/or Lgr6 on the epithelial stem cell surface; these proteins belong to the large G protein-coupled receptor (GPCR) superfamily (see, for example, WO 2009/022907, the contents of which are incorporated herein in their entirety).
  • GPCR G protein-coupled receptor
  • the Lgr subfamily is unique in carrying a large leucine-rich ectodomain important for ligand binding.
  • a method of the invention therefore comprises preparing a cell suspension from said epithelial tissue as described above, contacting said cell suspension with an Lgr5 and/or 6 binding compound (such as an antibody, e.g. an anti-Lgr5 monoclonal antibody, e.g. as described in WO 2009/022907), isolating the Lgr5 and/or 6 binding compound, and isolating the stem cells from said binding compound.
  • an Lgr5 and/or 6 binding compound such
  • An organoid is preferably obtained using a cell from an adult tissue, preferably an epithelial stem cell from an adult tissue.
  • the epithelial stem cells are normal cells.
  • the epithelial stem cells are cancer stem cells.
  • the stem cells may be Lgr5 positive cancer stem cells. Accordingly, the cells may be obtained from a tumour, if required.
  • the epithelial stem cells are diseased stem cells, for example stem cells infected with intracellular pathogens (e.g. bacteria, viruses or parasites).
  • Binding compounds that bind to breast cancer cell markers may be used for the isolation of breast cancer stem cells for culturing in a method of the invention.
  • breast cancer cell markers include Epithelial Membrane Antigen, Cancer antigen 15-3 (CA 15-3), cancer antigen 27.29 (CA 27.29), carcinoembryonic antigen (CEA), Urokinase plasminogen activator (uPA), plasminogen activator inhibitor (PAI-1), Podocalyxin, EZH2 and kallikrein-10.
  • Epithelial Membrane Antigen (EMA) binding compounds e.g. anti-EMA antibodies
  • EMA Epithelial Membrane Antigen
  • a method of the invention therefore comprises preparing a cell suspension from primary or secondary cancerous tissue, contacting said cell suspension with a breast cancer cell marker binding compound (e.g. EMA binding compounds), isolating the breast cancer cell marker binding compound and isolating the cells from said binding compound.
  • a breast cancer cell marker binding compound e.g. EMA binding compounds
  • an organoid originates from a single cell, optionally expressing Lgr5.
  • the single cell comprises a nucleic acid construct comprising a nucleic acid molecule of interest.
  • Lgr5 and/or 6 binding compounds comprise antibodies, such as monoclonal antibodies that specifically recognize and bind to the extracellular domain of either Lgr5 or Lgr6, such as monoclonal antibodies including mouse and rat monoclonal antibodies (see, for example, WO 2010/016766, the contents of which are incorporated herein in their entirety).
  • Lgr5 and/or Lgr6-expressing stem cells can be isolated, for example with the aid of magnetic beads or through fluorescence-activated cell sorting, as is clear to a skilled person.
  • Using a method of the invention it is possible to isolate one single Lgr5 and/or Lgr6 expressing cell and to apply a method of the invention to it.
  • An organoid or a population of breast epithelial stem cells may therefore be derived from one single cell. Accordingly, in some embodiments, the starting cell to be cultured is a single cell.
  • a population of cells may be used as the starting point, for example, a population of cells contained in a breast tissue fragment as described above.
  • the methods of the invention are not restricted to using single cells as the starting point.
  • a method for obtaining an organoid comprising culturing breast epithelial stem cells in a culture medium of the invention.
  • the method comprises culturing the breast epithelial stem cells in a culture medium of the invention using a culture method as described herein.
  • the method comprises culturing the epithelial stem cells or obtaining the organoid/population of adult epithelial stem cells from a single cell.
  • this allows a homogenous population of cells to form.
  • the method comprises culturing the stem cells in a culture medium of the invention for a period of time, for example, 3 days to 10 weeks, 1 to 10 weeks, 1 to 4 weeks or 10 days to 3 weeks, and then passaging the cells (e.g. dissociating the cells to a single cell density, seeding one or more cells at a ratio of 1 cell per container (e.g.
  • a culture medium of the invention for a period of time, for example, 3 days to 10 weeks, 1 to 10 weeks, 1 to 4 weeks or 10 days to 3 weeks and repeating the passaging and expanding steps at least once, at least twice, at least three times, at least four times, at least five times, at least six times, at least seven times, at least eight times, at least nine times, at least ten times, at least eleven times, at least twelve times, at least thirteen times or at least fourteen times.
  • the method may further comprise obtaining and/or isolating one or more epithelial stem cells or an organoid.
  • the stem cells may be useful to remove one or more stem cells and/or one or more organoids cultured in the culture medium from the culture medium for use in subsequent applications.
  • Any one of a number of physical methods of separation known in the art may be used to select the cells of the invention and distinguish these from other cell types. Such physical methods may involve FACS and various immuno-affinity methods based upon makers specifically expressed by the cells of the invention.
  • LGR5 is a cell marker that may be expressed in the cells of the invention. Therefore, by way of illustration only, the cells of the invention may be isolated by a number of physical methods of separation, which rely on the presence of this marker. Similarly, any of the other markers expressed by the cells may be used.
  • the cells of the invention may be isolated by FACS utilizing an antibody, for example, against one of these markers.
  • an antibody for example, against one of these markers.
  • this may be achieved through a fluorescent labeled antibody, or through a fluorescent labeled secondary antibody with binding specificity for the primary antibody.
  • suitable fluorescent labels includes, but is not limited to, FITC, Alexa Fluor® 488, GFP, CFSE, CFDA-SE, DyLight 488, PE, PerCP, PE-Alexa Fluor® 700, PE-Cy5 (TRI-COLOR®), PE-Cy5.5, PI, PE-Alexa Fluor® 750, and PE-Cy7. This list is provided by way of example only, and is not intended to be limiting.
  • FACS analysis using an anti-Lgr5 antibody may provide a purified cell population.
  • the cells of the invention may be isolated by immuno-affinity purification, which is a separation method well known in the art.
  • the cells of the invention may be isolated by immuno-affinity purification directed towards c-kit.
  • this method relies upon the immobilisation of antibodies on a purification column. The cell sample is then loaded onto the column, allowing the appropriate cells to be bound by the antibodies, and therefore bound to the column. Following a washing step, the cells are eluted from the column using a competitor which binds preferentially to the immobilised anti-c-kit antibody, and permits the cells to be released from the column.
  • immuno-affinity purification using an immobilised antibody will provide a purified cell population.
  • the other identifiable markers for example Sox9, Slug, CD44 and/or ALDH1
  • the cells may be purified through a FACS step using an anti-Lgr5 antibody, followed by an immuno-affinity purification step using a SSEA-1 affinity column.
  • the cells may be cultured after isolation for at least about 15, at least about 20 days, at least about 25 days, or at least about 30 days.
  • the cells are expanded in culture longer to improve the homogeneity of the cell phenotype in the cell population.
  • Single-cell suspensions or small clusters of cells (2-50 cells/cluster) will normally be seeded, rather than large clusters of cells, as in known in the art. As they divide, such cells will be seeded onto a support at a density that promotes cell proliferation.
  • the plating density of at least 1-500 cells/well is used, the surface of the well is 0.32 cm 2 .
  • clusters are seeded the plating density is preferably 250-2500 cells/cm 2 .
  • a density of between about 2500 cells/cm 2 and about 5,000 cells/cm 2 may be used in some embodiments.
  • single-cell suspensions or small cluster of cells will normally be seeded, rather than large clusters of cells, as in known in the art.
  • the invention provides a population of cells or one or more organoids comprising said stem cells that have been generated or obtained by culturing stem cells or tissue fragments according to the invention, which have been cultured for at least 2 months, at least 3 months, preferably at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 9 months, or at least 12 months or more, wherein the stem cells have been passaged approximately every 1 to 4 weeks.
  • a ‘population’ of cells is any number of cells greater than 1, but is preferably at least 1 ⁇ 10 3 cells, at least 1 ⁇ 10 4 cells, at least 1 ⁇ 10 5 cells, at least 1 ⁇ 10 6 cells, at least 1 ⁇ 10 7 cells, at least 1 ⁇ 10 8 cells, or at least 1 ⁇ 10 9 cells.
  • the stem cells or progenitor cells of the invention cultured according to the invention may be human stem cells or human progenitor cells.
  • the stem cells of the invention cultured according to the invention may be epithelial stem cells or epithelial progenitor cells.
  • the stem cells of the invention and/or cultured according to the invention are not embryonic stem cells. In some embodiments the stem cells of the invention and/or cultured according to the invention are not human embryonic stem cells. Preferably, the stem cells of the invention are adult stem cells.
  • the stem cells are human breast epithelial stem cells.
  • Human epithelial stem cells include stem cells of human epithelial tissue origin. Epithelial stem cells are able to form the distinct cell types of which the epithelium is composed.
  • the organoids are human breast organoids.
  • the epithelial stem cell is a cancer cell or a non-cancerous tumour cell e.g. is derived from a tumour.
  • the cancer cell is a circulating tumour cell.
  • the cancer cell is not a circulating tumour cell.
  • the term “cancer” it is to be understood that it applies equally to non-cancerous (benign) tumours. Accordingly, in some embodiments the cell is derived from a malignant tumour or from a metastasis. In some embodiments, the cell is derived from a benign tumour.
  • Cancer cells tend to have mutations that constitutively activate or deactivate certain growth pathways and which mean that certain factors in the culture medium that may normally be required for growth, are no longer necessary. For example, some cancers result in constitutive activation of the Wnt pathway. In such cases, a culture medium may not require an Lgr5 agonist and/or may not require a Wnt agonist, but one or both of these may still be present in the culture medium. Other mutations would allow other factors to be left out of the culture media described herein. Other epithelial cancers (carcinomas) or non-cancerous tumours (e.g. adenomas) can also be grown in culture media of the invention.
  • a cancer organoid obtained from cancer stem cells is grown in a culture medium of the invention that is suitable for growth of the corresponding normal tissue organoid obtained from normal stem cells, optionally with certain factors excluded from the medium.
  • a breast cancer organoid obtained by culturing breast cancer stem cells may be grown in the same culture conditions as a normal breast organoid obtained by culturing breast epithelial stem cells, optionally with certain factors excluded from the medium.
  • the normal tissue medium should allow cancers with all genetic backgrounds to grow, without excluding any particular cancer mutations. The methods of the invention for culturing stem cells can be practised accordingly.
  • the cancer organoid is grown in a culture medium of the invention that further comprises a p53 stabilizing agent.
  • a p53 stabilizing agent may be added to the culture medium in order to ensure that the cell population is predominantly tumour cells.
  • the methods of the invention for culturing stem cells can be practised accordingly.
  • the invention provides a method for culturing a single breast epithelial stem cell, a population of breast epithelial stem cells, or an isolated breast tissue fragment, preferably to generate an organoid, wherein the method comprises:
  • the invention also provides a method for expanding breast epithelial stem cells, wherein the method comprises:
  • the cells are expanded to generate one or more (e.g. at least 2, 3, 4, 5, 6, 10, 15, 20 or more than 20) organoids.
  • the culture medium used in the methods of the invention is a culture medium according to the invention.
  • a method for ‘expanding’ a population of cells or isolated tissue fragments is one that involves maintaining or increasing the number of stem cells in an initial population to generate an expanded population of stem cells which retain their undifferentiated phenotype and self-renewing properties.
  • a breast organoid comprising culturing stem cells or tissue fragments comprising said stem cells in a culture medium of the invention.
  • the invention provides a method for expanding a single breast stem cell or a population of breast stem cells, preferably to generate an organoid, wherein the method comprises culturing the single stem cell or population of stem cells in a culture medium according to the invention.
  • the organoid may be a normal organoid or it may be a cancer organoid, for example, a carcinoma organoid or an adenocarcinoma organoid, or a benign tumour organoid, for example.
  • the method for expanding a single stem cell or a population of stem cells, preferably to generate an organoid comprises expanding the single stem cell, population of stem cells or tissue fragment in a culture medium according to the invention.
  • the invention provides a method for expanding a single breast epithelial stem cell or a population of breast epithelial stem cells, preferably to generate an organoid, wherein the method comprises:
  • the method comprises bringing the stem cell, the population of stem cells or the isolated tissue fragment and the culture medium into contact with an extracellular matrix or a 3D matrix as described herein, for example, a 3D matrix that mimics the extracellular matrix by its interaction with the cellular membrane proteins such as integrins, for example a laminin-containing extracellular matrix such as MatrigelTM (BD Biosciences) or Cultrex® Basement Membrane Extract (Trevigen, Inc.).
  • the culture medium is diffused into the extracellular matrix.
  • the invention provides methods for culturing breast epithelial stem cells.
  • the invention further provides methods for expanding breast epithelial stem cells and methods for obtaining breast organoids.
  • expanding stem cells is synonymous with “obtaining breast organoids”.
  • the method comprises bringing the stem cell, the population of stem cells or the isolated tissue fragment and the culture medium into contact with an extracellular matrix or a 3D matrix as described herein, for example, a 3D matrix that mimics the extracellular matrix by its interaction with the cellular membrane proteins such as integrins, for example a laminin-containing extracellular matrix such as Cultrex® Basement Membrane Extract (Trevigen, Inc.) or MatrigelTM (BD Biosciences).
  • the culture medium is diffused into the extracellular matrix.
  • the tissue fragments or isolated cells are cultured in suspension plates. In some embodiments, the tissue fragments or isolated cells are cultured at between 35° C. and 39° C. or between 36° C. and 38° C. For example, the tissue fragments or isolated cells may be cultured at approximately 37° C.
  • the tissue fragments or isolated cells are cultured at ambient levels of Oz.
  • the level of Oz may be, for example, between 0.5% and 4%, between 1% and 4%, between 1% and 3% or between 2% and 4%.
  • the tissue fragments or isolated cells may be cultured under conditions of approximately 2% Oz.
  • the tissue fragments or isolated cells may be cultured under conditions of between 3% and 7% CO 2 or between 4% and 6% CO 2 .
  • the tissue fragments or isolated cells may be cultured under conditions of approximately 5% CO 2 .
  • the culture medium may be changed at any suitable interval, for example, every 1-7 days, every 2-6 days or every 3-5 days.
  • the culture medium may be changed every 4 days.
  • Organoids may be passaged at any suitable interval, for example, every 3 days to every 10 weeks, every 1 to 10 weeks, every 1 to 4 weeks or every 10 days to 3 weeks. In some embodiments, the organoids are passaged approximately every 10 days to 3 weeks.
  • the timing for passaging of organoids is determined by the organoids reaching a particular size (e.g. an average size of approximately 100-200 ⁇ m) and/or a reduction in viability which can manifest in the loss of integrity of the epithelium, the organoids having a dense appearance and/or the presence of dead cells.
  • a particular size e.g. an average size of approximately 100-200 ⁇ m
  • a reduction in viability which can manifest in the loss of integrity of the epithelium, the organoids having a dense appearance and/or the presence of dead cells.
  • the invention provides an organoid or a population of epithelial stem cells obtainable or obtained by a method of the invention.
  • the method further comprises obtaining and/or isolating an organoid.
  • the organoid or population of epithelial stem cells of the invention comprises breast cells.
  • a breast organoid which comprises a population breast epithelial stem cells.
  • the organoid or population of epithelial stem cells of the invention consists of breast cells.
  • Pasic et al. did not test a culture medium comprising an ErbB3/4 ligand combined with an Lgr5 agonist or a FGFR2b ligand.
  • Use of the Pasic et al. culture system results in the formation of cellular structures that have a morphology that is distinct from that of the organoids of the present invention.
  • cellular structures have a budding morphology in which the structures have a rough edge, whereas the organoids obtained using the methods of the present invention do not have this morphology but instead have a smooth cyst-like structure or a morphology that is similar to a cluster of grapes.
  • Cultures using the Pasic et al. system have been observed to stop expanding after 2-3 passages (see Example 4 herein), whereas the inventors have found that in some embodiments using a method of the invention it is possible to expand the breast epithelial stem cells for more than 14 passages.
  • Yu uses a culture method which comprises EGF and FGF2. They do not use culture media comprising an ErbB3/4 ligand in combination with an Lgr5 agonist or a FGFR2b ligand, as is used in the present invention. Indeed, they do not use any culture media comprising an ErbB3/4 ligand.
  • Yu's method results in formation of aggregates of single tumour cells without strong intercellular adhesion. No continuous epithelium is formed using this method.
  • the organoids obtained using the methods of the present invention display intercellular adhesion and a continuous epithelium. In some embodiments, the intercellular adhesion is strong.
  • the invention provides a breast organoid which has a cyst-like structure.
  • the cyst-like structure comprises a central lumen.
  • the central lumen of the cyst-like structure does not comprise any cells.
  • the central lumen of the cyst-like structure comprises breast epithelial stem cells, optionally wherein the central lumen is filled with breast epithelial stem cells.
  • the invention further provides a breast organoid that has a morphology that is similar to a cluster of grapes.
  • the invention further provides a breast epithelial stem cell, a population of breast epithelial stem cells or a breast organoid that has been cultured or can be cultured for more than 3 passages.
  • the invention further provides a culture medium comprising an ErbB3/4 ligand combined with an Lgr5 agonist and/or a FGFR2b ligand.
  • Organoids of the invention obtainable by expansion of stem cells, provide a population of cells which resemble their in vivo counterparts.
  • an organoid comprises at least one epithelial stem cell.
  • stem cell preferably refers to a cell which can divide and produce further epithelial stem cells or can generate differentiated progeny, e.g. progenitor cells, which are able to divide and further differentiate into one or more cell types.
  • progenitor cells e.g. progenitor cells
  • stem cell and “progenitor cell” may be used interchangeably.
  • a progenitor cell is able to dedifferentiate into a stem cell and so culturing a progenitor cell in the culture medium of the invention may result in the progenitor cell dedifferentiating into a stem cell.
  • the majority of cells are expanding cells (i.e. dividing cells) that retain an undifferentiated phenotype. Although some spontaneous differentiation may occur, the cell population is generally an expanding population.
  • the epithelial stem cells used in the invention are preferably multipotent, oligopotent or unipotent organ-specific adult stem cells.
  • the stem cells are not totipotent stem cells. In some embodiments, the stem cells are not pluripotent stem cells.
  • the organoids also have a distinctive structure that arises from these cellular properties, as described in detail below.
  • Image analysis may be used to assess characteristics of cells in culture such as cell morphology; cell structures; evidence for apoptosis or cell lysis; and organoid composition and structure.
  • Many types of imaging analysis are well known in the art, such as electron microscopy, confocal microscopy, stereomicroscopy, fluorescence microscopy. Histological analysis can reveal basic architecture and cell types.
  • an organoid of the invention preferably has a three dimensional structure, i.e. the organoid is preferably a three-dimensional organoid.
  • the organoid comprises only epithelial cells, i.e., non-epithelial cells are absent from the organoid.
  • the culture medium of the invention is specifically designed to expand epithelial stem cells, for example Lgr5+ epithelial stem cells. Therefore, even if other cell types are transiently present in the culture medium, e.g. in the tissue fragment that is the starting material of the invention, these cells are unlikely to survive and instead will be replaced by the longer term expansion of the stem cells which generate a pure population of epithelial cells.
  • the epithelial cells surround a lumen. In some embodiments, the epithelial cells surrounding the lumen are polarized, (meaning that proteins are differentially expressed on the apical or basolateral side of the epithelial cell). In some embodiments the organoids comprise stem cells which are able to actively divide and which are preferably able to differentiate to all major differentiated cell lineages present in the corresponding in vivo tissue.
  • the organoids of the invention have a section which is formed of multiple layers. In some embodiments the organoids of the invention have a monolayer section which appears to be formed of multiple layers referred to herein as regions of “pseudo-stratified” cells. In some embodiments the monolayer of pseudo-stratified cells folds so that it encloses lumina between the folds.
  • the organoids of the invention comprise single monolayers that are folded (or invaginated) to form two or more layers. It can sometimes be difficult to distinguish between folded (or invaginated) monolayers and regions of stratified cells.
  • an organoid comprises both regions of stratified cells and regions of folded monolayers.
  • the organoids of the invention have a section which is formed of multiple layers and a section comprising a single monolayer of cells.
  • the organoids of the invention comprise or consist of a single monolayer of cells.
  • the organoids of the invention comprise or consist of epithelial cells. In some embodiments, the organoids comprise or consist of a single layer of epithelial cells. In some embodiments non-epithelial cells are absent from the organoids.
  • an organoid of the invention has a structure essentially as presented in FIG. 2 (for example, in some embodiments, a normal organoid has a structure essentially as presented for the W894N or R1100N organoid lines in FIG. 2B or a cancer organoid has a structure essentially as presented for the W1007T, W1012T or W859T organoid lines in FIG. 2A ) or in FIG. 6 .
  • an organoid of the invention exhibits cell staining essentially as presented in FIG. 5 or FIG. 6 .
  • the organoids are mainly cystic structures with few budding structures. In some embodiments, the organoids generated according to the invention do not have budding structures.
  • the cystic structures comprise mainly monolayers but some regions of stratified cells may be present.
  • cystic it is meant that the organoid is approximately spherical.
  • budding it is meant that the organoid has multiple regions growing out of the basic structure.
  • breast organoids displayed cystic organoid structures, rather than the budding structures seen under previous culture conditions.
  • the invention also provides an organoid, preferably obtainable by the methods of the invention, which is a three-dimensional organoid comprising epithelial cells surrounding a central lumen.
  • said organoid comprises a single layer of cells. In other embodiments, said organoid comprises a multi-layered epithelium.
  • the organoid is obtained from normal breast tissue and has a cystic structure.
  • said normal organoid comprises a monolayer of basal and luminal cells surrounding a central lumen.
  • the organoid is obtained from cancerous breast tissue and has a cystic structure.
  • said organoid comprises: (i) a central lumen, multiple small lumina or no lumen and/or (ii) a monolayer of epithelial cells or a multi-layered epithelium.
  • the breast cancer organoid of the invention comprises a monolayer of epithelial cells surrounding a lumen.
  • the breast cancer organoid comprises a multi-layered epithelium and multiple small lumens.
  • the breast cancer organoid comprises a multi-layered epithelium and no lumen.
  • Cancer breast organoids optionally comprise solid balls of tumour cells.
  • Tumour status of cancer cells or cancer organoids may be confirmed using any suitable method. For example, karyotyping of cells or organoids can be performed to assess whether aneuploidy is present, which would suggest that the cells or organoids are tumour cells or tumour organoids.
  • Tumour cells or cancer organoids may also be identified by adding a p53 stabilizing agent to the culture medium.
  • Tumour cells and cancer organoids commonly contain p53 mutations that result in p53 stabilizing agents having no effect on the degradation of p53.
  • tumour cells unlike normal cells, are often able to escape senescence triggered by the addition of a p53 stabilizing agent (e.g. Nutlin-3) to a culture medium.
  • the breast cancer organoids stain positively for luminal cell markers, e.g. keratin-8 (Krt-8), and stain negatively for basal cell markers, e.g., keratin-14 (Krt-14) or keratin-5 (Krt-5).
  • the breast cancer organoid comprises luminal cells but does not comprise basal cells.
  • breast cancer organoids express KRT-8 at levels that are comparable to those seen in normal breast tissue or non-cancerous breast organoids.
  • breast cancer organoids express KRT-5 and/or KRT-14 at levels that are reduced compared to those seen in normal breast tissue or non-cancerous breast organoids.
  • Breast cancer organoids of the invention may express levels of CDH1 that are reduced compared to those seen in normal breast tissue or non-cancerous breast organoids.
  • Markers of epithelial to mesenchymal transition e.g., VIM, ZEB1
  • VIM epithelial to mesenchymal transition
  • Breast cancer organoids may display a reduced level of E-cadherin expression compared to normal breast tissue or non-cancerous breast organoids.
  • Breast cancer organoids of the invention may express other breast cancer cell markers such as Epithelial Membrane Antigen, Cancer antigen 15-3 (CA 15-3), cancer antigen 27.29 (CA 27.29), carcinoembryonic antigen (CEA), Urokinase plasminogen activator (uPA), plasminogen activator inhibitor (PAI-1), Podocalyxin, EZH2 and/or kallikrein-10.
  • breast cancer cell markers such as Epithelial Membrane Antigen, Cancer antigen 15-3 (CA 15-3), cancer antigen 27.29 (CA 27.29), carcinoembryonic antigen (CEA), Urokinase plasminogen activator (uPA), plasminogen activator inhibitor (PAI-1), Podocalyxin, EZH2 and/or kallikrein-10.
  • CA 15-3 Cancer antigen 15-3
  • CA 27.29 cancer antigen 27.29
  • CEA carcinoembryonic antigen
  • uPA Urokinas
  • an organoid or a population of epithelial stem cells which has been cultured in culture media of the invention for at least 2 months, for example at least 10 weeks, at least 12 weeks, at least 14 weeks, at least 16 weeks, at least 4 months, at least 5 months, at least 6 months, at least 9 months, at least one year.
  • the cells are human cells.
  • the cells have been passaged approximately every 1 to 4 weeks.
  • an organoid or a population of epithelial stem cells which has been that has been passaged or which is capable of being passaged for more than 3 passages, more than 4 passages, more than 5 passages, more than 6 passages, more than 7 passages, more than 8 passages, more than 9 passages, more than 10 passages, more than 11 passages, more than 12 passages, more than 13 passages or more than 14 passages.
  • a ErbB3/4 ligand allows the breast cells and organoids to be cultured long-term. It is not possible to culture and expand human breast epithelial stem cells or breast organoids for two months or more in the culture medium described in Pasic et al., WO2012/014076 or WO2012/168930, wherein the cells or organoids are passaged at least three times. Therefore, such breast organoids or populations of breast epithelial stem cells that had been cultured long-term did not exist before the present invention. Accordingly, there is provided an organoid or a population of stem cells that has been cultured/passaged or that is capable of being cultured/passaged as described herein.
  • a tissue fragment is a part of an adult tissue, preferably a human adult tissue, such as part of a human breast.
  • the tissue may be normal (healthy) tissue or it may be diseased or infected tissue.
  • an organoid as identified herein is therefore not a tissue fragment.
  • An organoid is preferably obtained using a cell from an adult tissue, preferably an epithelial stem cell from an adult tissue, optionally from an adult tissue fragment.
  • an organoid is obtained using an epithelial stem cell from an adult tissue or adult tissue fragment expressing Lgr5. Therefore, within the context of this invention, in some embodiments a tissue fragment comprises Lgr5+ stem cells.
  • an organoid is an organoid which is still being cultured using a method of the invention (preferably using a culture medium of the invention) and is therefore in contact with an extracellular matrix.
  • an organoid is embedded in a non-mesenchymal extracellular matrix.
  • in contact means a physical or mechanical or chemical contact, which means that for separating said organoid from said extracellular matrix a force needs to be used.
  • the extracellular matrix is a gelatinous protein mixture secreted by Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells, such as Matrigel (BD Biosciences) or Cultrex® Basement Membrane Extract (Trevigen, Inc.).
  • EHS Engelbreth-Holm-Swarm
  • the culture medium of the invention may optionally further comprise a population of cells of invention and/or one or more organoids of the invention.
  • the invention provides a composition comprising an organoid or cell of the invention, an extracellular matrix and a culture medium, preferably a culture medium of the invention.
  • An organoid comprises stem cells, such as epithelial stem cells, which retain their undifferentiated phenotype and self-renewal properties but also have differentiating progeny that grow into tissue-like structures.
  • breast organoids which more closely mimic the basic physiology of breast tissue, may be used in toxicity assays, or assays for drugs. They may also be useful for culturing pathogens which currently lack suitable tissue culture or animal models. Furthermore, such organoids may be useful in regenerative medicine, for example in post-radiation and/or post-surgery repair of the breast.
  • the invention provides the use of an organoid or expanded population of cells of the invention for use in medicine. Accordingly, the invention provides the use of an organoid or expanded population of cells of the invention for use in treating a disorder, condition or disease. The invention provides the use of an organoid or expanded population of cells of the invention for use in drug screening, (drug) target validation, (drug) target discovery, toxicology and toxicology screens, personalized medicine, regenerative medicine and/or as ex vivo cell/organ models, such as disease models.
  • Organoids or expanded population of cells of the invention can be used as ex vivo disease models.
  • Organoids of the invention can also be used for culturing of a pathogen and thus can be used as ex vivo infection models.
  • pathogens that may be cultured using an organoid of the invention include viruses, bacteria, prions or fungi that cause disease in its animal host.
  • an organoid of the invention can be used as a disease model that represents an infected state.
  • the organoids can be used in vaccine development and/or production.
  • Diseases that can be studied by the organoids of the invention thus include genetic diseases, metabolic diseases, pathogenic diseases, inflammatory diseases etc, for example including, but not limited to: breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g. adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • genetic diseases e.g. adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas
  • fibrocystic changes e.g. adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas
  • mastitis
  • iPS cells have been used as ex vivo cell/organ and/or disease models (for example, see Robinton et al. Nature 481, 295, 2012).
  • these methods suffer a number of challenges and disadvantages.
  • cell lines cannot be obtained from all patients (only certain biopsies result in successful cell lines) and therefore, cell lines cannot be used in personalised diagnostics and medicine.
  • iPS cells usually require some level of genetic manipulation to reprogramme the cells into specific cell fates. Alternatively, they are subject to culture conditions that affect karotypic integrity and so the time in culture must be kept to a minimum (this is also the case for human embryonic stem cells). This means that iPS cells cannot accurately represent the in vivo situation but instead are an attempt to mimic the behaviour of in vivo cells. Cell lines and iPS cells also suffer from genetic instability.
  • the organoids of the invention provide a genetically stable platform which faithfully represents the in vivo situation.
  • the organoids of the invention can also be expanded continuously, providing a good source of genetically stable cells.
  • an expanding population can be “split”, meaning that the organoid is split apart and all cells of the organoid are divided into new culture dishes or flasks. The divided cells are removed from the organoid and can then themselves be cultured and expanded to produce new organoids containing further expanded populations that can then be split again. Splits are also referred to herein as “passages”.
  • An organoid of the invention may be cultured for 1 or more passages, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more passages, for example, 20-30 passages, 30-35 passages, 32-40 passages or more.
  • an expanding cell population or organoid is split at every 3 days to every 10 weeks, every 1 to 10 weeks, every 1 to 4 weeks or every 10 days to 3 weeks.
  • the organoids are passaged approximately every 10 days to 3 weeks.
  • the organoids of the invention can provide an ongoing source of genetically stable cellular material.
  • the organoids of the invention can be used to gain mechanistic insight into a variety of diseases and therapeutics, to carry out in vitro drug screening, to evaluate potential therapeutics, to identify possible targets (e.g., proteins) for future novel (drug) therapy development and/or to explore gene repair coupled with cell-replacement therapy.
  • targets e.g., proteins
  • organoids or expanded populations of cells of the invention can be a tool for drug screening, target validation, target discovery, toxicology and toxicology screens and personalized medicine.
  • said expanded stem cell population or organoid of the invention is cultured in multiwell plates such as, for example, 96 well plates or 384 well plates.
  • Libraries of molecules are used to identify a molecule that affects said organoids.
  • Preferred libraries comprise antibody fragment libraries, peptide phage display libraries, peptide libraries (e.g. LOPAPTM, Sigma Aldrich), lipid libraries (BioMol), synthetic compound libraries (e.g. LO PACTM, Sigma Aldrich) or natural compound libraries (Specs, TimTec).
  • genetic libraries can be used that induce or repress the expression of one of more genes in the progeny of the stem cells.
  • These genetic libraries comprise cDNA libraries, antisense libraries, and siRNA or other non-coding RNA libraries.
  • the cells are preferably exposed to multiple concentrations of a test agent for a certain period of time. At the end of the exposure period, the cultures are evaluated.
  • the term “affecting” is used to cover any change in a cell, including, but not limited to, a reduction in, or loss of, proliferation, a morphological change, and cell death.
  • Said expanded stem cell population or organoid of the invention can also be used to identify drugs that specifically target breast cancer cells, e.g. the breast cancer cells or breast cancer organoids of the invention, but not normal breast cells, e.g. not said normal expanded stem cell population or normal organoid of the invention.
  • organoids are also useful for wider drug discovery purposes. It will be understood by the skilled person that the organoids of the invention would be widely applicable as drug screening tools for infectious, inflammatory and neoplastic pathologies of the human breast and other diseases of the breast. In some embodiments the organoids of the invention could be used for screening for cancer drugs.
  • the expanded cell populations for example the organoids of the invention or organoids obtained using media and methods of the invention can be used to test libraries of chemicals, antibodies, natural product (plant extracts), etc or specific known drugs for suitability for use as drugs, cosmetics and/or preventative medicines.
  • a cell biopsy from a patient of interest such as tumour cells from a cancer patient
  • organoids for identifying drugs in this way is that it is also possible to screen normal organoids (organoids derived from healthy tissue) to check which drugs and compounds have minimal effect on healthy tissue. This allows screening for drugs with minimal off-target activity or unwanted side-effects.
  • the organoids of the invention can be used for screening for drugs for breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • fibrocystic changes e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas
  • mastitis e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas
  • mastitis e.g., adenosis, metaplasia, dysplasi
  • the testing parameters depend on the disease of interest. For example, when screening for cancer drugs, cancer cell death is usually the ultimate aim. In other embodiments, metabolics or gene expression may be evaluated to study the effects of compounds and drugs of the screen on the cells or organoids of interest.
  • the invention provides a method for screening for a therapeutic or prophylactic drug, wherein the method comprises:
  • computer- or robot-assisted culturing and data collection methods are employed to increase the throughput of the screen.
  • expanded cell population for example, an organoid
  • expanded cell population is obtained from a patient biopsy.
  • the candidate molecule that causes a desired effect on the cultured expanded cell population is administered to said patient.
  • a method of treating a patient comprising:
  • the invention provides a method of treating breast cancer in a patient, comprising testing the patient's responsiveness to one or more drugs of interest using a drug screening method of the invention and then treating a patient with the drug if the patient is found to be responsive to the drug.
  • a breast cancer drug for use in treating a patient with breast cancer, wherein said treatment comprises testing the patient's responsiveness to the drug using a drug screening method of the invention and treating the patient with the drug if the patient is found to be responsive to the drug.
  • Examples of drugs for use in the invention which can be tested using a personalized medicine approach as outlined above include Abitrexate (Methotrexate), Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation), Ado-Trastuzumab Emtansine, Adriamycin PFS (Doxorubicin Hydrochloride), Adriamycin RDF (Doxorubicin Hydrochloride), Adrucil (Fluorouracil), Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Clafen (Cyclophosphamide), Cyclophosphamide, Cytoxan (Cyclophosphamide), Docetaxel, Doxorubicin Hydrochloride, Efudex (Fluorouracil), Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride,
  • Drugs for use in the invention which can be tested using a personalized medicine approach may be tested alone or in combination with one or more other drugs.
  • (a) Doxorubicin Hydrochloride, Adriamycin PFS (Doxorubicin Hydrochloride) or Adriamycin RDF (Doxorubicin Hydrochloride) may be tested in combination with (b) Cyclophosphamide, Clafen (Cyclophosphamide), Cytoxan (Cyclophosphamide) or Neosar (Cyclophosphamide), and, optionally, in further combination with (cxi) Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation) or Taxol (Paclitaxel), (cxii) Fluorouracil, Adrucil (Fluorouracil), Efudex (Fluorouracil) or Fluoroplex (Fluorouracil) or (cxiii) Docetaxel or Taxotere (Docetaxel).
  • Cyclophosphamide, Clafen (Cyclophosphamide), Cytoxan (Cyclophosphamide) or Neosar may be tested in combination with (b) Fluorouracil, Adrucil (Fluorouracil), Efudex (Fluorouracil) or Fluoroplex (Fluorouracil) and (cxi) Methotrexate, Abitrexate (Methotrexate), Folex (Methotrexate), Folex PFS (Methotrexate), Methotrexate LPF (Methotrexate), Mexate (Methotrexate) or Mexate-AQ (Methotrexate) or (cxii) Epirubicin Hydrochloride or Ellence (Epirubicin Hydrochloride).
  • the CMF and FEC drug combinations may be tested using a personalized medicine approach according to the invention.
  • the drug is used for treating, preventing or ameliorating symptoms of genetic diseases, metabolic diseases, pathogenic diseases, inflammatory diseases etc, for example including, but not limited to: breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • genetic diseases e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas
  • fibrocystic changes e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas
  • the organoids of the invention or cells grown using the culture media and methods of the invention can be used for target discovery.
  • Cells of the organoids originating from healthy or diseased tissue may be used for target identification.
  • the organoids of the invention may be used for discovery of drug targets for: breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • a library of compounds may be used to transduce the cells and inactivate specific genes.
  • cells are transduced with siRNA to inhibit the function of a (large) group of genes.
  • Any functional read out of the group of genes or specific cellular function can be used to determine if a target is relevant for the study.
  • a disease-specific read out can be determined using assays well known in the art. For example, cellular proliferation is assayed to test for genes involved in cancer.
  • a Topflash assay as described herein may be used to detect changes in Wnt activity caused by siRNA inhibition. Where growth reduction or cell death occurs, the corresponding siRNA related genes can be identified by methods known in the art. These genes are possible targets for inhibiting growth of these cells. Upon identification, the specificity of the identified target for the cellular process that was studied will need to be determined by methods well known in the art. Using these methods, new molecules can be identified as possible drug targets for therapy.
  • Patient-specific organoids obtained from diseased and/or normal tissue can be used for target validation of molecules identified in high throughput screens. The same goes for the validation of compounds that were identified as possible therapeutic drugs in high throughput screens.
  • the use of primary patient material expanded in the organoid culture system can be useful to test for false positives, etc. from high throughput drug discovery cell line studies.
  • the expanded stem cell population (for example, organoid of the invention) can be used for validation of compounds that have been identified as possible drugs or cosmetics in a high-throughput screen.
  • Said expanded stem cell population (for example, organoid of the invention) can further replace the use of cell lines in toxicity assays of potential novel drugs or of known drugs.
  • normal cells or organoids are used for these embodiments.
  • cancer cells or organoids may be used.
  • Toxicology screens work in a similar way to drug screens (as described above), but they test for the toxic effects of drugs and not therapeutic effects. Therefore, in some embodiments, the effects of the candidate compounds are toxic.
  • said expanded stem cell population (for example, organoid of the invention) can be used for culturing of a pathogen such as mouse mammary tumour virus (MMTV), human mammary tumor virus (HMTV), Epstein Barr virus (EBV), high risk human papillomavirus (HPV), Mycobacterium tuberculosis, Streptococcus uberis, Streptococcus dysgalactiae, Escherichia coli, Staphylococcus aureus , or Streptococcus agalactiae.
  • MMTV mouse mammary tumour virus
  • HMTV human mammary tumor virus
  • EBV Epstein Barr virus
  • HPV high risk human papillomavirus
  • Mycobacterium tuberculosis Streptococcus uberis
  • Streptococcus dysgalactiae Escherichia coli
  • Staphylococcus aureus or Streptococcus agalactiae
  • said expanded stem cell population (for example, organoid of the invention) can be used for monitoring the progression of a disease in the expanded stem cell population alongside monitoring the progression of a disease in a patient.
  • an expanded population of breast cancer stem cells could be obtained from a patient with breast cancer, the patient could then be treated with a certain drug or combination of drugs, and if the patient develops resistance to the drug or combination of drugs then a second expanded population of breast cancer stem cells could be obtained from the patient and compared to the first population of breast cancer stem cells, in order to identify causes of drug resistance and an appropriate therapy.
  • said expanded stem cell population (for example, organoid of the invention) can be used for identifying causes of drug resistance.
  • an expanded population of breast cancer stem cells could be obtained from a patient with breast cancer, the patient could then be treated with a certain drug or combination of drugs, and if the patient develops resistance to the drug or combination of drugs then a second expanded population of breast cancer stem cells could be obtained from the patient and compared to the first population of breast cancer stem cells, in order to identify causes of drug resistance.
  • the causes of drug resistance may be identified by identifying genetic changes in the resistant population of stem cells compared to the population of stem cells that responds to treatment with the drug.
  • Cultures comprising the expanded stem cell population are useful in regenerative medicine, for example in post-radiation and/or post-surgery repair of breast tissue, for example post-mastectomy, or in tissue repair of injured breast tissue.
  • breast epithelial stem cells with BRCA mutations may be obtained from a cancer patient, the BRCA mutations may then be repaired, resulting in the generation of autologous breast epithelial stem cells, which may then be transplanted back into the cancer patient.
  • the expanded epithelial stem cells are reprogrammed into related tissue fates.
  • gene therapy can additionally be used in a method directed at repairing damaged or diseased tissue.
  • Use can, for example, be made of an adenoviral or retroviral gene delivery vehicle to deliver genetic information, like DNA and/or RNA to stem cells.
  • a skilled person can replace or repair particular genes targeted in gene therapy.
  • a normal gene may be inserted into a nonspecific location within the genome to replace a non-functional gene.
  • an abnormal gene sequence can be replaced for a normal gene sequence through homologous recombination.
  • selective reverse mutation can return a gene to its normal function.
  • a further example is altering the regulation (the degree to which a gene is turned on or off) of a particular gene.
  • the stem cells are ex vivo treated by a gene therapy approach and are subsequently transferred to the mammal, preferably a human being in need of treatment.
  • organoids embedded in, or in contact with, an ECM can be transplanted into a mammal, preferably into a human.
  • organoids and ECM can be transplanted simultaneously into a mammal, preferably into a human.
  • an ECM can be used as a 3D scaffold for obtaining tissue-like structures comprising expanded populations of cells or organoids according to the invention. Such structures can then be transplanted into a patient by methods well known in the art.
  • An ECM scaffold can be made synthetically using ECM proteins, such as collagen and/or laminin, or alternatively an ECM scaffold can be obtained by “decellularising” an isolated organ or tissue fragment to leave behind a scaffold consisting of the ECM (for example see Macchiarini et al. The Lancet, Volume 372, Issue 9655, Pages 2023-2030, 2008).
  • an ECM scaffold can be obtained by decellularising an organ or tissue fragment, wherein optionally said organ or tissue fragment is from the breast.
  • the invention provides an organoid or population of cells of the invention for use in transplantation into a mammal, preferably into a human.
  • the invention enables a small biopsy to be taken from an adult donor and expanded without any apparent limit or genetic harm and so the technology provided herein may serve to generate transplantable epithelium for regenerative purposes.
  • the patient is preferably a human, but may alternatively be a non-human mammal, such as a cat, dog, horse, cow, pig, sheep, rabbit or mouse.
  • a patient can be treated with allogeneic or autologous stem cells or organoids.
  • autologous cells are cells which originated from the same organism into which they are being re-introduced for cellular therapy, for example in order to permit tissue regeneration. However, the cells have not necessarily been isolated from the same tissue as the tissue they are being introduced into. An autologous cell does not require matching to the patient in order to overcome the problems of rejection.
  • Allogeneic cells are cells which originated from an individual which is different from the individual into which the cells are being introduced for cellular therapy, for example in order to permit tissue regeneration, although of the same species. Some degree of patient matching may still be required to prevent the problems of rejection.
  • the cells or organoids of the invention are introduced into the body of the patient by injection or implantation. Generally, the cells will be directly injected into the tissue in which they are intended to act.
  • a syringe containing cells of the invention and a pharmaceutically acceptable carrier is included within the scope of the invention.
  • a catheter attached to a syringe containing cells of the invention and a pharmaceutically acceptable carrier is included within the scope of the invention.
  • the skilled person will be able to select an appropriate method and route of administration depending on the material that is being transplanted (i.e., population of cells, single cells in cell suspension, organoids or fragments of organoids).
  • cells of the invention can be used in the regeneration of tissue.
  • cells may be injected or implanted directly into the damaged tissue, where they may multiply and eventually differentiate into the required cell type.
  • the organoid can be injected or implanted directly into the damaged tissue.
  • Breast tissues that are susceptible to treatment include all damaged tissues, particularly including those which may have been damaged by disease (e.g. cancer), injury, trauma, an autoimmune reaction, or by a viral or bacterial infection.
  • the cells or organoids of the invention are used to regenerate the breast.
  • the cells or organoids of the invention are injected into a patient using a Hamilton syringe.
  • the invention provides a syringe comprising the cells or organoids of the invention.
  • the cells or organoids of the invention either in solution, in microspheres or in microparticles of a variety of compositions, will be administered into the artery irrigating the tissue or the part of the damaged organ in need of regeneration.
  • administration will be performed using a catheter.
  • the catheter may be one of the large variety of balloon catheters used for angioplasty and/or cell delivery or a catheter designed for the specific purpose of delivering the cells to a particular local of the body.
  • the cells or organoids may be encapsulated into microspheres made of a number of different biodegradable compounds, and with a diameter of about 15 ⁇ m.
  • the cells or organoids of the invention may be implanted into the damaged tissue adhered to a biocompatible implant.
  • the cells may be adhered to the biocompatible implant in vitro, prior to implantation into the patient.
  • adherents may include fibrin, one or more members of the integrin family, one or more members of the cadherin family, one or more members of the selectin family, one or more cell adhesion molecules (CAMs), one or more of the immunoglobulin family and one or more artificial adherents.
  • CAMs cell adhesion molecules
  • the cells or organoids of the invention may be embedded in a matrix, prior to implantation of the matrix into the patient.
  • the matrix will be implanted into the damaged tissue of the patient.
  • matrices include collagen based matrices, fibrin based matrices, laminin based matrices, fibronectin based matrices and artificial matrices. This list is provided by way of illustration only, and is not intended to be limiting.
  • the cells or organoids of the invention may be implanted or injected into the patient together with a matrix forming component.
  • a matrix forming component may allow the cells to form a matrix following injection or implantation, ensuring that the cells or organoids remain at the appropriate location within the patient.
  • matrix forming components include fibrin glue liquid alkyl, cyanoacrylate monomers, plasticizers, polysaccharides such as dextran, ethylene oxide-containing oligomers, block co-polymers such as poloxamer and Pluronics, non-ionic surfactants such as Tween and Triton ‘8’, and artificial matrix forming components. This list is provided by way of illustration only, and is not intended to be limiting. It will be clear to a person skilled in the art, that any combination of one or more matrix forming components may be used.
  • the cells or organoids of the invention may be contained within a microsphere.
  • the cells may be encapsulated within the centre of the microsphere.
  • the cells may be embedded into the matrix material of the microsphere.
  • the matrix material may include any suitable biodegradable polymer, including but not limited to alginates, Poly ethylene glycol (PLGA), and polyurethanes. This list is provided by way of example only, and is not intended to be limiting.
  • the cells or organoids of the invention may be adhered to a medical device intended for implantation.
  • medical devices include stitches and artificial skin.
  • the cells may be adhered to the medical device by a variety of methods.
  • the cells or organoids may be adhered to the medical device using fibrin, one or more members of the integrin family, one or more members of the cadherin family, one or more members of the selectin family, one or more cell adhesion molecules (CAMs), one or more of the immunoglobulin family and one or more artificial adherents.
  • CAMs cell adhesion molecules
  • the cells or organoids of the invention may be used to define patient groups for drug development.
  • the cells or organoids of the invention may also be used for pre-screening patients in phase I or phase II trials.
  • the cells or organoids of the invention may be used to identify patients on which to perform a clinical trial.
  • the cells or organoids of the invention may also be used in diagnostics after a drug is approved.
  • the cells or organoids of the invention may be used to identify patients who are most likely to benefit from a particular drug, to identify patients likely to be at increased risk for serious side effects as a result of treatment with a particular drug or to monitor the response to treatment with a particular drug for the purpose of adjusting treatment to achieve improved safety or effectiveness.
  • the cells or organoids of the invention may in some embodiments be used for veterinary therapies.
  • the cells or organoids may be used in the treatment of mastitis in cows.
  • the invention provides a pharmaceutical composition comprising the cells or organoids of the invention and further comprising a pharmaceutically acceptable carrier or excipient.
  • the invention provides a composition comprising a culture medium according to the invention and stem cells.
  • the invention also provides a composition comprising a culture medium according to the invention and organoids.
  • the invention provides a composition comprising a culture medium according to the invention and an extracellular matrix.
  • the invention also provides a composition comprising a culture medium of the invention, an extracellular matrix and stem cells of the invention.
  • the invention also provides a composition comprising a culture medium of the invention, an extracellular matrix and one or more organoids of the invention.
  • the invention also provides a culture medium supplement that can be used to produce a culture medium as disclosed herein.
  • a ‘culture medium supplement’ is a mixture of ingredients that cannot itself support stem cells, but which enables or improves stem cell culture when combined with other cell culture ingredients. The supplement can therefore be used to produce a functional cell culture medium of the invention by combining it with other cell culture ingredients to produce an appropriate medium formulation.
  • the use of culture medium supplements is well known in the art.
  • the invention provides a culture medium supplement that comprises an ErbB3/4 ligand according to the invention.
  • the supplement may contain any ErbB3/4 ligand (or combination of ErbB3/4 ligand) disclosed herein.
  • the supplement may also contain one or more additional cell culture ingredients as disclosed herein, e.g. one or more cell culture ingredients selected from the group consisting of amino acids, vitamins, inorganic salts, carbon energy sources and buffers.
  • a culture medium or culture medium supplement may be a concentrated liquid culture medium or supplement (e.g. a 2 ⁇ to 250 ⁇ concentrated liquid culture medium or supplement) or may be a dry culture medium or supplement. Both liquid and dry culture media or supplements are well known in the art.
  • a culture medium or supplement may be lyophilised.
  • a culture medium or supplement of the invention will typically be sterilized prior to use to prevent contamination, e.g. by ultraviolet light, heating, irradiation or filtration.
  • a culture medium or culture medium supplement may be frozen (e.g. at ⁇ 20° C. or ⁇ 80° C.) for storage or transport.
  • the culture medium may be stored as a liquid (e.g. at approximately 4° C.).
  • the culture medium may be split and stored as two components: a frozen component (e.g. at between approximately ⁇ 20° C. and approximately ⁇ 80° C.) and a liquid component (e.g. at approximately 4° C.).
  • temperature-sensitive or time-sensitive degradable material is preferably included in the frozen component, whereas less sensitive material (for example DMEM or FCS) can be stored in the liquid form and thus included in the liquid component for storage and shipping.
  • the invention also provides a hermetically-sealed vessel containing a culture medium or culture medium supplement of the invention.
  • Hermetically-sealed vessels may be preferred for transport or storage of the culture media or culture media supplements disclosed herein, to prevent contamination.
  • the vessel may be any suitable vessel, such as a flask, a plate, a bottle, a jar, a vial or a bag.
  • the invention also provides a kit comprising a culture medium, culture medium supplement and/or a composition of the invention.
  • the kit further comprises at least one other additional component, for example selected from the list comprising: an ECM (for example, MatrigelTM or Cultrex® Basement Membrane Extract), a population of cells and an organoid.
  • ECM for example, MatrigelTM or Cultrex® Basement Membrane Extract
  • composition “comprising” encompasses “including” as well as “consisting” e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X+Y.
  • a process comprising a step of mixing two or more components does not require any specific order of mixing.
  • components can be mixed in any order. Where there are three components then two components can be combined with each other, and then the combination may be combined with the third component, etc.
  • FIG. 1A 20 breast tumors were sampled, three of which were triple negative (15%, FIG. 1A and FIG. 13 ).
  • the first 12 samples were used to optimize cell isolation and organoid culture conditions. Samples 13-20 were grown in six different conditions.
  • tissue samples were photographed and cut into 1-3 mm 3 pieces. Two random pieces were snap frozen and stored at ⁇ 80° C. for DNA isolation, two random pieces were fixed in formalin for histopathological analysis and immunohistochemistry, and the remainder was processed for the isolation of viable cells. The remaining tissue was minced, washed with 10 ml AdDF+++ and digested in 10 ml base-F 7/10 N medium containing 1-2 mg/ml collagenase (Sigma-C9407) at 120 rpm and 37° C. for 1-2h. The digested tissue suspension was sequentially sheared using 10 ml and 5 ml plastic and flamed glass Pasteur pipettes.
  • the suspension was strained over a 100 ⁇ m filter with retained tissue pieces entering a subsequent shearing step with ⁇ 10 ml AdDF+++.
  • 2% FCS were added to the strained suspension before centrifugation at 400 g.
  • the pellet was resuspended in 10 ml AdDF+++ and centrifuged again at 400.
  • erythrocytes were lysed in 2 ml red blood cell lysis buffer (Roche-11814389001) for 5 min at room temperature before the addition of 10 ml AdDF+++ and centrifugation at 400 g.
  • the pellet was resuspended in 10 mg/ml cold Cultrex growth factor reduced BME type 2 (Trevigen-3533-010-02) and 40 ⁇ l drops of BME-cell suspension were allowed to solidify on prewarmed 24-well suspension culture plates (Greiner-M9312) at 37° C. for 10-20 min.
  • 400 ⁇ l of organoid media base-F 7/10 N, base-F 7/10 N, or base-F 7/10 N
  • base-F 7/10 N base-F 7/10 N
  • cystic organoids were resuspended in 2 ml cold AdDF+++ and mechanically sheared through flamed glass Pasteur pipettes. Dense organoids were dissociated by resuspension in 2 ml TrypLE Express (Invitrogen-12605036), incubation for 1-5 min at room temperature, and mechanical shearing through flamed glass Pasteur pipettes. Following the addition of 10 ml AdDF+++ and centrifugation at 300 g or 400 g respectively, organoid fragments were resuspended in cold BME and reseeded as above at ratios (1:1-1:6) allowing the formation of new organoids. Single cell suspensions were initially seeded at high density and reseeded at a lower density after ⁇ 1 week.
  • Mammary tumor organoid lines W854T, W855T, and W859T readily expand in base-F 7/10 N medium and were therefore characterized in more detail.
  • Line W854T furthermore readily grew in medium containing 5 ⁇ M Nutlin-3 indicating the presence of mutant p53.
  • RNA and DNA were isolated and sent to BI for gene expression profiling and exome sequencing.
  • normal organoid line W855N consists of mutually exclusive basal and luminal cells
  • tumor organoids solely express luminal cell marker Keratin-8 ( FIGS. 6 and 7 ).
  • E-cadherin expression is present but decreased in tumor compared to normal organoid lines indicating their epithelial origin ( FIGS. 6 and 7 ).
  • markers of epithelial to mesenchymal transition (EMT) are mildly upregulated in tumor organoid lines W855T and W859T corresponding to their incoherent phenotype ( FIGS. 6 and 7 ).
  • lines W854T, W855T, and W859T were plated in duplicate into a 96-well format and exposed to medium containing the EGFR inhibitor Iressa (1 nM-30 ⁇ M at 1:3 dilution steps, S1025 Selleck Chemicals) or the p53 stabilizer Nutlin-3 (1 nM-30 ⁇ M at 1:3 dilution steps, 10004372 Cayman Chemicals) for 7 days. Organoids were photographed every 3-4 days ( FIG. 8 ) and cell viability measured at day 7 using the CellTiter-Glo 2.0 assay (G9241, Promega) ( FIG. 9 ).
  • Mammary tumor organoids are suitable for cell viability based low throughput drug screens in a 96-well format. We are currently testing the assay in a 384-well plate format which has been successfully established for colon cancer organoids.
  • Normal mammary organoids in Pasic's culture medium slowed expanding at passage 2 and stopped at passage 3; organoids from the same patient grown in base-F 7/10 E continued expanding up to passage 4 (see FIG. 10 ).
  • Normal mammary organoids from different patients expand at least up to passage 6 in medium base F 7/10 EN.

Abstract

The invention relates to improved culture methods for expanding epithelial stem cells and obtaining organoids, to culture media involved in said methods, and to uses of said organoids.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. application Ser. No. 15/529,150, filed May 24, 2017, which is a national phase entry pursuant to 35 U.S.C. § 371 of International Application No. PCT/EP2015/077988, filed Nov. 27, 2015, which claims the benefit of priority of Great Britain Application No. 1421094.2, filed Nov. 27, 2014, each of which is incorporated by reference herein in its entirety for any purpose.
  • SEQUENCE LISTING
  • The present application is filed with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled “2021-02-22_01169-0008-01US_Sequence_Listing_ST25.txt” created on Feb. 22, 2021, which is 126,976 bytes in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
  • All documents cited herein are incorporated by reference in their entirety.
  • TECHNICAL FIELD
  • The invention is in the field of breast epithelial stem cell culture media and methods, in particular culture media and methods for expanding populations of breast epithelial stem cells, e.g. human breast epithelial stem cells.
  • BACKGROUND
  • There is great interest in culture media and methods for expanding populations of stem cells. Populations of stem cells have many uses. For example, stem cells and their differentiated progeny can be used in cellular assays, drug screening, and toxicity assays. Stem cells also show promise for cell-based therapies, such as in regenerative medicine for the treatment of damaged tissue. Furthermore, efficient cell culture media are important for providing and maintaining populations of cells for research purposes.
  • Methods for the long-term culture of epithelial stem cells or tissue fragments derived from several tissues (e.g. pancreas, colon, intestinal crypts, stomach, liver and prostate) have been described (see WO 2010/090513, WO2012/014076 and WO2012/168930). However, these methods have been found to be unable to support the long-term culture of epithelial stem cells or tissue fragments derived from breast tissue.
  • A culture system for culturing epithelial stem cells derived from normal breast tissue is described in Pasic et al. (2011) Genes & Development 25:1641-1653. However, this system is unable to support long-term culture of breast organoids. Cultures using this system have been observed to stop expanding after 2-3 passages. Accordingly, it is not possible to obtain large numbers of stem cells using this technique.
  • A culture system for culturing circulating tumour cells from breast cancer patients is described in Yu et al. (2014) Science 345(6193):216-220. However, the authors of this paper provided no guidance with respect to culturing breast epithelial stem cells obtained from normal breast tissue, primary tumour or metastases. Furthermore, circulating breast tumour cells cultured using the Yu et al. system form aggregates of cells.
  • Therefore, there is a need for culture media and methods for culturing breast epithelial stem cells obtained from normal breast tissue, primary tumours or metastases that permit long-term culture. There is also a need for culture media and methods for culturing circulating breast tumour cells that permit the formation of breast organoids that closely resemble breast tumours.
  • SUMMARY OF THE INVENTION
  • The invention provides a method for expanding breast epithelial stem cells comprising:
      • providing a population of breast stem cells;
      • providing a culture medium comprising an ErbB3/4 ligand, one or more FGFR2b ligands and a Wnt agonist;
      • contacting the stem cells with the culture medium; and
      • culturing the cells under appropriate conditions.
  • The invention further provides a culture medium comprising an ErbB3/4 ligand and one or more FGFR2b ligands and a Wnt agonist.
  • The invention further provides a culture medium comprising an ErbB3/4 ligand suitable for expanding a population of breast epithelial stem cells for at least 4 passages.
  • The invention further provides a breast organoid which comprises a population of breast epithelial stem cells. In some embodiments, said breast organoid is obtainable or obtained by a method of the invention.
  • The invention further provides a breast organoid of the invention in a culture medium of the invention.
  • The invention further provides the use of a breast organoid of the invention, or a cell derived from said organoid, in a drug discovery screen; toxicity assay; research of tissue embryology, cell lineages, or differentiation pathways; gene expression studies including recombinant gene expression; research of mechanisms involved in tissue injury or repair; research of inflammatory and infectious diseases; studies of pathogenetic mechanisms; or studies of mechanisms of cell transformation or aetiology of cancer.
  • The invention further provides an organoid of the invention, or a cell derived from said organoid, for use in medicine.
  • The invention further provides a culture medium comprising a p53 stabilising agent.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1A-1B. (A) Breast tumour tissues obtained from patients displayed according to subtype (estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor-2 (HER2) status); (B) established or promising organoid lines displayed according to subtype (ER, PR and HER2 status).
  • FIG. 2. Phase contrast images of established and promising mammary (A) tumour and (B) normal organoid lines. Scale bars equal 150 μm.
  • FIG. 3A-3B. Example of media optimization. Normal and tumour mammary cells of patient W855 were suspended in Cultrex® Basement Membrane Extract (Trevigen, Inc.) and supplemented with the indicated media. Phase contrast images are displayed for: (A) Passage 0 day 4 and (B) Passage 0 day 14. The most beneficial culture conditions are marked by bold grey boxes. Image widths: 1.5 mm top panels, 300 μm bottom panels.
  • FIG. 4. Aneuploid karyotypes of established mammary tumour organoid lines.
  • FIG. 5. Histological and immunohistochemical comparison of original mammary gland tissue (W854 tumour (W854T), W855 normal and tumour (W855N and W855T), W859 tumour (W859T)) with the respectively derived organoid lines. Scale bars equal 100 μm (overview) and 20 μm (detailed view). Key: HE=haematoxylin and eosin stain; PR=progesterone receptor (clone PgR 636, M356929 Dako), ER=estrogen receptor alpha (clone SP1, ab6660 Abcam), HER2=human epidermal growth factor receptor 2 (c-erbB2) (clone SP3, #RM-9103, Thermoscientific).
  • FIG. 6. Immunofluorescent analysis of whole mounted normal (W855) and tumour (W854, W855 and W859) mammary organoid lines. Nuclear counterstain is DRAQ5 (DR50050 Biostatus limited). Scale bar equals 100 μm. Key: Krt8=keratin-8 (clone Ks8.7, sc101459 Santa Cruz), Krt14=keratin-14 (clone AF64, PRB-155P Covance), ER=estrogen receptor alpha (clone C1355, Fisher 50-172-167), E-cadherin (clone 36/E-cadherin, 61082 BD Biosciences), HER2=human epidermal growth factor receptor 2 (c-erbB2) (clone SP3, #RM-9103, Thermoscientific), PR=progesterone receptor (clone PgR 636, M356929 Dako).
  • FIG. 7. Quantitative PCR on organoids for basal (KRT5/14), luminal (KRT8), epithelial (CDH1) and mesenchymal (VIM, ZEB1) markers normalized to normal mammary organoid line W855N.
  • FIG. 8. Phase-contrast images of mammary tumour organoid lines W854T, W855T and W859T during a pilot drug screen with increasing doses of the EGFR inhibitor Iressa or the p53 stabilizer Nutlin-3. Visibly affected organoids are marked by bold black boxes. Image widths are 500 μm.
  • FIG. 9. Viability of mammary tumour organoid lines W854T, W855T and W859T treated with increasing does of the EGFR inhibitor Iressa or the p53 stabilizer Nutlin-3 for 7 days, as measured by a luminescent ATP assay (CellTiter-Glo 2.0, G9241 Promega).
  • FIG. 10. Normal human mammary organoids in the culture medium described in Pasic et al. compared with organoids from the same patient grown in base-F7/10E medium.
  • FIG. 11. Mouse mammary tumour organoids. Phase-contrast images of organoids established from KB2P mammary tumors lacking Brca2 and p53, Passage 4, Image widths are 7 mm, 1.5 mm, and 600 μm.
  • FIG. 12. Human metastatic mammary tumor organoids. Phase-contrast images of organoids established from a human breast cancer metastasis to the skin, Passage 5, Image widths are 7 mm, 1.5 mm, and 600 μm.
  • FIG. 13. Overview of mammary organoid cultures.
  • FIG. 14. Organoid culture media.
  • DETAILED DESCRIPTION
  • Methods for culturing epithelial stem cells from a variety of tissues have previously been described in WO2010/090513, WO2012/014076 and WO2012/168930. The present inventors have surprisingly found that adding an ErbB3/4 ligand to the culture medium allows breast epithelial stem cells to be cultured for an increased number of passages compared to when the ErbB3/4 ligand is absent from the medium.
  • The ability to keep the cells and resulting organoids alive for longer and increase the passage number advantageously allows more cells to be obtained from a collection of starting cells than was possible using previous methods. This enables a large number of cells to be available for various applications, for example, drug screening, in which a large amount of material is required to test various different drugs. The ability to generate the cells from a single starting source is advantageous for such applications where it is necessary to compare results between experiments. Furthermore, the enhanced ability to obtain more cells from a small collection of starting cells (e.g. a collection of approximately 100-300 cells) is advantageous for applications where little starting material is available, such as biopsies of primary or metastatic cancer or circulating tumour cells. Similarly, it means that many cells are available for use in transplants, e.g. in breast reconstruction or breast enhancement, and that multiple patients may be transplanted with cells obtained from a useful donor.
  • Culturing the cells in a medium of the invention allows the cells to multiply whilst retaining their stem or progenitor cell phenotype, which is referred to herein as expansion. Organoids are formed comprising these stem or progenitor cells. Use of the medium of the invention is therefore advantageous for providing increased numbers of these useful stem or progenitor cells and for obtaining organoids containing these cells.
  • Accordingly, there is provided a method for culturing breast epithelial stem cells, wherein said method comprises culturing one or more breast epithelial stem cells in contact with an extracellular matrix in the presence of a culture medium, the culture medium comprising a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands.
  • The culture medium used in the method of the invention is preferably a culture medium of the invention as described herein.
  • Accordingly, there is also provided a culture medium comprising a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands (e.g. 1, 2, 3, 4, or more than 4), and one or more ligands of FGFR2b (e.g. 1, 2, 3, 4 or more than 4), preferably wherein the one or more ligands of FGFR2b is FGF7 and/or FGF10, for example, for use in the above method.
  • Accordingly, there is also provided a culture medium comprising a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands (e.g. 1, 2, 3, 4, or more than 4), and one or more Wnt agonists (e.g. 1, 2, 3, 4 or more than 4), for example, for use in the above method.
  • In some embodiments, the one or more Wnt agonists is a Lgr5 agonist, Lgr4 agonist, Lgr6 agonist or Wnt3a. In some embodiments, the Lgr5 agonist is an Rspondin, e.g. any one of Rspondin 1-4.
  • In preferred embodiments, the culture medium of the invention comprises a basal medium for animal or human cells to which is added one or more ErbB3/4 ligands (e.g. 1, 2, 3, 4, or more than 4), one or more ligands of FGFR2b (e.g. 1, 2, 3, 4 or more than 4), preferably wherein the one or more ligands of FGFR2b is FGF7 and/or FGF10, and one or more Wnt agonists (e.g. 1, 2, 3, 4 or more than 4), preferably wherein the one or more Wnt agonists is a Lgr5 agonist, preferably wherein the Lgr5 agonist is an Rspondin, e.g. any one of Rspondin 1-4.
  • In some embodiments, the culture medium of the invention or the culture medium used in the method of the invention comprises one or more additional components selected from: a ligand of FGFR2b (e.g. FGF7 and/or FGF10), a Lgr5 agonist (e.g. Rspondin, e.g. any one of Rspondin 1-4), a BMP inhibitor (for example, Noggin), B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor (for example, an ALK 4, 5, 7 inhibitor, e.g. A83-01) and a p38 MAP kinase inhibitor (for example, SB 202190). In some embodiments, the culture medium further comprises at least two, at least three, at least four, at least five, at least six, at least seven, (e.g. 2, 3, 4, 5, 6, 7) of these additional components. In some embodiments, the culture medium further comprises all of these additional components.
  • In some embodiments, the culture medium further comprises a further receptor tyrosine kinase ligand, e.g. EGF.
  • In some embodiments, the culture medium further comprises a Wnt agonist, e.g. Wnt3a.
  • In some embodiments, the culture medium further comprises one or more of amphiregulin, TGF-alpha and PDGF-CC. For example, in some embodiments the culture medium further comprises PDGF-CC. In some embodiments, the culture medium further comprises amphiregulin, TGF-alpha and PDGF-CC. The invention therefore provides the use of an ErbB3/4 ligand for culturing breast epithelial stem cells.
  • The invention also provides a method for culturing breast epithelial stem cells which uses an expansion medium as described in WO2012/168930, WO2010/090513 or WO2012/014076 to which at least one (e.g. 1, 2, 3, 4, or more than 4) ErbB3/4 ligand and at least one (e.g. 1, 2, 3, 4, or more than 4) FGFR2b ligands are added.
  • The present inventors have also surprisingly found that the addition of a p53 stabilizing agent to a culture medium can ensure that the cell population is predominantly tumour cells. Without wishing to be bound by any theory, it is believed that p53 stabilizing agents increase the cellular concentration of p53 (e.g. by blocking the interaction between p53 and Mdm2), stimulate p53-dependent expression and induce cellular senescence. p53 mutations commonly occur in tumour cells, which may result in the mutant p53 protein being unable to be stabilised by a p53 stabilizing agent. Thus, a subset of tumour cells with the mutant p53 protein would escape the senescence induced by a p53 stabilizing agent and outgrow normal cells in a mixed population.
  • The invention therefore further provides the use of a p53 stabilizing agent for culturing tumour cells.
  • The invention also provides a method for culturing tumour stem cells which uses an expansion medium as described herein or in WO2012/168930, WO2010/090513 or WO2012/014076 to which at least one (e.g. 1, 2, 3, 4, or more than 4) p53 stabilizing agent is added. The invention also provides a culture medium comprising a p53 stabilizing agent. Preferably, the culture medium is a culture medium as described herein or in W2012/168930, WO2010/090513 or WO2012/014076. Preferably, the culture medium is suitable for culturing epithelial stem cells.
  • A preferred p53 stabilizing agent is a member of the Nutlin family, e.g. Nutlin-1, Nutlin-2 or Nutlin-3. For example, in some embodiments, the p53 stabilizing agent is Nutlin-3. Other p53 stabilising agents are known in the art (e.g. CP-31398) and the skilled person will be able to use any of these accordingly.
  • ErbB3/4 Ligand
  • The ErbB receptor tyrosine kinase family consists of four cell surface receptors: i) ErbB1/EGFR/HER1, ii) ErbB2/HER2, iii) ErbB3/HER3, and iv) ErbB4/HER4. An ErbB3/4 ligand is herein defined as a ligand that is capable of binding to ErbB3 and/or ErbB4. Accordingly, EGF is not an ErbB3/4 ligand because it does not bind to ErbB3/4. In some embodiments, the Erb3/4 ligand binds to ErbB3 and does not bind to ErbB4. In some embodiments, the Erb3/4 ligand binds to Erb4 and does not bind to Erb3. In some embodiments, binding of the ErbB3/4 ligand to ErbB3 or ErbB4 induces the heterodimerization of said ErbB3 or said ErbB4 with ErbB2. In some embodiments, the induction of heterodimerization of ErbB3 or ErbB4 with ErbB2 stimulates intrinsic kinase activity, which leads to tyrosine phosphorylation. In the context of a culture medium of the invention, an ErbB3/4 ligand is known as “N”.
  • Various ErbB3/4 ligands are known in the art. In preferred embodiments, the one or more ErbB3/4 ligands of the culture medium are members of the neuregulin/heregulin family. The neuregulin/heregulin family is referred to herein as the neuregulin family. The neuregulin family is a family of structurally related polypeptide growth factors that are gene products of alternatively spliced genes NRG1, NRG2, NRG3 and NRG4. In more preferred embodiments, the one or more ErbB3/4 ligands of the culture medium are polypeptides that are gene products of one or more of NRG1, NRG2, NRG3 and NRG4 (i.e., a neuregulin polypeptide). In some embodiments, the ErbB3/4 ligand is a polypeptide that is a gene product of NRG. The polypeptide that is a gene product of NRG1, NRG2, NRG3 and NRG4 may be any of the isoforms that result from the alternative splicing of NRG1, NRG2, NRG3 or NRG4 mRNA. Thus, for example, the polypeptide that is a gene product of NRG1 may be any of the following isoforms: Type I NRG1 (also known as heregulin, NEU differentiation factor (NDF) or acetylcholine receptor inducing activity (ARIA)), Type II NRG1 (also known as Glial Growth Factor-2 (GGF2)), Type III NRG1 (also known as Sensory and motor neuron-derived factor (SMDF)), Type IV NRG1, Type V NRG1 or Type VI NRG1. In some embodiments, the neuregulin polypeptide is pro-neuregulin-1, membrane-bound isoform isoform HRG-beta1 (NP_039250.2), pro-neuregulin-1, membrane-bound isoform isoform HRG-beta1b (NP_001153471.1), pro-neuregulin-1, membrane-bound isoform isoform HRG-beta1c (NP_001153467.1), pro-neuregulin-1 or membrane-bound isoform isoform HRG-beta1d (NP_001153473.1).
  • In some embodiments the ErbB3/4 ligand is of human origin. Accordingly, in some embodiments the ErbB3/4 ligand is a human gene product of one or more of NRG1, NRG2, NRG3 and NRG4 (i.e. a human neuregulin polypeptide).
  • In some embodiments, the NRG1 gene has the sequence of Gene ID: 3084 or NG_012005.1. In some embodiments, the NRG2 gene has the sequence of Gene ID: 9542. In some embodiments, the NRG3 gene has the sequence of Gene ID: 10718 or NG_013373.1. In some embodiments, the NRG4 gene has the sequence shown of Gene ID: 145957.
  • In some embodiments a neuregulin polypeptide is a gene product of NRG1 and has the sequence shown in NP_001153467.1 (SEQ ID NO: 1), NP_001153468.1 (SEQ ID NO: 2), NP_001153471.1 (SEQ ID NO: 3), NP_001153473.1 (SEQ ID NO: 4), NP_001153474.1 (SEQ ID NO: 5), NP_001153476.1 (SEQ ID NO: 6), NP_001153477.1 (SEQ ID NO: 7), NP_001153479.1 (SEQ ID NO: 8), NP_001153480.1 (SEQ ID NO: 9), NP_004486.2 (SEQ ID NO: 10), NP_039250.2 (SEQ ID NO: 11), NP_039251.2 (SEQ ID NO: 12), NP_039252.2 (SEQ ID NO: 13), NP_039253.1 (SEQ ID NO: 14), NP_039254.1 (SEQ ID NO: 15), NP_039256.2 (SEQ ID NO: 16) or NP_039258.1 (SEQ ID NO: 17). In some embodiments a neuregulin polypeptide is a gene product of NRG2 and has the sequence shown in NP_001171864.1 (SEQ ID NO: 18), NP_004874.1 (SEQ ID NO: 19), NP_053584.1 (SEQ ID NO: 20), NP_053585.1 (SEQ ID NO: 21), or NP_053586.1 (SEQ ID NO: 22). In some embodiments a neuregulin polypeptide is a gene product of NRG3 and has the sequence shown in NP_001010848.2 (SEQ ID NO: 23), NP_001159444.1 (SEQ ID NO: 24), or NP_001159445.1 (SEQ ID NO: 25). In some embodiments a neuregulin polypeptide is a gene product of NRG4 and has the sequence shown in NP_612640.1 (SEQ ID NO: 26).
  • In some embodiments, the at least one ErbB3/4 ligand is a biologically active variant of one or more naturally occurring ErB3/4 ligands, for example, of one or more members of the neuregulin family. Neuregulin variants, which may be naturally occurring (e.g. allelic variants that occur at the NRG1 locus) or recombinantly produced, have amino acid sequences that are the same as, similar to or substantially similar to a neuregulin polypeptide.
  • In some embodiments, a neuregulin variant has a sequence that is at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a neuregulin polypeptide, for example the neuregulin variant has a sequence that is at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to any one of SEQ ID NOs: 1-26.
  • In some embodiments, the at least one ErbB3/4 ligand is at least one biologically active fragment of at least one naturally occurring ErbB3/4 ligand, for example a biologically active fragment of of one or more neuregulin polypeptides.
  • “Biologically active” is defined herein as meaning that the ErbB3/4 ligand, for example, the variant or fragment, is capable of binding to ErbB3 and/or ErbB4, optionally wherein binding to ErbB3 and/or ErbB4 induces the heterodimerization of said ErbB3 or said ErbB4 with ErbB2, and optionally wherein said induction of heterodimerization of ErbB3 or ErbB4 with ErbB2 stimulates intrinsic kinase activity, which leads to tyrosine phosphorylation.
  • In some embodiments, the ErbB3/4 ligand is a fragment of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26 wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95 or at least 100 amino acids of the sequence recited in any one of SEQ ID NOs: 1-26.
  • In some embodiments, the ErbB3/4 ligand is a variant of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26.
  • References to a percentage sequence identity between two amino acid sequences means that, when aligned, that percentage of amino acids are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987) Supplement 30. A preferred alignment is determined by the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. The Smith-Waterman homology search algorithm is disclosed in Smith & Waterman (1981) Adv. Appl. Math. 2:482-489.
  • In some embodiments, the ErbB3/4 ligand is a variant of a fragment of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95 or at least 100 amino acids of the sequence recited in any one of SEQ ID NOs: 1-26, and wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the fragment.
  • In some embodiments, the ErbB3/4 ligand is a fragment of a biologically active variant of the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in any one of SEQ ID NOs: 1-26, and wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95 or at least 100 amino acids of the variant.
  • In some embodiments, the ErbB3/4 ligand is a non-naturally occurring ligand, for example a synthetic ligand or an anti-ErbB3/4 antibody. Methods of generating antibodies against a target of interest are well known in the art. In some embodiments, the anti-ErbB3/4 antibody is an agonistic anti-ErbB3/4 antibody. Preferably, the antibody is biologically active. Any suitable antibodies may be used, for example, as described herein.
  • ErbB3/4 ligands can be identified using methods known in the art, for example, binding of GST-tagged candidate ligands to insect-cell-expressed ErbB receptors causing tyrosine phosphorylation (Carraway 3rd et al., (1997) Nature 387(6632):512-6).
  • In some embodiments, the ErbB3/4 ligand comprises an EGF domain or EGF-like domain. EGF and EGF-like domains are evolutionary conserved protein domains that are recognised in the art (see, for example, Wouters et al. (2005) Protein Sci. 14(4): 1091-1103). Accordingly, in some embodiments in which the ErbB3/4 ligand is a biologically active fragment of at least one naturally occurring ErbB3/4 ligand, the fragment comprises an EGF domain or an EGF-like domain.
  • In some embodiments, the ErbB3/4 ligand is human neuregulin β-1. In some embodiments, the human neuregulin β-1 has the amino acid sequence recited in SEQ ID NO: 27. Human neuregulin β-1 is a fragment of a neuregulin polypeptide, wherein the fragment comprises an EGF-like domain. In a preferred embodiment, the ErbB3/4 ligand comprises or consists of the EGF-like domain of human neuregulin β-1.
  • In some embodiments the ErbB3/4 ligand has similar biological activity to recombinant human neuregulin β-1.
  • In some embodiments, the ErbB3/4 ligand is a fragment of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 62, or at least 64 amino acids of the sequence recited in SEQ ID NO: 27.
  • In some embodiments, the ErbB3/4 ligand is a variant of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in SEQ ID NO: 27.
  • In some embodiments, the ErbB3/4 ligand is a variant of a fragment of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 62 or at least 64 amino acids of the sequence recited in SEQ ID NO: 27, and wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the fragment.
  • In some embodiments, the ErbB3/4 ligand is a fragment of a biologically active variant of the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, wherein the variant has an amino acid sequence that is at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the polypeptide having the amino acid sequence recited in SEQ ID NO: 27, and wherein the fragment comprises at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 62 or at least 64 amino acids of the variant.
  • The ErbB3/4 ligand may be present at any suitable concentration. For example, the ErbB3/4 ligand may be present at a concentration of between 0.05 and 500 nM, between 0.05 and 100 nM, between 0.05 and 50 nM, between 0.05 and 10 nM, between 0.05 and 5 nM, between 0.05 and 1 nM, between 0.5 and 500 nM, between 0.5 and 100 nM, between 0.5 and 50 nM, between 0.5 and 10 nM, between 0.5 nM and 1 nM, between 1 and 10 nM, between 3 and 10 nM, between 3 and 8 nM, between 4 and 6 nM. For example, the ErbB3/4 ligand may be present at a concentration of approximately 5 nM. In some embodiments, the ErbB3/4 ligand is used at a concentration of at least 0.05 nM, at least 0.5 nM, at least M, at least 3 nM, at least 4 nM, at least 5 nM, at least 10 nM, at least 50 nM, or at least 100 nM.
  • Additionally to the ErbB3/4 ligand, cell culture media generally contain a number of components which are necessary to support maintenance and/or expansion of the cultured cells. A cell culture medium of the invention will therefore normally contain many other components in addition to an ErbB3/4 ligand. Suitable combinations of components can readily be formulated by the skilled person, taking into account the disclosure herein. A culture medium according to the invention will generally be a nutrient solution comprising standard cell culture components, such as amino acids, vitamins, inorganic salts, a carbon energy source, and a buffer as described in more detail below. Other standard cell culture components that may be included in the culture include hormones, such as progesterone, proteins, such as albumin, catalase, insulin and transferrin. These other standard cell culture components make up the “basal” culture medium.
  • A culture medium according to the invention may be generated by modification of an existing cell culture medium. The skilled person will understand from common general knowledge the types of culture media that might be suitable for modification for use in epithelial stem cell culture. Suitable cell culture media are available commercially, and include, but are not limited to, Dulbecco's Modified Eagle Media (DMEM), Minimal Essential Medium (MEM), Knockout-DMEM (KO-DMEM), Glasgow Minimal Essential Medium (G-MEM), Basal Medium Eagle (BME), DMEM/Ham's F12, Advanced DMEM/Ham's F12, Iscove's Modified Dulbecco's Media and Minimal Essential Media (MEM), Ham's F-10, Ham's F-12, Medium 199, and RPMI 1640 Media. Examples of suitable “basal” culture are also provided in WO2012/168930, WO2010/090513, and WO 2012/014076.
  • In some embodiments, the basal medium is AdDF+++. In some embodiments, the AdDF+++ comprises Advanced DMEM/F12 (Dulbecco's Modified Eagle Medium/Ham's F-12), GlutaMax, HEPES, penicillin/streptomycin, primocin.
  • In some embodiments, the basal medium is D10F. In some embodiments, the D10F comprises 31966-DMEM (Dulbecco's Modified Eagle Medium), fetal bovine serum (FBS) and penicillin/streptomycin.
  • Thus, in some embodiments, one of these pre-existing cell culture media is used as the basal culture medium to which is added the one or more Erb3/4 ligands. As mentioned above, the culture medium of the invention additionally comprises one or more ligands of FGFR2b (for example, FGF7 and/or FGF10) and/or one or more Wnt agonists (preferably an agonistic Lgr5 ligand, preferably Rspondin). In some embodiments, the culture medium of the invention comprises the following additional components: (i) FGF7 and/or FGF10 and (ii) Rspondin. In some embodiments, the culture medium of the invention comprises the following additional components: (i) FGF7 and/or FGF10, (ii) Rspondin and (iii) one or more further receptor tyrosine kinase ligands.
  • In some embodiments, the culture medium of the invention further comprises one or more components selected from: a BMP inhibitor (for example, Noggin), B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor (for example, A83-01), a p38 MAP kinase inhibitor (for example, SB 202190).
  • In some embodiments, the culture medium of the invention comprises an ErbB3/4 ligand, Rspondin, a BMP inhibitor (for example, Noggin), B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor (for example, A83-01), a p38 MAP kinase inhibitor (for example, SB 202190), FGF7 and FGF10, and optionally one or more additional components selected from: one or more further receptor tyrosine kinase ligands (for example, EGF, amphiregulin, TGF-alpha, PDGF), a p53 stabilizing agent and a Wnt agonist (for example, Wnt3a).
  • As will be apparent to the skilled reader, the preferred culture methods of the invention are advantageous because feeder cells are not required. Feeder cell layers are often used to support the culture of stem cells, and to inhibit their differentiation. A feeder cell layer is generally a monolayer of cells that is co-cultured with, and which provides a surface suitable for growth of, the cells of interest. The feeder cell layer provides an environment in which the cells of interest can grow. Feeder cells are often mitotically inactivated (e.g. by irradiation or treatment with mitomycin C) to prevent their proliferation. The use of feeder cells is undesirable, because it complicates passaging of the cells (the cells must be separated from the feeder cells at each passage, and new feeder cells are required at each passage). The use of feeder cells can also lead to contamination of the desired cells with the feeder cells. This is clearly problematic for any medical applications, and even in a research context, complicates analysis of the results of any experiments performed on the cells. The culture media of the invention are particularly advantageous because they can be used to culture cells without feeder cell contact, i.e. the methods of the invention do not require a layer of feeder cells to support the cells whose growth is being sponsored.
  • Accordingly, the compositions of the invention may be feeder cell-free compositions. A composition is conventionally considered to be feeder cell-free if the cells in the composition have been cultured for at least one passage in the absence of a feeder cell layer. A feeder cell-free composition of the invention will normally contain less than about 5%, less than about 4%, less than about 3%, less than about 2%, less than about 1% feeder cells (expressed as a % of the total number of cells in the composition) or preferably no feeder cells at all.
  • BMP Inhibitors
  • The culture media of the invention may comprise one or more bone morphogenetic protein (BMP) inhibitor.
  • In some embodiments, the culture medium may comprise any suitable BMP inhibitor. BMPs bind as a dimeric ligand to a receptor complex consisting of two different receptor serine/threonine kinases, type I and type II receptors. The type II receptor phosphorylates the type I receptor, resulting in the activation of this receptor kinase. The type I receptor subsequently phosphorylates specific receptor substrates (SMAD), resulting in a signal transduction pathway leading to transcriptional activity. Advantageously, BMP inhibitors promote expression of Lgr5, and so the presence of a BMP inhibitor in a culture medium of the invention will likely result in more proliferative organoids than if the BMP inhibitor is absent. In some embodiments, cells cultured with a BMP inhibitor have upregulated expression of Lgr5 compared to cells cultured without a BMP inhibitor. Therefore, addition of a BMP inhibitor typically results in more proliferative organoids.
  • A BMP inhibitor is defined as an agent that binds to a BMP molecule to form a complex wherein the BMP activity is neutralized, for example by preventing or inhibiting the binding of the BMP molecule to a BMP receptor. Alternatively, said inhibitor is an agent that acts as an antagonist or reverse agonist. This type of inhibitor binds with a BMP receptor and prevents binding of a BMP to said receptor. An example of a latter agent is an antibody that binds a BMP receptor and prevents binding of BMP to the antibody-bound receptor.
  • A BMP inhibitor may be added to the media in an amount effective to inhibit a BMP-dependent activity in a cell to at most 90%, more preferred at most 80%, more preferred at most 70%, more preferred at most 50%, more preferred at most 30%, more preferred at most 10%, more preferred 0%, relative to a level of a BMP activity in the absence of said inhibitor, as assessed in the same cell type. As is known to a skilled person, a BMP activity can be determined by measuring the transcriptional activity of BMP, for example as exemplified in Zilberberg et al., 2007. BMC Cell Biol. 8:41.
  • Several classes of natural BMP-binding proteins are known, including Noggin (Peprotech), Chordin and chordin-like proteins (R&D systems) comprising chordin domains, Follistatin and follistatin-related proteins (R&D systems) comprising a follistatin domain, DAN and DAN-like proteins (R&D systems) comprising a DAN cysteine-knot domain, sclerostin/SOST (R&D systems), decorin (R&D systems), and alpha-2 macroglobulin (R&D systems).
  • Examples of BMP inhibitors for use in a method of the invention are Noggin, DAN, and DAN-like proteins including Cerberus and Gremlin (R&D systems). These diffusible proteins are able to bind a BMP ligand with varying degrees of affinity and inhibit their access to signalling receptors. The addition of any of these BMP inhibitors to the basal culture medium prevents the loss of stem cells.
  • A preferred BMP inhibitor is Noggin. Accordingly, in some embodiments, the culture medium as described herein comprises Noggin. In the context of a culture medium of the invention, Noggin is also referred to herein as “No”. Noggin is preferably added to the basal culture medium at a concentration of at least 10 ng/ml, for example, at least 20 ng/ml, more preferred at least 25 ng/ml. A still more preferred concentration is about 25 ng/ml.
  • During culturing of stem cells, said BMP inhibitor may be added to the culture medium when required, for example, daily or every other day. The BMP inhibitor is preferably added to the culture medium every second day. The culture medium may be refreshed when required, for example, daily or every other day.
  • Wnt Agonists
  • The culture media of the invention may comprise one or more Wnt agonist. The Wnt signalling pathway is defined by a series of events that occur when the cell-surface Wnt receptor complex, comprising a Frizzled receptor, LRP and LGR is activated, usually be an extracellular signalling molecule, such as a member of the Wnt family. This results in the activation of Dishevelled family proteins which inhibit a complex of proteins that includes axin, GSK-3, and the protein APC to degrade intracellular β-catenin. The resulting enriched nuclear β-catenin enhances transcription by TCF/LEF family transcription factors.
  • A Wnt agonist is defined as an agent that activates TCF/LEF-mediated transcription in a cell. Wnt agonists are therefore selected from true Wnt agonists that bind and activate the Wnt receptor complex including any and all of the Wnt family proteins, an inhibitor of intracellular β-catenin degradation, a GSK inhibitor (such as CHIR9901) and activators of TCF/LEF.
  • In some embodiments, a Wnt agonist is a secreted glycoprotein including Wnt-1/Int-1, Wnt-2/Irp (1 nM-related Protein), Wnt-2b/13, Wnt-3/Int-4, Wnt-3a (R&D systems), Wnt-4, Wnt-5a, Wnt-5b, Wnt-6 (Kirikoshi H et al 2001 Biochem Biophys Res Com 283 798-805), Wnt-7a (R&D systems), Wnt-7b, Wnt-8a/8d, Wnt-8b, Wnt-9a/14, Wnt-9b/14b/15, Wnt-10a, Wnt-10b/12, WnM 1, and Wnt-16. An overview of human Wnt proteins is provided in “THE WNT FAMILY OF SECRETED PROTEINS”, R&D Systems Catalog, 2004. In some embodiments, the Wnt agonist is an inhibitor of RNF43 or ZNRF3. It has been shown that RNF43 and ZNRF3 reside in the cell membrane and negatively regulate levels of the Wnt receptor complex in the membrane, probably by ubiquitination of Frizzled. Therefore, the inventors hypothesise that inhibition of RNF43 or ZNRF3 with antagonistic antibodies, RNAi or small molecule inhibitors would indirectly stimulate the Wnt pathway. RNF43 and ZNRF3 have a catalytic ring domain (with ubiquitination activity), which can be targeted in small molecule inhibitor design. Several anti-RNF43 antibodies and several anti-ZNRF3 antibodies are available commercially. In some embodiments, such antibodies are suitable Wnt agonists in the context of the invention.
  • In a preferred embodiment, the Wnt agonist in the culture medium is any agonist able to stimulate the Wnt pathway via the Lgr5 cell surface receptor, i.e. in a preferred embodiment, the Wnt agonist in the culture medium is an Lgr5 agonist. Known Lgr5 agonists include Rspondin, fragments and derivatives thereof, and anti-Lgr5 antibodies (e.g. see WO 2012/140274 and De Lau, W. et al. Nature, 2011 Jul. 4; 476(7360):293-7). A preferred Lgr5 agonist is Rspondin. Any suitable Rspondin may be used, for example, it may be selected from one or more of Rspondin 1, Rspondin 2, Rspondin 3 and Rspondin 4 or derivatives thereof. For example, any of Rspondin 1 (NU206, Nuvelo, San Carlos, Calif.), Rspondin 2 ((R&D systems), Rspondin 3, and Rspondin 4) may be used. Rspondin 1, 2, 3, and 4 are also referred to herein as “Rspondin 1-4”. Fragments of Rspondin may be used as the Wnt agonist. For example, in some embodiments the Wnt agonist is a fragment of Rspondin comprising or consisting of the furin domain. Examples of suitable Rspondin fragments are represented by the sequence of amino acids recited in SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30 or SEQ ID NO: 31 or of sequences with more than 70, 80, 90 or 99% identity to any one of SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30 or SEQ ID NO: 31. In some embodiments the Lgr5 agonist is an anti-Lgr5 antibody, more preferably an agonistic anti-Lgr5 antibody. An example of an agonistic anti-Lgr5 antibody is 1D9 (available commercially from BD Biosciences, BDB562733, No.:562733). Therefore, in one embodiment, the agonist is the antibody 1D9. The VL of antibody 1D9 is represented by SEQ ID NO: 32 and the VH is represented by SEQ ID NO: 33. Therefore, in one embodiment, the agonist is an antibody comprising or consisting of SEQ ID NO: 32 and/or SEQ ID NO: 33.
  • In some embodiments, the Wnt agonist in the culture medium is any agonist able to stimulate the Wnt pathway via the Lgr4 cell surface receptor, i.e. in some embodiments, the Wnt agonist in the culture medium is an Lgr4 agonist.
  • In some embodiments, the Wnt agonist in the culture medium is any agonist able to stimulate the Wnt pathway via the Lgr6 cell surface receptor, i.e. in some embodiments, the Wnt agonist in the culture medium is an Lgr6 agonist.
  • The Wnt agonist may be added to the media in an amount effective to stimulate a Wnt activity in a cell by at least 10%, more preferred at least 20%, more preferred at least 30%, more preferred at least 50%, more preferred at least 70%, more preferred at least 90%, more preferred at least 100%, relative to a level of said Wnt activity in the absence of said molecule, as assessed in the same cell type. As is known to a skilled person, Wnt activity can be determined by measuring the transcriptional activity of Wnt, for example by pTOPFLASH and pFOPFLASH Tcf luciferase reporter constructs (Korinek et al., 1997. Science 275:1784-1787).
  • A soluble Wnt agonist, such as Wnt-3a, may be provided in the form of Wnt conditioned media. For example, about 10% to about 30%, e.g. about 10 ng/ml to about 10 μg/ml, preferably about 1 μg/ml, Wnt conditioned media may be used.
  • Rspondin 1-4 may be provided in the form of Rspo conditioned media. For example, about 10% to about 30%, e.g. about 10 ng/ml to about 10 μg/ml, preferably about 1 μg/ml, Rspo conditioned media may be used.
  • One or more, for example, 2, 3, 4 or more Wnt agonists may be used in the culture medium. In one embodiment, the culture medium comprises an Lgr5 agonist, for example Rspondin, and additionally comprises a further Wnt agonist. In this context, the further Wnt agonist may, for example, be selected from the group consisting of Wnt-3a, a GSK-inhibitor (such as CHIR99021), Wnt-5, Wnt-6a and Norrin. In one embodiment, the culture medium comprises Rspondin and additionally comprises a soluble Wnt ligand, such as Wnt3a. Addition of a soluble Wnt ligand has been shown to be particularly advantageous for expansion of human epithelial stem cells (as described in WO2012/168930).
  • Any suitable concentration of Wnt agonist, e.g. Rspondin, may be used, for example, at least 200 ng/ml, more preferred at least 300 ng/ml, more preferred at least 500 ng/ml. A still more preferred concentration of Rspondin is at least 500 ng/ml or about 1 μg/ml.
  • During culturing of stem cells, said Wnt agonist may be added to the culture medium when required, for example, daily or every other day. The Wnt agonist is preferably added to the culture medium every second day.
  • Antibodies
  • Antibodies, such as anti-ErbB3/4 antibodies, agonistic anti-Lgr5 antibodies or antagonistic TGF-beta inhibitors (see below), used in the invention may be any antibodies, fragments, etc. A conventional antibody is comprised of two identical heavy chains and two identical light chains that are joined by disulfide bonds. Each heavy and light chain contains a constant region and a variable region. Each variable region contains three CDRs which are primarily responsible for binding an epitope of an antigen. They are referred to as CDR1, CDR2, and CDR3, numbered sequentially from the N-terminus, of which the CDR3 region comprises the most variable region and normally provides a substantial part of the contact residues to a target. The more highly conserved portions of the variable regions are called the “framework regions”.
  • The term antibody is used herein in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies) of any isotype such as IgG, IgM, IgA, IgD and IgE, polyclonal antibodies including recombinant polyclonal antibodies, Oligoclonics, multispecific antibodies, chimeric antibodies, nanobodies, diabodies, BiTE's, Tandabs, mimetobodies, bispecific antibodies, humanized antibodies, human antibodies, deimmunised antibodies and antibody fragments. In addition, scaffolds will be covered under this term, such as Anticalins, Ankarins, etc. An antibody reactive with a the specific epitopes of the Lgr proteins discussed above can be generated by recombinant methods such as selection of libraries of recombinant antibodies in phage or similar vectors, or by immunizing an animal with the Lgr epitopes of nucleic acid encoding them.
  • In one embodiment, an antibody according to the invention comprises a single domain antibody, a F(ab′)2, Fab, Fab′, or single chain Fv (scFv) fragment. An Fc fragment, which for example activates complement and may bind to Fc receptors, can be present but is not required for an antibody and variants or derivatives thereof. A scFv fragment is an epitope-binding fragment that contains at least one fragment of an antibody heavy chain variable region (VH) linked to at least one fragment of an antibody light chain variable region (VL). The linker may be a short, flexible peptide selected to assure that the proper three-dimensional folding of the VL and VH regions occurs once they are linked so as to maintain the target molecule binding-specificity of the whole antibody from which the single-chain antibody fragment is derived. The carboxyl terminus of the VL or VH sequence may be covalently linked by a linker to the amino acid terminus of a complementary VL or VH sequence.
  • The antibody may be a diabody, mimetibody, nanobody, and/or a bispecific antibody. A nanobody is a single domain antibody that occurs naturally in camelids. In contrast to standard antibodies, nanobodies are relatively simple proteins comprising only a heavy chain-like variable region. Bispecific antibodies are artificially engineered monoclonal antibodies that consist of two distinct binding sites and are capable of binding two different epitopes. Examples of bispecific antibodies are discussed in more detail below in the section on dual-targeting and multi-targeting agonists.
  • The antibody may be a chimeric antibody comprising a binding portion, for example the variable region or part thereof of the heavy and light chains, of a non-human antibody, while the remainder portion, for example the constant region of the heavy and light chains, is of a human antibody. A chimeric antibody may be produced by recombinant processes well known in the art, and has an animal variable region and a human constant region.
  • The antibody may be a human antibody or a humanized antibody. The term “human antibody” means an antibody in which the variable and constant domain sequences are derived from human sequences. In a humanized antibody, only the complementarity determining regions (CDRs), which are responsible for antigen binding and specificity are animal derived and have an amino acid sequence corresponding to the animal antibody, and substantially all of the remaining portions of the molecule (except, in some cases, small portions of the framework regions within the variable region) are human derived and correspond in amino acid sequence to a human antibody. Methods for humanizing non-human antibodies are known in the art. As is known to the skilled person, antibodies such as rat antibodies can be humanized by grafting their CDRs onto the variable light (VL) and variable heavy (VH) frameworks of human Ig molecules, while retaining those rat framework residues deemed essential for specificity and affinity. Overall, CDR grafted antibodies consist of more than 80% human amino acid sequences.
  • In some embodiments, the antibody is a deimmunised antibody in which the T and B cell epitopes have been eliminated. They have reduced immunogenicity when applied in vivo.
  • TGF-Beta Inhibitor
  • The culture media of the invention may comprise a TGF-beta inhibitor. The presence of a TGF-beta inhibitor in the media is advantageous because it increases human organoid formation efficiency. TGF-beta signalling is involved in many cellular functions, including cell growth, cell fate and apoptosis. Signalling typically begins with binding of a TGF-beta superfamily ligand to a type II receptor which recruits and phosphorylates a type I receptor. The type I receptor then phosphorylates SMADs, which act as transcription factors in the nucleus and regulate target gene expression.
  • The TGF-beta superfamily ligands comprise bone morphogenic proteins (BMPs), growth and differentiation factors (GDFs), anti-mullerian hormone (AMH), activin, nodal and TGF-betas. In general, Smad2 and Smad3 are phosphorylated by the ALK4, 5 and 7 receptors in the TGF-beta/activin pathway. By contrast, Smad1, Smad5 and Smad8 are phosphorylated as part of the bone morphogenetic protein (BMP) pathway. Although there is some cross-over between pathways, in the context of this invention, a “TGF-beta inhibitor” or an “inhibitor of TGF-beta signalling” is preferably an inhibitor of the TGF-beta pathway which acts via Smad2 and Smad3. Therefore, in some embodiments the TGF-beta inhibitor is not a BMP inhibitor, e.g. the TGF-beta inhibitor is not Noggin. In some embodiments, a BMP inhibitor is added to the culture medium in addition to the TGF-beta inhibitor (see above).
  • Thus the TGF-beta inhibitor may be any agent that reduces the activity of the TGF-beta signalling pathway, preferably the signalling pathway that acts via Smad2 and/or Smad3, more preferably the signalling pathway that acts via ALK4, ALK5 or ALK7. There are many ways of disrupting the TGF-beta signaling pathway that are known in the art and that can be used in conjunction with this invention. For example, the TGF-beta signaling may be disrupted by: inhibition of TGF-beta expression by a small-interfering RNA strategy; inhibition of furin (a TGF-beta activating protease); inhibition of the pathway by physiological inhibitors; neutralisation of TGF-beta with a monoclonal antibody; inhibition with small-molecule inhibitors of TGF-beta receptor kinase 1 (also known as activin receptor-like kinase, ALK5), ALK4, ALK6, ALK7 or other TGF-beta-related receptor kinases; inhibition of Smad 2 and Smad 3 signaling e.g. by overexpression of their physiological inhibitor, Smad 7, or by using thioredoxin as an Smad anchor disabling Smad from activation (Fuchs, O. Inhibition of TGF-Signaling for the Treatment of Tumor Metastasis and Fibrotic Diseases. Current Signal Transduction Therapy, Volume 6, Number 1, January 2011, pp. 29-43(15)).
  • Various methods for determining if a substance is a TGF-beta inhibitor are known and might be used in conjunction with the invention. For example, a cellular assay may be used in which cells are stably transfected with a reporter construct comprising the human PAI-1 promoter or Smad binding sites, driving a luciferase reporter gene. Inhibition of luciferase activity relative to control groups can be used as a measure of compound activity (De Gouville et al., Br J Pharmacol. 2005 May; 145(2): 166-177).
  • A TGF-beta inhibitor according to the present invention may be a protein, peptide, small-molecules, small-interfering RNA, antisense oligonucleotide, aptamer or antibody. The inhibitor may be naturally occurring or synthetic. In one embodiment, the TGF-beta inhibitor is an inhibitor of ALK4, ALK5 and/or ALK7. For example, the TGF-beta inhibitor may bind to and directly inhibit ALK4, ALK5 and/or ALK7. Examples of preferred small-molecule TGF-beta inhibitors that can be used in the context of this invention include the small molecule inhibitors listed in Table 1:
  • TABLE 1
    Small-molecule TGF-beta inhibitors targeting receptor kinases
    IC50 Mol
    Inhibitor Targets (nM) Wt Name Formula
    A83-01 ALK5 12 421.52 3-(6-Methyl-2- C25H19N5S
    (TGF- pyridinyl)-N-phenyl-
    βR1) 4-(4-quinolinyl)-1H-
    ALK4 45 pyrazole-1-
    ALK7 7.5 carbothioamide
    SB-431542 ALK5 94 384.39 4-[4-(1,3- C22H16N4O3
    ALK4 benzodioxol-5-yl)-5-
    ALK7 (2-pyridinyl)-1H-
    imidazol-2-
    yl]benzamide
    SB-505124 ALK5 47 335.4 2-(5- C20H21N3O2
    ALK4 129 benzo[1,3]dioxol-5-
    yl-2-tert-butyl-
    3Himidazol-
    4-yl)-6-
    methylpyridine
    hydrochloride hydrate
    SB-525334 ALK5 14.3 343.42 6-[2-(1,1- C21H21N5
    Dimethylethyl)-5-(6-
    methyl-2-pyridinyl)-
    1H-imidazol-4-
    yl]quinoxaline
    SD-208 ALK5 49 352.75 2-(5-Chloro-2- C17H10ClFN6
    fluorophenyl)-4-[(4-
    pyridyl)amino]pteridine
    LY-36494 TGR-βRI 59 272.31 4-[3-(2-Pyridinyl)-1H- C17H12N4
    TGF-βRII 400 pyrazol-4-yl]-quinoline
    MLK-7K 1400
    SJN-2511 ALK5 23 287.32 2-(3-(6-Methylpyridine- C17H13N5
    2-yl)-1H-pyrazol-4-yl)-
    1,5-naphthyridine
  • In some embodiments, the TGF-beta inhibitor is a small molecule inhibitor optionally selected from the group consisting of: A83-01, SB-431542, SB-505124, SB-525334, LY 364947, SD-208 and SJN 2511.
  • In some embodiments, no more than one TGF beta inhibitor is present in the medium. In other embodiments, more than one TGF beta inhibitor is present in the medium, e.g. 2, 3, 4 or more. In some embodiments, a medium of the invention comprises one or more of any of the inhibitors listed in table 1. A medium may comprise any combination of one inhibitor with another inhibitor listed. For example, a medium may comprise SB-525334 or SD-208 or A83-01; or SD-208 and A83-01. The skilled person will appreciate that a number of other small-molecule inhibitors exist that are primarily designed to target other kinases, but at high concentrations may also inhibit TGF-beta receptor kinases. For example, SB-203580 is a p38 MAP kinase inhibitor that, at high concentrations (for example, approximate 10 μM or more) is thought to inhibit ALK5. Any such inhibitor that inhibits the TGF-beta signalling pathway can also be used in the context of this invention.
  • In some embodiments, the TGF beta inhibitor is present at at least 5 nM, for example, at least 50 nM, at least 100 nM, at least 300 nM, at least 450 nM, at least 475 nM, for example 5 nM-500 mM, 10 nM-100 mM, 50 nM-700 uM, 50 nM-10 uM, 100 nM-1000 nM, 350-650 nM or more preferably about 500 nM.
  • A83-01 may be added to the medium at a concentration of between 10 nM and 10 uM, or between 1 uM and 8 uM, or between 4 uM and 6 uM. For example, A83-01 may be added to the medium at about 5 uM. The skilled person would know how to determine the concentration of other TGF beta inhibitors for use in the invention.
  • Receptor Tyrosine Kinase Ligands
  • ErbB3/4 ligands are receptor tyrosine kinase ligands. The culture media of the invention may further comprise one or more additional receptor tyrosine kinase ligands that are not themselves ErbB3/4 ligands. An example of a receptor tyrosine kinase ligand for use in the invention is EGF, which is the ligand for the receptor tyrosine kinase EGFR. Many receptor tyrosine kinase ligands are also mitogenic growth factors.
  • The culture media of the invention may comprise one or more mitogenic growth factor. The one or more mitogenic growth factor may be selected from a family of growth factors comprising epidermal growth factor (EGF, Peprotech), Transforming Growth Factor-alpha (TGF-alpha, Peprotech), basic Fibroblast Growth Factor (bFGF, Peprotech), brain-derived neurotrophic factor (BDNF, R&D Systems), Platelet Derived Growth Factor (PDGF, Peprotech), amphiregulin (R&D Systems). A preferred PDGF is PDGF-CC. In the context of a culture medium of the invention, EGF is also referred to herein as “E” and FGF is also referred to herein as “F”.
  • In some embodiments, the receptor tyrosine kinase ligand for use in the invention is one or more mitogenic growth factors selected from human EGF, human PDGF (e.g. human PDGF-CC), human amphiregulin and human TGF-alpha.
  • EGF
  • EGF is a potent mitogenic factor for a variety of cultured ectodermal and mesodermal cells and has a profound effect on the differentiation of specific cells in vivo and in vitro and of some fibroblasts in cell culture. The EGF precursor exists as a membrane-bound molecule which is proteolytically cleaved to generate the 53-amino acid peptide hormone that stimulates cells.
  • The inventors have found that EGF is not essential in the culture medium of the invention for the growth of breast organoids. Therefore, in some embodiments, the culture medium of the invention does not contain EGF. However, the inventors found that the addition of EGF may be beneficial for growth of breast organoids. Therefore, in some embodiments, the culture media of the invention further comprises EGF.
  • In embodiments where the culture medium of the invention comprises EGF, EGF is preferably added to the basal culture medium at a concentration of between 1 and 500 ng/ml or of at least 1 and not higher than 500 ng/ml. A preferred concentration is at least 1, 5, 10, 20, 25, 30, 40, 45, or 50 ng/ml and not higher than 500, 450, 400, 350, 300, 250, 200, 150, or 100 ng/ml. A more preferred concentration is at least 1 and not higher than 100 ng/ml. The same concentrations could be used for a different mitogenic growth factor, such as a PDGF or a FGF. If more than one FGF is used, for example FGF7 and FGF10, the concentration of a FGF is as defined above and refers to the total concentration of FGF used.
  • FGFR2b Ligands
  • FGF7 and FGF10 are proteins that belong to the fibroblast growth factor (FGF) family of proteins. FGF family members possess broad mitogenic and cell survival activities, and are involved in a variety of biological processes, including embryonic development, cell growth, morphogenesis, tissue repair, tumor growth and invasion. FGFs stimulate cells by interacting with cell surface tyrosine kinase receptors (FGFR). Four closely related receptors (FGFR1-FGFR4) have been identified. FGFR1-FGFR3 genes have been shown to encode multiple isoforms, and these isoforms can be critical in determining ligand specificity. Most FGFs bind more than one receptor (Ornitz J Biol Chem. 1998 Feb. 27; 273 (9):5349-57). However, FGF10 and FGF7 are unique among FGFs in that they interact only with a specific isoform of FGFR2, designated FGFR2b which is expressed exclusively by epithelial cells (Igarashi, J Biol Chem. 1998 273(21):13230-5).
  • In preferred embodiments, the culture medium of the invention comprises a ligand of FGFR2b (e.g. FGF1, FGF3, FGF7, FGF10 or FGF22). In some embodiments, the ligand of FGFR2b is a high-affinity ligand of FGFR2b. In some embodiments, the ligand of FGFR2b binds to FGFR2b but does not bind to any other FGFR isoforms. In some embodiments, the FGFR2b ligand is a member of the FGF7 subfamily (e.g. FGF7, FGF10 or FGF22). In a preferred embodiment, the FGFR2b ligand is FGF10 and/or FGF7. In some embodiments, no more than one FGFR2b ligand is used. In other embodiments, two or more FGFR2b ligands are used, e.g. 2, 3 or more. In some embodiments, the culture medium comprises FGF7 and FGF10. In some embodiments, biologically active fragments or variants of naturally occurring FGFR2b ligands, for example, biologically active fragments or variants of FGF7 and/or FGF10 are used as FGFR2b ligands. In some embodiments, biologically active synthetic ligands are used as FGFR2b ligands in the culture medium of the invention. “Biologically active” as used in this context means that the FGFR2b ligand, for example, the fragment or variant of a naturally occurring FGFR2b ligand, or a synthetic FGFR2b ligand, is capable of binding to FGFR2b and eliciting similar downstream signalling compared to FGF7 and/or FGF10, for example eliciting tyrosine phosphorylation of FRS2a (FGF receptor substrate 2a) or FRS2p (FGF receptor substrate 2p). Tyrosine phosphorylation of FRS2a and/or FRS2p is the earliest cytosolic event in the signalling pathways activated by FGF7 and/or FGF10. In some embodiments, the FGFR2b ligand is substituted with a compound that activates the FGFR2 or FGFR4 pathway (a “FGF-pathway activator”).
  • In some embodiments, the one or more FGFR2b ligands used in the culture medium of the invention is selected from the following: human FGF7, human FGF10, human FGF22, human FGF1 or human FGF22.
  • In a further embodiment, a combination of mitogenic growth factors such as, for example, EGF and FGF7, or EGF and FGF10, is added to the basal culture medium. In a further preferred embodiment, a combination of mitogenic growth factors such as, for example, (i) FGF7 and FGF10, (ii) EGF, FGF7 and FGF10, or (iii) PDGF, FGF7 and FGF10, or (iv) EGF, PDGF, amphiregulin, FGF7 and FGF10 is added to the culture medium.
  • In some embodiments, TGF-alpha or amphiregulin are added to the basal culture medium. These mitogenic growth factors may replace EGF. In some embodiments, hepatocyte growth factor (HGF) is added to the culture medium.
  • During culturing of stem cells, a combination of receptor tyrosine kinase ligands (e.g. EGF, FGF10 and FGF7) is preferably added to the culture medium when required, for example, daily or every other day. They may be added singularly or in combination. It is preferable that they are added every second day.
  • ROCK Inhibitors
  • ROCK inhibitors, such as Y-27632 (10 μM; Sigma), can be included in any of the media described, in particular in the first few days of culture before performing cell sorting experiments, because it is known to avoid anoikis (a form of programmed cell death which is induced by anchorage-dependent cells detaching from the surrounding extracellular matrix). Therefore, any of the media defined herein, may additionally comprise a ROCK inhibitor for the first few days. In some embodiments, the culture medium of the invention additionally comprises a ROCK inhibitor, such as Y-27632, for example for the first few days of culture before performing cell sorting experiments.
  • A further embodiment a culture medium of the invention comprises a Rock (Rho-kinase) inhibitor. The addition of a Rock inhibitor was found to prevent anoikis, especially when culturing single stem cells. Said Rock inhibitor is preferably selected from R-(+)-trans-4-(1-aminoethyl)-N-(4-Pyridyl)cyclohexanecarboxamide dihydrochloride monohydrate (Y-27632, Sigma-Aldrich), 5-(1,4-diazepan-1-ylsulfonyl)isoquinoline (fasudil or HA1077, Cayman Chemical), and (S)-(+)-2-methyl-1-[(4-methyl-5-isoquinolinyl)sulfonyl]-hexahydro-1H-1,4-diazepine dihydrochloride (H-1 152, Tocris Bioschience). Said Rho-kinase inhibitor, for example Y-27632, is preferably added to the culture medium every second day during the first seven days of culturing said stem cells. A Rock inhibitor is preferably included in the medium in the first few days e.g. for the first 1, 2, 3, 4, 5, 6 or 7 days of culture after single cell seeding or after a split. Any suitable concentration of the Rock inhibitor may be used, for example, 1-200 uM, 1-100 uM, 5-50 uM or approximately 10 uM. A preferred concentration for Y27632 is 10 uM. Therefore, in some embodiments, the invention provides a method for culturing stem cells and/or a method for obtaining an organoid wherein a Rock inhibitor is added to the culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day. In some embodiments, the Rock inhibitor is not added to the culture medium after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • Addition of a Rock inhibitor is particularly important when culturing single stem cells (as mentioned above), i.e. when the starting material for an organoid is a single stem cell. Therefore, in some embodiments the invention provides a method for obtaining an organoid, wherein the method comprises culturing stem cells, optionally single stem cells, wherein a Rock inhibitor is added to the culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day, and optionally not adding the Rock inhibitor to the culture medium after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • The Rock inhibitor is less important, and sometimes not necessary, when culturing multiple cells, for example when the starting material for an organoid is a tissue fragment. Therefore, in some embodiments, the invention provides a method for obtaining an organoid, wherein the method comprises culturing stem cells, optionally a tissue fragment, wherein the Rock inhibitor is not added to the culture medium either at all or after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • After the cells are split into multiple cultures, a Rock inhibitor may be added to the culture medium in the same way, meaning for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day, after the split, particularly when the split involves taking single stem cells from a first culture and placing these into a second culture. If the split involves taking multiple stem cells from the first culture and placing these into a second culture then addition of a Rock inhibitor is less important, and sometimes not necessary. Therefore, in some embodiments, wherein the method for obtaining organoids or for culturing stem cells involves a split, optionally where a single cell is involved in the split, a Rock inhibitor is added to the new culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day, after the split. In some embodiments, wherein the method for obtaining organoids or for culturing stem cells involves a split, optionally where multiple cells are involved in the split, is not added to the culture medium either at all or after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • Notch Agonists
  • In yet a further embodiment, the culture medium of the invention further comprises a Notch agonist. Notch signaling has been shown to play an important role in cell-fate determination, as well as in cell survival and proliferation. Notch receptor proteins can interact with a number of surface-bound or secreted ligands, including but not limited to Delta 1, Jagged 1 and 2, and Delta-like 1, Delta-like 3, Delta-like 4. Upon ligand binding, Notch receptors are activated by serial cleavage events involving members of the ADAM protease family, as well as an intramembranous cleavage regulated by the gamma secretase presenilin. The result is a translocation of the intracellular domain of Notch to the nucleus where it transcriptionally activates downstream genes. A preferred Notch agonist is selected from Jagged 1 and Delta 1, or an active fragment or derivative thereof. A most preferred Notch agonist is DSL peptide (Dontu et al., 2004. Breast Cancer Res 6. R605-R615) with the sequence CDDYYYGFGCNKFCRPR. Said DSL peptide is preferably used at a concentration between 10 μM and 100 nM or at least 10 μM and not higher than 100 nM. The addition of a Notch agonist, especially during the first week of culturing, increases the culture efficiency by a factor of 2-3. Said Notch agonist is preferably added to the culture medium every second day during the first seven days of culturing said stem cells. Therefore, in some embodiments, the invention provides a method for culturing stem cells and/or a method for obtaining an organoid wherein a Notch agonist is added to the culture medium for the first 1, 2, 3, 4, 5, 6 or 7 days, optionally every second day. In some embodiments, the Notch agonist is not added to the culture medium after the first 2, 3, 4, 5, 6, 7, 8, 9 or 10 days.
  • A Notch agonist is defined as a molecule that stimulates a Notch activity in a cell by at least 10%, more preferred at least 20%, more preferred at least 30%, more preferred at least 50%, more preferred at least 70%, more preferred at least 90%, more preferred at least 100%, relative to a level of a Notch activity in the absence of said molecule. As is known to a skilled person, a Notch activity can be determined by measuring the transcriptional activity of Notch, for example by a 4xwtCBFl-luciferase reporter construct as described (Hsieh et al, 1996 Mol Cell. Biol. 16, 952-959).
  • Additional Components
  • The culture medium optionally comprises Nicotinamide and is preferably supplemented with one or more (e.g., 1, 2, 3 or all) of the compounds selected from the group consisting of B27, N-acetylcysteine and N2. Thus in some embodiments the culture medium further comprises one or more components selected from the group consisting of: nicotinamide, B27, N2 and N-Acetylcysteine. For example, in some embodiments, the culture medium further comprises B27, N-Acetylcysteine and Nicotinamide.
  • B27 (Invitrogen), N-Acetylcysteine (Sigma) and N2 (Invitrogen), and Nicotinamide (Sigma) are believed to control proliferation of the cells and assist with DNA stability. In the context of the invention, Nicotinamide is also referred to herein as “Nic”.
  • In some embodiments, Nicotinamide is present at 7-15 mM, for example about 10 mM.
  • In some embodiments, the B27 supplement is ‘B27 Supplement minus Vitamin A’ (available from Invitrogen, Carlsbad, Calif.; www.invitrogen.com; currently catalog no. 12587010; and from PAA Laboratories GmbH, Pasching, Austria; www.paa.com; catalog no. F01-002; Brewer et al., J Neurosci Res., 35(5):567-76, 1993) may be used to formulate a culture medium that comprises biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin.
  • The B27 Supplement supplied by PAA Laboratories GmbH comes as a liquid 50× concentrate, containing amongst other ingredients biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinol, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin. Of these ingredients at least linolenic acid, retinol, retinyl acetate and tri-iodothyronine (T3) are nuclear hormone receptor agonists. B27 Supplement may be added to a culture medium as a concentrate or diluted before addition to a culture medium. It may be used at a x final concentration or at other final concentrations. Use of B27 Supplement is a convenient way to incorporate biotin, cholesterol, linoleic acid, linolenic acid, progesterone, putrescine, retinol, retinyl acetate, sodium selenite, tri-iodothyronine (T3), DL-alpha tocopherol (vitamin E), albumin, insulin and transferrin into a culture medium of the invention. It is also envisaged that some or all of these components may be added separately to the culture medium instead of using the B27 Supplement. Thus, the culture medium may comprise some or all of these components.
  • In some embodiments, retinoic acid is absent from the B27 Supplement used in the culture medium, and/or is absent from the culture medium.
  • N2 Supplement′ is available from Invitrogen, Carlsbad, Calif.; www.invitrogen.com; catalog no. 17502-048; and from PAA Laboratories GmbH, Pasching, Austria; www.paa.com; catalog no. F005-004; Bottenstein & Sato, PNAS, 76(1):514-517, 1979. The N2 Supplement supplied by PAA Laboratories GmbH comes as a 100× liquid concentrate, containing 500 μg/ml human transferrin, 500 μg/ml bovine insulin, 0.63 μg/ml progesterone, 1611 μg/ml putrescine, and 0.52 μg/ml sodium selenite. N2 Supplement may be added to a culture medium as a concentrate or diluted before addition to a culture medium. It may be used at a x final concentration or at other final concentrations. Use of N2 Supplement is a convenient way to incorporate transferrin, insulin, progesterone, putrescine and sodium selenite into a culture medium of the invention. It is of course also envisaged that some or all of these components may be added separately to the culture medium instead of using the N2 Supplement. Thus, the culture medium may comprise some or all of these components.
  • In some embodiments in which the medium comprises B27, it does not also comprise N2. The embodiments of the present invention can therefore be adapted to exclude N2 when B27 is present, if desired.
  • In some embodiments N2 is not present in the culture medium.
  • In some embodiments in which the medium comprises N2, it does not also comprise B27. The embodiments of the present invention can therefore be adapted to exclude B27 when N2 is present, if desired.
  • In some embodiments B27 is not present in the culture medium.
  • In some embodiments the culture medium is supplemented with B27 and/or N2.
  • In some embodiments the basal medium is supplemented with 150 ng/ml to 250 ng/ml N-Acetylcysteine; preferably, the basal medium is supplemented with about 200 ng/ml N-Acetylcysteine.
  • In some embodiments the culture medium comprises progestin. In other embodiments the culture medium does not comprise progestin.
  • In some embodiments the culture medium comprises estradiol. In other embodiments the culture medium does not comprise estradiol.
  • In some embodiments the culture medium comprises hepatocyte growth factor. In other embodiments the culture medium does not comprise hepatocyte growth factor.
  • In some embodiments the culture medium comprises receptor activator of nuclear factor kappa-B ligand (RANKL). In other embodiments the culture medium does not comprise RANKL. In some embodiments, the culture medium of the invention is supplemented with nicotinamide. Addition of nicotinamide has been found to improve culture efficiency and lifespan of human organoids. Nicotinamide may be added to the culture medium to a final concentration of between 1 and 100 mM, between 5 and 50 mM, or preferably between 5 and 20 mM. For example, nicotinamide may be added to the culture medium to a final concentration of approximately 10 mM.
  • In some embodiments, the culture medium of the invention comprises a p53 stabilizing agent. A p53 stabilizing agent may be added to the culture medium in order to ensure that the cell population is predominantly tumour cells. Without wishing to be bound by any theory, it is believed that p53 stabilizing agents increase the cellular concentration of p53 (e.g. by blocking the interaction between p53 and Mdm2), stimulate p53-dependent expression and induce cellular senescence. p53 mutations commonly occur in tumour cells, which may result in the mutant p53 protein being unable to be stabilised by a p53 stabilizing agent. Thus, a subset of tumour cells with the mutant p53 protein would escape the senescence induced by a p53 stabilizing agent and outgrow normal cells in a mixed population. A preferred p53 stabilizing agent is a member of the Nutlin family, e.g. Nutlin-1, Nutlin-2 or Nutlin-3. For example, in some embodiments, the p53 stabilizing agent is Nutlin-3. Any suitable concentration of the p53 stabilizing agent may be used, for example between 1 nM and 10 mM, between 10 nM and 1 mM, between 100 nM and 100 μM, between 1 μM and 10 μM. For example, Nutlin-3 may be added to the culture medium of the invention to a final concentration of approximately 5 μM. Accordingly, in some embodiments of methods of the invention in which the breast epithelial stem cells are cancer cells, the culture medium used in the method advantageously comprises a p53 stabilizing agent. However, other embodiments of such methods for culturing breast epithelial cancer stem cells do not use a culture medium comprising a p53 stabilizing agent. It is also envisaged that the p53 stabilizing agent may be present in the culture medium when the method is used for culturing normal breast epithelial stem cells, although it is not required.
  • In some embodiments, the culture medium of the invention comprises a p38 MAP kinase inhibitor. A p38 MAP kinase inhibitor is an inhibitor that directly or indirectly negatively regulates p38 signalling. In some embodiments, the p38 MAP kinase inhibitor is SB202190. However, other suitable p38 MAP kinase inhibitors may alternatively be used and these are readily available to the skilled person.
  • Any suitable pH may be used. For example, the pH of the medium may be in the range from about 7.0 to 7.8, in the range from about 7.2 to 7.6, or about 7.4. The pH may be maintained using a buffer. A suitable buffer can readily be selected by the skilled person. Buffers that may be used include carbonate buffers (e.g. NaHCO3), and phosphates (e.g. NaH2PO4). These buffers are generally used at about 50 to about 500 mg/l. Other buffers such as N-[2-hydroxyethyl]-piperazine-N′-[2-ethanesul-phonic acid] (HEPES) and 3-[N-morpholino]-propanesulfonic acid (MOPS) may also be used, normally at around 1000 to around 10,000 mg/l. A culture medium may comprise a pH indicator, such as phenol red, to enable the pH status of the medium to be easily monitored (e.g. at about 5 to about 50 mg/litre).
  • A culture medium for use in the invention may comprise one or more amino acids. The skilled person understands the appropriate types and amounts of amino acids for use in stem cell culture media. Amino acids which may be present include L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine and combinations thereof. Some culture media will contain all of these amino acids. Generally, each amino acid when present is present at about 0.001 to about 1 g/L of medium (usually at about 0.01 to about 0.15 g/L), except for L-glutamine which is present at about 0.05 to about 1 g/L (usually about 0.1 to about 0.75 g/L). The amino acids may be of synthetic origin.
  • A culture medium for use in the invention may comprise one or more vitamins. The skilled person understands the appropriate types and amounts of vitamins for use in stem cell culture media. Vitamins which may be present include thiamine (vitamin B1), riboflavin (vitamin B2), niacin (vitamin B3), D-calcium pantothenate (vitamin B5), pyridoxal/pyridoxamine/pyridoxine (vitamin B6), folic acid (vitamin B9), cyanocobalamin (vitamin B12), ascorbic acid (vitamin C), calciferol (vitamin D2), DL-alpha tocopherol (vitamin E), biotin (vitamin H) and menadione (vitamin K).
  • A culture medium for use in the invention may comprise one or more inorganic salts. The skilled person understands the appropriate types and amounts of inorganic salts for use in stem cell culture media. Inorganic salts are typically included in culture media to aid maintenance of the osmotic balance of the cells and to help regulate membrane potential. Inorganic salts which may be present include salts of calcium, copper, iron, magnesium, potassium, sodium, zinc. The salts are normally used in the form of chlorides, phosphates, sulphates, nitrates and bicarbonates. Specific salts that may be used include CaCl2, CuSO4-5H2O, Fe(NO3)-9H2O, FeSO4-7H2O, MgCl, MgSO4, KCl, NaHCO3, NaCl, Na2HPO4, Na2HPO4—H2O and ZnSO4-7H2O.
  • The osmolarity of the medium may be in the range from about 200 to about 400 mOsm/kg, in the range from about 290 to about 350 mOsm/kg, or in the range from about 280 to about 310 mOsm/kg. The osmolarity of the medium may be less than about 300 mOsm/kg (e.g. about 280 mOsm/kg).
  • A culture medium for use in the invention may comprise a carbon energy source, in the form of one or more sugars. The skilled person understands the appropriate types and amounts of sugars to use in stem cell culture media. Sugars which may be present include glucose, galactose, maltose and fructose. The sugar is preferably glucose, particularly D-glucose (dextrose). A carbon energy source will normally be present at between about 1 and about 10 g/L.
  • A culture medium of the invention may contain serum. Serum obtained from any appropriate source may be used, including fetal bovine serum (FBS), goat serum or human serum.
  • Preferably, human serum is used. Serum may be used at between about 1% and about 30% by volume of the medium, according to conventional techniques.
  • In other embodiments, a culture medium of the invention may contain a serum replacement. Various different serum replacement formulations are commercially available and are known to the skilled person. Where a serum replacement is used, it may be used at between about 1% and about 30% by volume of the medium, according to conventional techniques.
  • In other embodiments, a culture medium of the invention may be serum-free and/or serum replacement-free. A serum-free medium is one that contains no animal serum of any type. Serum-free media may be preferred to avoid possible xeno-contamination of the stem cells. A serum replacement-free medium is one that has not been supplemented with any commercial serum replacement formulation.
  • In a preferred embodiment, the cell culture medium is supplemented with a purified, natural, semi-synthetic and/or synthetic growth factor and does not comprise an undefined component, such as fetal bovine serum or fetal calf serum. For example, supplements such as B27 (Invitrogen), N-Acetylcysteine (Sigma) and N2 (Invitrogen) stimulate proliferation of some cells. In some embodiments, the cell culture medium is supplemented with one or more of these supplements, for example one, any two or all three of these supplements.
  • A culture medium for use in the invention may comprise one or more trace elements, such as ions of barium, bromium, cobalt, iodine, manganese, chromium, copper, nickel, selenium, vanadium, titanium, germanium, molybdenum, silicon, iron, fluorine, silver, rubidium, tin, zirconium, cadmium, zinc and/or aluminium.
  • The medium may comprise a reducing agent, such as beta-mercaptoethanol at a concentration of about 0.1 mM.
  • A culture medium of the invention may comprise one or more additional agents, such as nutrients or growth factors previously reported to improve stem cell culture, such as cholesterol/transferrin/albumin/insulin/progesterone, putrescine, selenite/other factors.
  • In some embodiments, the culture medium of the invention comprises recombinant human neuregulin β-1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7 and FGF10.
  • In some embodiments, the culture medium of the invention comprises recombinant human neuregulin β-1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10 and PDGF-CC.
  • In some embodiments, the culture medium of the invention comprises recombinant human neuregulin β-1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10 and EGF.
  • In some embodiments, the culture medium of the invention comprises recombinant human neuregulin β-1, an Rspondin (e.g. Rspondin 1), Noggin, B27, N-Acetylcysteine, Nicotinamide, a ROCK 1,2 inhibitor (for example, Y-27632), an ALK 4, 5, 7 inhibitor (for example, A83-01), a p38 MAP Kinase inhibitor (for example, SB 202190), FGF7, FGF10, amphiregulin, TGF-alpha and EGF.
  • Extracellular Matrix
  • As described above, the method for culturing epithelial stem cells comprises culturing one or more epithelial stem cells in contact with an extracellular matrix. Any suitable extracellular matrix may be used. Isolated epithelial stem cells are preferably cultured in a microenvironment that mimics at least in part a cellular niche in which said stem cells naturally reside. This cellular niche may be mimicked by culturing said stem cells in the presence of biomaterials, such as an extracellular matrix that provides key regulatory signals controlling stem cell fate.
  • A cellular niche is in part determined by the stem cells and surrounding cells, and the extracellular matrix (ECM) that is produced by the cells in said niche. In a preferred method of the invention, epithelial stem cells are cultured in contact with an ECM. “In contact” means a physical or mechanical or chemical contact, which means that for separating said resulting organoid or population of epithelial stem cells from said extracellular matrix a force needs to be used. Preferably, the epithelial stem cells are embedded in the ECM.
  • A culture medium of the invention may be diffused into an extracellular matrix (ECM). In a preferred method of the invention, isolated tissue fragments or isolated epithelial stem cells are attached to an ECM. ECM is composed of a variety of polysaccharides, water, elastin, and glycoproteins, wherein the glycoproteins comprise collagen, entactin (nidogen), fibronectin, and laminin. ECM is secreted by epithelial cells, endothelial cells, parietal endoderm-like cells (e.g. Englebreth-Holm-Swarm Parietal Endoderm-Like cells described in Hayashi et al. (2004) Matrix Biology 23:47-62) and connective tissue cells. Different types of ECM are known, comprising different compositions including different types of glycoproteins and/or different combination of glycoproteins. Said ECM can be provided by culturing ECM-producing cells, such as for example epithelial cells, endothelial cells, parietal endoderm-like cells or fibroblast cells, in a receptacle, prior to the removal of these cells and the addition of isolated tissue fragments or isolated epithelial stem cells. Examples of extracellular matrix-producing cells are chondrocytes, producing mainly collagen and proteoglycans, fibroblast cells, producing mainly type IV collagen, laminin, interstitial procollagens, and fibronectin, and colonic myofibroblasts producing mainly collagens (type I, III, and V), chondroitin sulfate proteoglycan, hyaluronic acid, fibronectin, and tenascin-C. Alternatively, said ECM is commercially provided. Examples of commercially available extracellular matrices are extracellular matrix proteins (Invitrogen) and basement membrane preparations from Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells (e.g. Cultrex® Basement Membrane Extract (Trevigen, Inc.) or Matrigel™ (BD Biosciences)). A synthetic extracellular matrix material, such as ProNectin (Sigma Z378666) may be used. Mixtures of extracellular matrix materials may be used, if desired. The use of an ECM for culturing stem cells enhanced long-term survival of the stem cells and the continued presence of undifferentiated stem cells. In the absence of an ECM, stem cell cultures could not be cultured for longer periods and no continued presence of undifferentiated stem cells was observed. In addition, the presence of an ECM allowed culturing of three-dimensional tissue organoids, which could not be cultured in the absence of an ECM. The extracellular matrix material will normally be a drop on the bottom of the dish in which cells are suspended. Typically, when the matrix solidifies at 37° C., the medium is added and diffuses into the ECM. The cells in the medium stick to the ECM by interaction with its surface structure, for example interaction with integrins.
  • An example of an ECM for use in a method of the invention comprises at least one glycoprotein, such as laminin.
  • A preferred ECM for use in a method of the invention comprises at least two distinct glycoproteins, such as two different types of collagen or a collagen and laminin. The ECM can be a synthetic hydrogel extracellular matrix or a naturally occurring ECM. A further preferred ECM comprises laminin, entactin, and collagen IV. A further preferred ECM is provided by Matrigel™ (BD Biosciences), which comprises laminin, entactin, and collagen IV. In some embodiments the extracellular matrix is a laminin-containing extracellular matrix such as Matrigel™ (BD Biosciences). In some embodiments, the ECM comprises laminin, entactin, collagen IV and heparin sulphate proteoglycan (e.g. Cultrex® Basement Membrane Extract Type 2 (Trevigen, Inc.)).
  • In some embodiments, the single stem cell, population of cells, or tissue fragment is embedded in matrigel, which is optionally growth factor reduced and/or phenol red-free.
  • In some embodiments, the culture medium is placed on top of the ECM. The culture medium can then be removed and replenished as and when required. In some embodiments, the culture medium is replenished every 1, 2, 3, 4, 5, 6 or 7 days. If components are “added” or “removed” from the media, then this can in some embodiments mean that the media itself is removed from the ECM and then a new media containing the “added” component or with the “removed” component excluded is placed on the ECM.
  • In some embodiments the culture medium of the invention is in contact with an extracellular matrix or a 3D matrix that mimics the extracellular matrix by its interaction with the cellular membrane proteins, such as integrins.
  • There is further provided a culture medium of the invention and an extracellular matrix, e.g. supplied as a kit.
  • In some embodiments, the culture medium of the invention is suitable for culturing adult breast stem cells. In some embodiments, the culture medium of the invention is suitable for obtaining breast organoids. In some embodiments, the culture medium of the invention is suitable for expanding a population of breast stem cells for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or more than 14 passages.
  • Thus, the invention provides a culture medium as described herein that comprises one or more ErbB3/4 ligands that is suitable for expanding a population of breast stem cells for at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or more than 14 passages.
  • In some embodiments, the culture medium of the invention is suitable for establishing populations of successful breast organoids in at least 50%, at least 60%, at least 70%, at least 80% or at least 90% of inoculations. A successful breast organoid is defined herein as an organoid that is capable of being passaged at least four times.
  • Thus, the invention provides a culture medium that comprises one or more ErbB3/4 ligands that is suitable for establishing populations of successful organoids with an efficiency of at least 50%, at least 60%, at least 70%, at least 80% or at least 90%.
  • Uses of Culture Media of the Invention
  • The invention provides the use of the culture medium of the invention for expanding an epithelial stem cell, population of epithelial stem cells, tissue fragment or organoid.
  • The invention also provides the use of a culture medium of the invention for expanding a breast epithelial stem cell, population of breast epithelial stem cells, breast tissue fragment or breast organoid.
  • In some embodiments, the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a normal tissue. For example, in some embodiments, the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a non-cancerous tissue.
  • In some embodiments, the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a diseased tissue, for example a cancerous tissue, such as from an adenoma, a fibroadenoma, an adenocarcinoma, a carcinoma, a sarcoma, circulating tumour cells or metastasized cancer. Accordingly, in some embodiments, the stem cell, population of stem cells, tissue fragment or organoid is obtainable from a benign or malignant breast tumour. In some embodiments, the stem cell, population of stem cells, tissue fragment or organoid is or comprises a breast cancer cell(s). In some embodiments, the stem cell, population of stem cells, or the tissue fragment is a biopsy from a breast tumour. The tumour is preferably a breast tumour by origin rather than being a metastatic tumour originating from a different tissue.
  • Isolation of Breast Epithelial Stem Cells for Culture
  • In a preferred embodiment, the epithelial stem cells to be cultured in the method of the invention and/or from which the organoids are derived are obtained from adult tissue, i.e. the epithelial stem cells are adult epithelial stem cells. In this context “adult” means mature tissue, i.e. includes newly-born baby or child but excludes embryonic or foetal. In a preferred embodiment the epithelial stem cells are not derived from embryonic stem cells or embryonic stem cell lines, e.g. which have been differentiated in vitro, for example human embryonic stem cells or human embryonic stem cell lines.
  • The cells and tissues are preferably mammalian cells and tissues. More preferably, the cells and tissues are human cells and tissues. In some embodiments they are from other mammals, for example from a laboratory animal (e.g. mouse, rabbit, rat, guinea pig), a companion animal (e.g. dog, cat, horse) or a farm animal (e.g. cow, pig, sheep, goat).
  • Cells taken directly from live tissue, i.e. freshly isolated cells, are also referred to as primary cells. In some embodiments the epithelial stem cells are primary epithelial stem cells. Primary cells represent the best experimental models for in vivo situations. In a preferred embodiment of the invention, the epithelial stem cells are (or are derived from) primary epithelial stem cells. Primary cell cultures can be passaged to form secondary cell cultures. With the exception of cancer cells, traditional secondary cell cultures have a limited lifespan. After a certain number of population doublings (e.g. 50-100 generations) cells undergo the process of senescence and stop dividing. Cells from secondary cultures can become immortalized to become continuous cell lines. Immortalization can occur spontaneously, or may be virally- or chemically-induced.
  • Immortalized cell lines are also known as transformed cells. By contrast, the methods of the present invention allow continuous passaging of epithelial stem cells without immortalisation or transformation. Thus in some embodiments, the epithelial stem cells are not immortalised or transformed cells or are not derived from an immortalised cell line or a transformed cell line. An advantage of the present invention is that the epithelial stem cells, undergoing multiple rounds of expansion and passaging, retain the characteristics of primary cells and have minimal or no genotypic or phenotypic changes.
  • The epithelial stem cells may be obtained by any suitable method. In some embodiments, cells are isolated by collagenase digestion, for example, as described in the examples and in Dorell et al., 2008 (Hepatology. 2008 October; 48(4):1282-91. Surface markers for the murine oval cell response. Dorrell C, Erker L, Lanxon-Cookson K M, Abraham S L, Victoroff T, Ro S, Canaday P S, Streeter P R, Grompe M). In some embodiments, collagenase digestion is performed on a tissue biopsy. In some embodiments, collagenase and accutase digestion are used to obtain the epithelial stem cells for use in the invention.
  • In some embodiments, breast tissue may be minced, washed (for example in basal culture medium) and incubated in a culture medium of the invention supplemented with a protease (for example, collagenase, dispase or trypsin) (see, for example, Example 1). The protease may be at any suitable concentration, for example, between 0.1 and 10 mg/ml, between 0.1 and 1 mg/ml or between 1 and 10 mg/ml. For example, the protease may be present at between 1 and 2 mg/ml. Incubation in the culture medium supplemented with a protease may be for any suitable length of time. For example, the tissue may be incubated in the medium for between 10 minutes and 10 hours, between 20 minutes and 5 hours, between 30 minutes and 5 hours, between 30 minutes and 3 hours, between 30 minutes and 2 hours, between 1 hour and 5 hours, between 1 hour and 3 hours or between 1 hour and 2 hours. In some embodiments, the tissue is incubated in the medium for at least 10 minutes, at least 20 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 3 hours or at least 5 hours.
  • The tissue fragments contained in the resultant digested tissue suspension may then be further reduced in size. For example, the suspension may be sheared. Filtration and/or centrifugation steps may be used to isolate tissue fragments or isolated cells. The isolated tissue fragments or isolated cells may then be cultured in a culture medium of the invention.
  • In some embodiments, the method comprises culturing a fragment of tissue which comprises epithelium. In some embodiments, the epithelial stem cells are isolated from a tissue fragment.
  • In some embodiments, the epithelial stem cells are obtained for culturing based on the expression of epithelial cell surface markers, such as EPCAM, MUC-1, KRT-5, KRT8 and KRT18. A method of the invention therefore comprises preparing a cell suspension from said epithelial tissue as described above, contacting said cell suspension with an epithelial cell surface marker binding compound (e.g. EPCAM, MUC-1, KRT-5, KRT8 and/or KRT18 binding compounds), isolating the epithelial marker binding compound, and isolating the cells from said binding compound.
  • In some embodiments, epithelial stem cells are obtained for culturing based on the expression of Lgr5 and/or Lgr6 on the epithelial stem cell surface; these proteins belong to the large G protein-coupled receptor (GPCR) superfamily (see, for example, WO 2009/022907, the contents of which are incorporated herein in their entirety). The Lgr subfamily is unique in carrying a large leucine-rich ectodomain important for ligand binding. In some embodiments, a method of the invention therefore comprises preparing a cell suspension from said epithelial tissue as described above, contacting said cell suspension with an Lgr5 and/or 6 binding compound (such as an antibody, e.g. an anti-Lgr5 monoclonal antibody, e.g. as described in WO 2009/022907), isolating the Lgr5 and/or 6 binding compound, and isolating the stem cells from said binding compound.
  • An organoid is preferably obtained using a cell from an adult tissue, preferably an epithelial stem cell from an adult tissue.
  • In some embodiments the epithelial stem cells are normal cells. In alternative embodiments, the epithelial stem cells are cancer stem cells. Thus, it is envisaged, for example, that the stem cells may be Lgr5 positive cancer stem cells. Accordingly, the cells may be obtained from a tumour, if required. In alternative embodiments, the epithelial stem cells are diseased stem cells, for example stem cells infected with intracellular pathogens (e.g. bacteria, viruses or parasites).
  • Binding compounds that bind to breast cancer cell markers may be used for the isolation of breast cancer stem cells for culturing in a method of the invention. Examples of breast cancer cell markers include Epithelial Membrane Antigen, Cancer antigen 15-3 (CA 15-3), cancer antigen 27.29 (CA 27.29), carcinoembryonic antigen (CEA), Urokinase plasminogen activator (uPA), plasminogen activator inhibitor (PAI-1), Podocalyxin, EZH2 and kallikrein-10. For example, Epithelial Membrane Antigen (EMA) binding compounds (e.g. anti-EMA antibodies) may be used for the isolation of breast cancer cells. In some embodiments, a method of the invention therefore comprises preparing a cell suspension from primary or secondary cancerous tissue, contacting said cell suspension with a breast cancer cell marker binding compound (e.g. EMA binding compounds), isolating the breast cancer cell marker binding compound and isolating the cells from said binding compound. In another embodiment, an organoid originates from a single cell, optionally expressing Lgr5. In some embodiments the single cell comprises a nucleic acid construct comprising a nucleic acid molecule of interest.
  • Preferred Lgr5 and/or 6 binding compounds comprise antibodies, such as monoclonal antibodies that specifically recognize and bind to the extracellular domain of either Lgr5 or Lgr6, such as monoclonal antibodies including mouse and rat monoclonal antibodies (see, for example, WO 2010/016766, the contents of which are incorporated herein in their entirety). Using such an antibody, Lgr5 and/or Lgr6-expressing stem cells can be isolated, for example with the aid of magnetic beads or through fluorescence-activated cell sorting, as is clear to a skilled person. Using a method of the invention, it is possible to isolate one single Lgr5 and/or Lgr6 expressing cell and to apply a method of the invention to it. An organoid or a population of breast epithelial stem cells may therefore be derived from one single cell. Accordingly, in some embodiments, the starting cell to be cultured is a single cell.
  • Alternatively, a population of cells may be used as the starting point, for example, a population of cells contained in a breast tissue fragment as described above. Thus, the methods of the invention are not restricted to using single cells as the starting point.
  • In a further aspect, there is provided a method for obtaining an organoid comprising culturing breast epithelial stem cells in a culture medium of the invention. Preferably, the method comprises culturing the breast epithelial stem cells in a culture medium of the invention using a culture method as described herein.
  • In some embodiments, the method comprises culturing the epithelial stem cells or obtaining the organoid/population of adult epithelial stem cells from a single cell. Advantageously, this allows a homogenous population of cells to form. In some embodiments, the method comprises culturing the stem cells in a culture medium of the invention for a period of time, for example, 3 days to 10 weeks, 1 to 10 weeks, 1 to 4 weeks or 10 days to 3 weeks, and then passaging the cells (e.g. dissociating the cells to a single cell density, seeding one or more cells at a ratio of 1 cell per container (e.g. per well)), expanding the cells using a culture medium of the invention for a period of time, for example, 3 days to 10 weeks, 1 to 10 weeks, 1 to 4 weeks or 10 days to 3 weeks and repeating the passaging and expanding steps at least once, at least twice, at least three times, at least four times, at least five times, at least six times, at least seven times, at least eight times, at least nine times, at least ten times, at least eleven times, at least twelve times, at least thirteen times or at least fourteen times.
  • Following culturing, the method may further comprise obtaining and/or isolating one or more epithelial stem cells or an organoid. For example, following culture of the stem cells, it may be useful to remove one or more stem cells and/or one or more organoids cultured in the culture medium from the culture medium for use in subsequent applications. Any one of a number of physical methods of separation known in the art may be used to select the cells of the invention and distinguish these from other cell types. Such physical methods may involve FACS and various immuno-affinity methods based upon makers specifically expressed by the cells of the invention. As described herein, LGR5 is a cell marker that may be expressed in the cells of the invention. Therefore, by way of illustration only, the cells of the invention may be isolated by a number of physical methods of separation, which rely on the presence of this marker. Similarly, any of the other markers expressed by the cells may be used.
  • In one embodiment, the cells of the invention may be isolated by FACS utilizing an antibody, for example, against one of these markers. As will be apparent to one skilled in the art, this may be achieved through a fluorescent labeled antibody, or through a fluorescent labeled secondary antibody with binding specificity for the primary antibody. Examples of suitable fluorescent labels includes, but is not limited to, FITC, Alexa Fluor® 488, GFP, CFSE, CFDA-SE, DyLight 488, PE, PerCP, PE-Alexa Fluor® 700, PE-Cy5 (TRI-COLOR®), PE-Cy5.5, PI, PE-Alexa Fluor® 750, and PE-Cy7. This list is provided by way of example only, and is not intended to be limiting.
  • It will be apparent to a person skilled in the art that FACS analysis using an anti-Lgr5 antibody may provide a purified cell population. However, in some embodiments, it may be preferable to purify the cell population further by performing a further round of FACS analysis using one or more of the other identifiable markers, for example, Sox9, Slug, CD44 and/or ALDH1, but others may also be used.
  • In another embodiment, the cells of the invention may be isolated by immuno-affinity purification, which is a separation method well known in the art. By way of illustration only, the cells of the invention may be isolated by immuno-affinity purification directed towards c-kit. As will be apparent to one skilled in the art, this method relies upon the immobilisation of antibodies on a purification column. The cell sample is then loaded onto the column, allowing the appropriate cells to be bound by the antibodies, and therefore bound to the column. Following a washing step, the cells are eluted from the column using a competitor which binds preferentially to the immobilised anti-c-kit antibody, and permits the cells to be released from the column.
  • It will be apparent to a person skilled in the art that immuno-affinity purification using an immobilised antibody will provide a purified cell population. However, in some embodiments, it may be preferable to purify the cell population further by performing a further round of immuno-affinity purification using one or more of the other identifiable markers, for example Sox9, Slug, CD44 and/or ALDH1, and use an aliquot of the isolated clones to ascertain the expression of other relevant intracellular markers.
  • It will be apparent to a person skilled in the art that the sequential purification steps are not necessarily required to involve the same physical method of separation. Therefore, it will be clear that, for example, the cells may be purified through a FACS step using an anti-Lgr5 antibody, followed by an immuno-affinity purification step using a SSEA-1 affinity column. In certain embodiments, the cells may be cultured after isolation for at least about 15, at least about 20 days, at least about 25 days, or at least about 30 days. In certain aspects, the cells are expanded in culture longer to improve the homogeneity of the cell phenotype in the cell population.
  • Other features of this method are defined in the part of the description dedicated to definitions. Single-cell suspensions or small clusters of cells (2-50 cells/cluster) will normally be seeded, rather than large clusters of cells, as in known in the art. As they divide, such cells will be seeded onto a support at a density that promotes cell proliferation. Typically, when single cells are isolated the plating density of at least 1-500 cells/well is used, the surface of the well is 0.32 cm2. When clusters are seeded the plating density is preferably 250-2500 cells/cm2. For replating, a density of between about 2500 cells/cm2 and about 5,000 cells/cm2 may be used in some embodiments. During replating, single-cell suspensions or small cluster of cells will normally be seeded, rather than large clusters of cells, as in known in the art.
  • In one embodiment, the invention provides a population of cells or one or more organoids comprising said stem cells that have been generated or obtained by culturing stem cells or tissue fragments according to the invention, which have been cultured for at least 2 months, at least 3 months, preferably at least 4 months, at least 5 months, at least 6 months, at least 7 months, at least 9 months, or at least 12 months or more, wherein the stem cells have been passaged approximately every 1 to 4 weeks.
  • A ‘population’ of cells is any number of cells greater than 1, but is preferably at least 1×103 cells, at least 1×104 cells, at least 1×105 cells, at least 1×106 cells, at least 1×107 cells, at least 1×108 cells, or at least 1×109 cells.
  • The stem cells or progenitor cells of the invention cultured according to the invention may be human stem cells or human progenitor cells. The stem cells of the invention cultured according to the invention may be epithelial stem cells or epithelial progenitor cells.
  • In some embodiments, the stem cells of the invention and/or cultured according to the invention are not embryonic stem cells. In some embodiments the stem cells of the invention and/or cultured according to the invention are not human embryonic stem cells. Preferably, the stem cells of the invention are adult stem cells.
  • In a preferred embodiment, the stem cells are human breast epithelial stem cells. Human epithelial stem cells include stem cells of human epithelial tissue origin. Epithelial stem cells are able to form the distinct cell types of which the epithelium is composed.
  • In a preferred embodiment, the organoids are human breast organoids.
  • Methods and Culture Media for Cancer Cells and Cancer Organoids
  • In some embodiments, the epithelial stem cell is a cancer cell or a non-cancerous tumour cell e.g. is derived from a tumour. In some embodiments, the cancer cell is a circulating tumour cell. In some embodiments, the cancer cell is not a circulating tumour cell. Where the term “cancer” is used herein, it is to be understood that it applies equally to non-cancerous (benign) tumours. Accordingly, in some embodiments the cell is derived from a malignant tumour or from a metastasis. In some embodiments, the cell is derived from a benign tumour. Cancer cells tend to have mutations that constitutively activate or deactivate certain growth pathways and which mean that certain factors in the culture medium that may normally be required for growth, are no longer necessary. For example, some cancers result in constitutive activation of the Wnt pathway. In such cases, a culture medium may not require an Lgr5 agonist and/or may not require a Wnt agonist, but one or both of these may still be present in the culture medium. Other mutations would allow other factors to be left out of the culture media described herein. Other epithelial cancers (carcinomas) or non-cancerous tumours (e.g. adenomas) can also be grown in culture media of the invention.
  • In a preferred embodiment, a cancer organoid obtained from cancer stem cells is grown in a culture medium of the invention that is suitable for growth of the corresponding normal tissue organoid obtained from normal stem cells, optionally with certain factors excluded from the medium. For example, a breast cancer organoid obtained by culturing breast cancer stem cells may be grown in the same culture conditions as a normal breast organoid obtained by culturing breast epithelial stem cells, optionally with certain factors excluded from the medium. In many situations it may be preferable (or at least more convenient) to grow cancer organoids in the normal tissue medium (without any factors excluded). Preferably, the normal tissue medium should allow cancers with all genetic backgrounds to grow, without excluding any particular cancer mutations. The methods of the invention for culturing stem cells can be practised accordingly.
  • In a further preferred embodiment, the cancer organoid is grown in a culture medium of the invention that further comprises a p53 stabilizing agent. A p53 stabilizing agent may be added to the culture medium in order to ensure that the cell population is predominantly tumour cells. The methods of the invention for culturing stem cells can be practised accordingly.
  • Methods
  • The invention provides a method for culturing a single breast epithelial stem cell, a population of breast epithelial stem cells, or an isolated breast tissue fragment, preferably to generate an organoid, wherein the method comprises:
      • providing a breast epithelial stem cell, a population of breast epithelial stem cells or an isolated breast tissue fragment;
      • providing a culture medium comprising one or more ErbB3/4 ligands and preferably further comprising one or more FGFR2b ligands and/or one or more Wnt agonists;
      • contacting the stem cell, population of stem cells, or isolated tissue fragment with the culture medium;
      • culturing the stem cell, population of stem cells, or isolated tissue fragment under appropriate conditions.
  • The invention also provides a method for expanding breast epithelial stem cells, wherein the method comprises:
      • providing a breast epithelial stem cell, population of breast epithelial stem cells or an isolated breast tissue fragment;
      • providing a culture medium comprising one or more ErbB3/4 ligands and (i) one or more FGFR2b ligands and/or (ii) one or more Wnt agonists (e.g. one or more Lgr5 agonists);
      • contacting the stem cell, population of stem cells, or isolated tissue fragment with the culture medium; and
      • culturing the cells, population of stem cells, or isolated tissue fragment under appropriate conditions.
  • Preferably, the cells are expanded to generate one or more (e.g. at least 2, 3, 4, 5, 6, 10, 15, 20 or more than 20) organoids.
  • In some embodiments, the culture medium used in the methods of the invention is a culture medium according to the invention.
  • A method for ‘expanding’ a population of cells or isolated tissue fragments is one that involves maintaining or increasing the number of stem cells in an initial population to generate an expanded population of stem cells which retain their undifferentiated phenotype and self-renewing properties.
  • However, it may also include the production of differentiating progeny, which may, for example, form tissue-like structures contributing to organoid formation. Hence, there are herein provided methods for obtaining a breast organoid comprising culturing stem cells or tissue fragments comprising said stem cells in a culture medium of the invention. The invention provides a method for expanding a single breast stem cell or a population of breast stem cells, preferably to generate an organoid, wherein the method comprises culturing the single stem cell or population of stem cells in a culture medium according to the invention. As mentioned above, the organoid may be a normal organoid or it may be a cancer organoid, for example, a carcinoma organoid or an adenocarcinoma organoid, or a benign tumour organoid, for example. In some embodiments, the method for expanding a single stem cell or a population of stem cells, preferably to generate an organoid, comprises expanding the single stem cell, population of stem cells or tissue fragment in a culture medium according to the invention.
  • Thus the invention provides a method for expanding a single breast epithelial stem cell or a population of breast epithelial stem cells, preferably to generate an organoid, wherein the method comprises:
      • providing a breast epithelial stem cell, a population of breast epithelial stem cells or an isolated breast tissue fragment;
      • providing a culture medium according to the invention;
      • contacting the stem cells with the culture medium;
      • culturing the cells under appropriate conditions.
  • In some embodiments, the method comprises bringing the stem cell, the population of stem cells or the isolated tissue fragment and the culture medium into contact with an extracellular matrix or a 3D matrix as described herein, for example, a 3D matrix that mimics the extracellular matrix by its interaction with the cellular membrane proteins such as integrins, for example a laminin-containing extracellular matrix such as Matrigel™ (BD Biosciences) or Cultrex® Basement Membrane Extract (Trevigen, Inc.). In some embodiments, the culture medium is diffused into the extracellular matrix.
  • The invention provides methods for culturing breast epithelial stem cells. The invention further provides methods for expanding breast epithelial stem cells and methods for obtaining breast organoids.
  • The term “expanding stem cells” is synonymous with “obtaining breast organoids”.
  • Isolation and Culture of Tissue Fragments and Stem Cells
  • The skilled person would be able to determine the appropriate culture conditions for the methods of the invention.
  • In some embodiments, the method comprises bringing the stem cell, the population of stem cells or the isolated tissue fragment and the culture medium into contact with an extracellular matrix or a 3D matrix as described herein, for example, a 3D matrix that mimics the extracellular matrix by its interaction with the cellular membrane proteins such as integrins, for example a laminin-containing extracellular matrix such as Cultrex® Basement Membrane Extract (Trevigen, Inc.) or Matrigel™ (BD Biosciences). In some embodiments, the culture medium is diffused into the extracellular matrix.
  • In some embodiments, the tissue fragments or isolated cells are cultured in suspension plates. In some embodiments, the tissue fragments or isolated cells are cultured at between 35° C. and 39° C. or between 36° C. and 38° C. For example, the tissue fragments or isolated cells may be cultured at approximately 37° C.
  • In some embodiments, the tissue fragments or isolated cells are cultured at ambient levels of Oz. Alternatively, in some embodiments the level of Oz may be, for example, between 0.5% and 4%, between 1% and 4%, between 1% and 3% or between 2% and 4%. For example, the tissue fragments or isolated cells may be cultured under conditions of approximately 2% Oz. The tissue fragments or isolated cells may be cultured under conditions of between 3% and 7% CO2 or between 4% and 6% CO2. For example, the tissue fragments or isolated cells may be cultured under conditions of approximately 5% CO2.
  • The culture medium may be changed at any suitable interval, for example, every 1-7 days, every 2-6 days or every 3-5 days. For example, the culture medium may be changed every 4 days.
  • Organoids may be passaged at any suitable interval, for example, every 3 days to every 10 weeks, every 1 to 10 weeks, every 1 to 4 weeks or every 10 days to 3 weeks. In some embodiments, the organoids are passaged approximately every 10 days to 3 weeks.
  • In some embodiments, the timing for passaging of organoids is determined by the organoids reaching a particular size (e.g. an average size of approximately 100-200 μm) and/or a reduction in viability which can manifest in the loss of integrity of the epithelium, the organoids having a dense appearance and/or the presence of dead cells.
  • Organoids
  • The invention provides an organoid or a population of epithelial stem cells obtainable or obtained by a method of the invention. Thus, in some embodiments, the method further comprises obtaining and/or isolating an organoid. The organoid or population of epithelial stem cells of the invention comprises breast cells. Accordingly, there is provided a breast organoid which comprises a population breast epithelial stem cells. Preferably, the organoid or population of epithelial stem cells of the invention consists of breast cells.
  • Methods of expanding breast cells are currently described in Pasic et al. (2011) Genes & Development 25:1641-1653, but the culture methods and resulting cellular structures are different from the culture methods and resulting cellular structures provided by the present invention. The methods described in Pasic et al. use a culture system which comprises (i) amphiregulin or EGF (which are HER1 ligands) and (ii) FGF2 or FGF7. They do not use culture media comprising an ErbB3/4 ligand in combination with an Lgr5 agonist or a FGFR2b ligand, as is used in the present invention. Pasic et al. tested a culture medium comprising an ErbB3/4 ligand, NRG1-β1, but did not find that the breast organoids are able to grow in this culture medium. Pasic et al. did not test a culture medium comprising an ErbB3/4 ligand combined with an Lgr5 agonist or a FGFR2b ligand. Use of the Pasic et al. culture system results in the formation of cellular structures that have a morphology that is distinct from that of the organoids of the present invention. In particular, the Pasic et al. cellular structures have a budding morphology in which the structures have a rough edge, whereas the organoids obtained using the methods of the present invention do not have this morphology but instead have a smooth cyst-like structure or a morphology that is similar to a cluster of grapes. Cultures using the Pasic et al. system have been observed to stop expanding after 2-3 passages (see Example 4 herein), whereas the inventors have found that in some embodiments using a method of the invention it is possible to expand the breast epithelial stem cells for more than 14 passages.
  • Methods of culturing circulating tumour cells are described in Yu et al. (2014) Science 345(6193):216-220. However, the authors of this paper provided no guidance with respect to culturing breast epithelial stem cells obtained from normal breast tissue, primary tumour or metastases. Again, the culture system is different from the present invention and results in different structures compared to the organoids of the present invention. In particular, Yu uses a culture method which comprises EGF and FGF2. They do not use culture media comprising an ErbB3/4 ligand in combination with an Lgr5 agonist or a FGFR2b ligand, as is used in the present invention. Indeed, they do not use any culture media comprising an ErbB3/4 ligand. Yu's method results in formation of aggregates of single tumour cells without strong intercellular adhesion. No continuous epithelium is formed using this method. In contrast, the organoids obtained using the methods of the present invention display intercellular adhesion and a continuous epithelium. In some embodiments, the intercellular adhesion is strong.
  • Accordingly, the invention provides a breast organoid which has a cyst-like structure. In some embodiments, the cyst-like structure comprises a central lumen. In some embodiments, the central lumen of the cyst-like structure does not comprise any cells. In other embodiments, the central lumen of the cyst-like structure comprises breast epithelial stem cells, optionally wherein the central lumen is filled with breast epithelial stem cells. The invention further provides a breast organoid that has a morphology that is similar to a cluster of grapes. The invention further provides a breast epithelial stem cell, a population of breast epithelial stem cells or a breast organoid that has been cultured or can be cultured for more than 3 passages. The invention further provides a culture medium comprising an ErbB3/4 ligand combined with an Lgr5 agonist and/or a FGFR2b ligand.
  • Organoids of the invention, obtainable by expansion of stem cells, provide a population of cells which resemble their in vivo counterparts.
  • In some embodiments, an organoid comprises at least one epithelial stem cell. The term “stem cell” preferably refers to a cell which can divide and produce further epithelial stem cells or can generate differentiated progeny, e.g. progenitor cells, which are able to divide and further differentiate into one or more cell types. However, in some embodiments, the term “stem cell” and “progenitor cell” may be used interchangeably. In some instances, a progenitor cell is able to dedifferentiate into a stem cell and so culturing a progenitor cell in the culture medium of the invention may result in the progenitor cell dedifferentiating into a stem cell. For example, it is to be understood that in a preferred organoid, the majority of cells are expanding cells (i.e. dividing cells) that retain an undifferentiated phenotype. Although some spontaneous differentiation may occur, the cell population is generally an expanding population. The epithelial stem cells used in the invention are preferably multipotent, oligopotent or unipotent organ-specific adult stem cells. The stem cells are not totipotent stem cells. In some embodiments, the stem cells are not pluripotent stem cells. The organoids also have a distinctive structure that arises from these cellular properties, as described in detail below.
  • Image analysis may be used to assess characteristics of cells in culture such as cell morphology; cell structures; evidence for apoptosis or cell lysis; and organoid composition and structure. Many types of imaging analysis are well known in the art, such as electron microscopy, confocal microscopy, stereomicroscopy, fluorescence microscopy. Histological analysis can reveal basic architecture and cell types.
  • An organoid of the invention preferably has a three dimensional structure, i.e. the organoid is preferably a three-dimensional organoid. In a preferred embodiment the organoid comprises only epithelial cells, i.e., non-epithelial cells are absent from the organoid. This is because the culture medium of the invention is specifically designed to expand epithelial stem cells, for example Lgr5+ epithelial stem cells. Therefore, even if other cell types are transiently present in the culture medium, e.g. in the tissue fragment that is the starting material of the invention, these cells are unlikely to survive and instead will be replaced by the longer term expansion of the stem cells which generate a pure population of epithelial cells.
  • In some embodiments, the epithelial cells surround a lumen. In some embodiments, the epithelial cells surrounding the lumen are polarized, (meaning that proteins are differentially expressed on the apical or basolateral side of the epithelial cell). In some embodiments the organoids comprise stem cells which are able to actively divide and which are preferably able to differentiate to all major differentiated cell lineages present in the corresponding in vivo tissue.
  • In some embodiments the organoids of the invention have a section which is formed of multiple layers. In some embodiments the organoids of the invention have a monolayer section which appears to be formed of multiple layers referred to herein as regions of “pseudo-stratified” cells. In some embodiments the monolayer of pseudo-stratified cells folds so that it encloses lumina between the folds.
  • In some embodiments the organoids of the invention comprise single monolayers that are folded (or invaginated) to form two or more layers. It can sometimes be difficult to distinguish between folded (or invaginated) monolayers and regions of stratified cells. In some embodiments an organoid comprises both regions of stratified cells and regions of folded monolayers. In some embodiments the organoids of the invention have a section which is formed of multiple layers and a section comprising a single monolayer of cells. In some embodiments the organoids of the invention comprise or consist of a single monolayer of cells.
  • In some embodiments the organoids of the invention comprise or consist of epithelial cells. In some embodiments, the organoids comprise or consist of a single layer of epithelial cells. In some embodiments non-epithelial cells are absent from the organoids.
  • Illustrative examples of organoids generated according to the invention are given in the accompanying figures. In one embodiment, an organoid of the invention has a structure essentially as presented in FIG. 2 (for example, in some embodiments, a normal organoid has a structure essentially as presented for the W894N or R1100N organoid lines in FIG. 2B or a cancer organoid has a structure essentially as presented for the W1007T, W1012T or W859T organoid lines in FIG. 2A) or in FIG. 6. In one embodiment, an organoid of the invention exhibits cell staining essentially as presented in FIG. 5 or FIG. 6.
  • In some embodiments, the organoids are mainly cystic structures with few budding structures. In some embodiments, the organoids generated according to the invention do not have budding structures. The cystic structures comprise mainly monolayers but some regions of stratified cells may be present.
  • By “cystic” it is meant that the organoid is approximately spherical. By “budding” it is meant that the organoid has multiple regions growing out of the basic structure.
  • Under the improved culture conditions of the invention, breast organoids displayed cystic organoid structures, rather than the budding structures seen under previous culture conditions.
  • The invention also provides an organoid, preferably obtainable by the methods of the invention, which is a three-dimensional organoid comprising epithelial cells surrounding a central lumen.
  • In some embodiments, said organoid comprises a single layer of cells. In other embodiments, said organoid comprises a multi-layered epithelium.
  • In some embodiments, the organoid is obtained from normal breast tissue and has a cystic structure. In some embodiments, said normal organoid comprises a monolayer of basal and luminal cells surrounding a central lumen.
  • In some embodiments, the organoid is obtained from cancerous breast tissue and has a cystic structure. In some embodiments, said organoid comprises: (i) a central lumen, multiple small lumina or no lumen and/or (ii) a monolayer of epithelial cells or a multi-layered epithelium. Thus, in some embodiments, the breast cancer organoid of the invention comprises a monolayer of epithelial cells surrounding a lumen. In other embodiments, the breast cancer organoid comprises a multi-layered epithelium and multiple small lumens. In further embodiments, the breast cancer organoid comprises a multi-layered epithelium and no lumen. Cancer breast organoids optionally comprise solid balls of tumour cells.
  • Tumour status of cancer cells or cancer organoids may be confirmed using any suitable method. For example, karyotyping of cells or organoids can be performed to assess whether aneuploidy is present, which would suggest that the cells or organoids are tumour cells or tumour organoids.
  • Tumour cells or cancer organoids may also be identified by adding a p53 stabilizing agent to the culture medium. Tumour cells and cancer organoids commonly contain p53 mutations that result in p53 stabilizing agents having no effect on the degradation of p53. Thus, tumour cells, unlike normal cells, are often able to escape senescence triggered by the addition of a p53 stabilizing agent (e.g. Nutlin-3) to a culture medium.
  • In some embodiments, the breast cancer organoids stain positively for luminal cell markers, e.g. keratin-8 (Krt-8), and stain negatively for basal cell markers, e.g., keratin-14 (Krt-14) or keratin-5 (Krt-5). Thus, in some embodiments, the breast cancer organoid comprises luminal cells but does not comprise basal cells. In some embodiments, breast cancer organoids express KRT-8 at levels that are comparable to those seen in normal breast tissue or non-cancerous breast organoids. In some embodiments, breast cancer organoids express KRT-5 and/or KRT-14 at levels that are reduced compared to those seen in normal breast tissue or non-cancerous breast organoids.
  • Breast cancer organoids of the invention may express levels of CDH1 that are reduced compared to those seen in normal breast tissue or non-cancerous breast organoids. Markers of epithelial to mesenchymal transition (e.g., VIM, ZEB1) may be more highly expressed in breast cancer organoids compared to normal breast tissue or non-cancerous breast organoids. Breast cancer organoids may display a reduced level of E-cadherin expression compared to normal breast tissue or non-cancerous breast organoids.
  • Breast cancer organoids of the invention may express other breast cancer cell markers such as Epithelial Membrane Antigen, Cancer antigen 15-3 (CA 15-3), cancer antigen 27.29 (CA 27.29), carcinoembryonic antigen (CEA), Urokinase plasminogen activator (uPA), plasminogen activator inhibitor (PAI-1), Podocalyxin, EZH2 and/or kallikrein-10.
  • There is provided an organoid or a population of epithelial stem cells which has been cultured in culture media of the invention for at least 2 months, for example at least 10 weeks, at least 12 weeks, at least 14 weeks, at least 16 weeks, at least 4 months, at least 5 months, at least 6 months, at least 9 months, at least one year. Preferably, the cells are human cells. Preferably, the cells have been passaged approximately every 1 to 4 weeks.
  • There is also provided an organoid or a population of epithelial stem cells which has been that has been passaged or which is capable of being passaged for more than 3 passages, more than 4 passages, more than 5 passages, more than 6 passages, more than 7 passages, more than 8 passages, more than 9 passages, more than 10 passages, more than 11 passages, more than 12 passages, more than 13 passages or more than 14 passages.
  • Advantageously, use of a ErbB3/4 ligand allows the breast cells and organoids to be cultured long-term. It is not possible to culture and expand human breast epithelial stem cells or breast organoids for two months or more in the culture medium described in Pasic et al., WO2012/014076 or WO2012/168930, wherein the cells or organoids are passaged at least three times. Therefore, such breast organoids or populations of breast epithelial stem cells that had been cultured long-term did not exist before the present invention. Accordingly, there is provided an organoid or a population of stem cells that has been cultured/passaged or that is capable of being cultured/passaged as described herein.
  • Within the context of the invention, a tissue fragment is a part of an adult tissue, preferably a human adult tissue, such as part of a human breast. The tissue may be normal (healthy) tissue or it may be diseased or infected tissue. Preferably an organoid as identified herein is therefore not a tissue fragment. An organoid is preferably obtained using a cell from an adult tissue, preferably an epithelial stem cell from an adult tissue, optionally from an adult tissue fragment. In some embodiments, an organoid is obtained using an epithelial stem cell from an adult tissue or adult tissue fragment expressing Lgr5. Therefore, within the context of this invention, in some embodiments a tissue fragment comprises Lgr5+ stem cells.
  • In an embodiment, an organoid is an organoid which is still being cultured using a method of the invention (preferably using a culture medium of the invention) and is therefore in contact with an extracellular matrix. Preferably, an organoid is embedded in a non-mesenchymal extracellular matrix. Within the context of the invention, “in contact” means a physical or mechanical or chemical contact, which means that for separating said organoid from said extracellular matrix a force needs to be used. In some embodiments, the extracellular matrix is a gelatinous protein mixture secreted by Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells, such as Matrigel (BD Biosciences) or Cultrex® Basement Membrane Extract (Trevigen, Inc.). Accordingly, the culture medium of the invention may optionally further comprise a population of cells of invention and/or one or more organoids of the invention.
  • The invention provides a composition comprising an organoid or cell of the invention, an extracellular matrix and a culture medium, preferably a culture medium of the invention.
  • Uses of Organoids
  • There is an interest in culture media and methods for culturing stem cells for the formation, maintenance and expansion of organoids, such as breast organoids. An organoid comprises stem cells, such as epithelial stem cells, which retain their undifferentiated phenotype and self-renewal properties but also have differentiating progeny that grow into tissue-like structures.
  • Similarly to populations of related or identical cells, breast organoids, which more closely mimic the basic physiology of breast tissue, may be used in toxicity assays, or assays for drugs. They may also be useful for culturing pathogens which currently lack suitable tissue culture or animal models. Furthermore, such organoids may be useful in regenerative medicine, for example in post-radiation and/or post-surgery repair of the breast.
  • It is clear that there are many clinical and research applications for stem cells and their differentiated progeny. For all these applications, reproducible culture methods are of the utmost importance for providing adequate numbers of cells of suitable quality. For example, for effective drug screening, conditions must be carefully controlled requiring precise culture methods for controlling differentiation and proliferation of cells, so that pure populations of phenotypically and karyotypically identical cells can be generated. Similarly, for cell-based therapies, wherein cultured cells may be directly provided to patients, the cells must be genetically and phenotypically sound so as to avoid undesirable immune responses or cell fates when provided to the patient.
  • The invention provides the use of an organoid or expanded population of cells of the invention for use in medicine. Accordingly, the invention provides the use of an organoid or expanded population of cells of the invention for use in treating a disorder, condition or disease. The invention provides the use of an organoid or expanded population of cells of the invention for use in drug screening, (drug) target validation, (drug) target discovery, toxicology and toxicology screens, personalized medicine, regenerative medicine and/or as ex vivo cell/organ models, such as disease models.
  • Cells and organoids cultured according to the media and methods of the invention are thought to faithfully represent the in vivo situation. This is true both for expanded populations of cells and organoids grown from normal tissue and for expanded populations of cells and organoids grown from diseased tissue. Therefore, as well as providing normal ex vivo cell/organ models, the organoids or expanded population of cells of the invention can be used as ex vivo disease models.
  • Organoids of the invention can also be used for culturing of a pathogen and thus can be used as ex vivo infection models. Examples of pathogens that may be cultured using an organoid of the invention include viruses, bacteria, prions or fungi that cause disease in its animal host. Thus, an organoid of the invention can be used as a disease model that represents an infected state. In some embodiments of the invention, the organoids can be used in vaccine development and/or production.
  • Diseases that can be studied by the organoids of the invention thus include genetic diseases, metabolic diseases, pathogenic diseases, inflammatory diseases etc, for example including, but not limited to: breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g. adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • Traditionally, cell lines and more recently iPS cells have been used as ex vivo cell/organ and/or disease models (for example, see Robinton et al. Nature 481, 295, 2012). However, these methods suffer a number of challenges and disadvantages. For example, cell lines cannot be obtained from all patients (only certain biopsies result in successful cell lines) and therefore, cell lines cannot be used in personalised diagnostics and medicine. iPS cells usually require some level of genetic manipulation to reprogramme the cells into specific cell fates. Alternatively, they are subject to culture conditions that affect karotypic integrity and so the time in culture must be kept to a minimum (this is also the case for human embryonic stem cells). This means that iPS cells cannot accurately represent the in vivo situation but instead are an attempt to mimic the behaviour of in vivo cells. Cell lines and iPS cells also suffer from genetic instability.
  • By contrast, the organoids of the invention provide a genetically stable platform which faithfully represents the in vivo situation. The organoids of the invention can also be expanded continuously, providing a good source of genetically stable cells. In particular, an expanding population can be “split”, meaning that the organoid is split apart and all cells of the organoid are divided into new culture dishes or flasks. The divided cells are removed from the organoid and can then themselves be cultured and expanded to produce new organoids containing further expanded populations that can then be split again. Splits are also referred to herein as “passages”. An organoid of the invention may be cultured for 1 or more passages, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30 or more passages, for example, 20-30 passages, 30-35 passages, 32-40 passages or more. In some embodiments, an expanding cell population or organoid is split at every 3 days to every 10 weeks, every 1 to 10 weeks, every 1 to 4 weeks or every 10 days to 3 weeks. In some embodiments, the organoids are passaged approximately every 10 days to 3 weeks. Thus, the organoids of the invention can provide an ongoing source of genetically stable cellular material. Thus, the organoids of the invention can be used to gain mechanistic insight into a variety of diseases and therapeutics, to carry out in vitro drug screening, to evaluate potential therapeutics, to identify possible targets (e.g., proteins) for future novel (drug) therapy development and/or to explore gene repair coupled with cell-replacement therapy.
  • For these reasons, the organoids or expanded populations of cells of the invention can be a tool for drug screening, target validation, target discovery, toxicology and toxicology screens and personalized medicine.
  • Drug Screening
  • For preferably high-throughput purposes, said expanded stem cell population or organoid of the invention is cultured in multiwell plates such as, for example, 96 well plates or 384 well plates. Libraries of molecules are used to identify a molecule that affects said organoids. Preferred libraries comprise antibody fragment libraries, peptide phage display libraries, peptide libraries (e.g. LOPAP™, Sigma Aldrich), lipid libraries (BioMol), synthetic compound libraries (e.g. LO PAC™, Sigma Aldrich) or natural compound libraries (Specs, TimTec). Furthermore, genetic libraries can be used that induce or repress the expression of one of more genes in the progeny of the stem cells. These genetic libraries comprise cDNA libraries, antisense libraries, and siRNA or other non-coding RNA libraries. The cells are preferably exposed to multiple concentrations of a test agent for a certain period of time. At the end of the exposure period, the cultures are evaluated. The term “affecting” is used to cover any change in a cell, including, but not limited to, a reduction in, or loss of, proliferation, a morphological change, and cell death. Said expanded stem cell population or organoid of the invention can also be used to identify drugs that specifically target breast cancer cells, e.g. the breast cancer cells or breast cancer organoids of the invention, but not normal breast cells, e.g. not said normal expanded stem cell population or normal organoid of the invention. Of course, it is not necessary for the drug screening always to be high throughput and the organoids and cells of the invention are also useful for testing individual drug candidates.
  • The organoids are also useful for wider drug discovery purposes. It will be understood by the skilled person that the organoids of the invention would be widely applicable as drug screening tools for infectious, inflammatory and neoplastic pathologies of the human breast and other diseases of the breast. In some embodiments the organoids of the invention could be used for screening for cancer drugs.
  • In some embodiments, the expanded cell populations, for example the organoids of the invention or organoids obtained using media and methods of the invention can be used to test libraries of chemicals, antibodies, natural product (plant extracts), etc or specific known drugs for suitability for use as drugs, cosmetics and/or preventative medicines. For instance, in some embodiments, a cell biopsy from a patient of interest, such as tumour cells from a cancer patient, can be cultured using culture media and methods of the invention and then treated with a chemical compound or a chemical library or one or more drugs of interest. It is then possible to determine which compounds effectively modify, kill and/or treat the patient's cells. This allows specific patient responsiveness to a particular drug to be tested thus allowing treatment to be tailored to a specific patient. Thus, this allows a personalized medicine approach. The added advantage of using the organoids for identifying drugs in this way is that it is also possible to screen normal organoids (organoids derived from healthy tissue) to check which drugs and compounds have minimal effect on healthy tissue. This allows screening for drugs with minimal off-target activity or unwanted side-effects.
  • Drugs for any number of diseases can be screened in this way. For example, the organoids of the invention can be used for screening for drugs for breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • The testing parameters depend on the disease of interest. For example, when screening for cancer drugs, cancer cell death is usually the ultimate aim. In other embodiments, metabolics or gene expression may be evaluated to study the effects of compounds and drugs of the screen on the cells or organoids of interest.
  • Therefore, the invention provides a method for screening for a therapeutic or prophylactic drug, wherein the method comprises:
      • culturing an expanded cell population (for example, an organoid) of the invention, for example with a culture medium of the invention;
      • exposing said expanded cell population (for example, an organoid) of the invention to one or a library of candidate molecules;
      • evaluating said expanded cell populations (for example, organoids) for any effects, for example any change in the cell, such as a reduction in or loss of proliferation, a morphological change and/or cell death;
      • identifying the candidate molecule that causes said effects as a potential drug.
  • In some embodiments, computer- or robot-assisted culturing and data collection methods are employed to increase the throughput of the screen.
  • In some embodiments, expanded cell population (for example, an organoid) is obtained from a patient biopsy. In some embodiments, the candidate molecule that causes a desired effect on the cultured expanded cell population (for example, an organoid) is administered to said patient.
  • Accordingly, in one aspect, there is provided a method of treating a patient comprising:
      • (a) obtaining a biopsy from the diseased tissue of interest in the patient;
      • (b) screening for a suitable drug using a screening method of the invention, for example: culturing an expanded cell population (for example, an organoid) of the invention with a culture medium of the invention;
        • exposing said expanded cell population (for example, an organoid) of the invention to one or a library of known drugs;
        • evaluating said expanded cell populations (for example, organoids) for any effects, for example any change in the cell, such as a reduction in or loss of proliferation, a morphological change and/or cell death;
        • identifying the candidate molecule that causes said effects as a suitable drug; and
      • (c) treating said patient with the drug obtained in step (b).
  • For example, the invention provides a method of treating breast cancer in a patient, comprising testing the patient's responsiveness to one or more drugs of interest using a drug screening method of the invention and then treating a patient with the drug if the patient is found to be responsive to the drug.
  • Similarly, there is provided a breast cancer drug for use in treating a patient with breast cancer, wherein said treatment comprises testing the patient's responsiveness to the drug using a drug screening method of the invention and treating the patient with the drug if the patient is found to be responsive to the drug.
  • Examples of drugs for use in the invention which can be tested using a personalized medicine approach as outlined above include Abitrexate (Methotrexate), Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation), Ado-Trastuzumab Emtansine, Adriamycin PFS (Doxorubicin Hydrochloride), Adriamycin RDF (Doxorubicin Hydrochloride), Adrucil (Fluorouracil), Afinitor (Everolimus), Anastrozole, Aredia (Pamidronate Disodium), Arimidex (Anastrozole), Aromasin (Exemestane), Capecitabine, Clafen (Cyclophosphamide), Cyclophosphamide, Cytoxan (Cyclophosphamide), Docetaxel, Doxorubicin Hydrochloride, Efudex (Fluorouracil), Ellence (Epirubicin Hydrochloride), Epirubicin Hydrochloride, Everolimus, Exemestane, Fareston (Toremifene), Faslodex (Fulvestrant), Femara (Letrozole), Fluoroplex (Fluorouracil), Fluorouracil, Folex (Methotrexate), Folex PFS (Methotrexate), Fulvestrant, Gemcitabine Hydrochloride, Gemzar (Gemcitabine Hydrochloride), Goserelin Acetate, Herceptin (Trastuzumab), Ixabepilone, Ixempra (Ixabepilone), Kadcyla (Ado-Trastuzumab Emtansine), Lapatinib Ditosylate, Letrozole, Megace (Megestrol Acetate), Megestrol Acetate, Methotrexate, Methotrexate LPF (Methotrexate), Mexate (Methotrexate), Mexate-AQ (Methotrexate), Neosar (Cyclophosphamide), Nolvadex (Tamoxifen Citrate), Novaldex (Tamoxifen Citrate), Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Pamidronate Disodium, Pejeta (Pertuzumab), Pertuzumab, Tamoxifen Citrate, Taxol (Paclitaxel), Taxotere (Docetaxel), Trastuzumab, Toremifene, Tykerb (Lapatinib Ditosylate), Xeloda (Capecitabine) and Zoladex (Goserelin Acetate).
  • Drugs for use in the invention which can be tested using a personalized medicine approach may be tested alone or in combination with one or more other drugs.
  • For example, (a) Doxorubicin Hydrochloride, Adriamycin PFS (Doxorubicin Hydrochloride) or Adriamycin RDF (Doxorubicin Hydrochloride) may be tested in combination with (b) Cyclophosphamide, Clafen (Cyclophosphamide), Cytoxan (Cyclophosphamide) or Neosar (Cyclophosphamide), and, optionally, in further combination with (cxi) Paclitaxel, Paclitaxel Albumin-stabilized Nanoparticle Formulation, Abraxane (Paclitaxel Albumin-stabilized Nanoparticle Formulation) or Taxol (Paclitaxel), (cxii) Fluorouracil, Adrucil (Fluorouracil), Efudex (Fluorouracil) or Fluoroplex (Fluorouracil) or (cxiii) Docetaxel or Taxotere (Docetaxel). Thus, the AC, AC-T, CAF and TAC drug combinations may be tested using a personalized medicine approach according to the invention.
  • For example, (a) Cyclophosphamide, Clafen (Cyclophosphamide), Cytoxan (Cyclophosphamide) or Neosar (Cyclophosphamide) may be tested in combination with (b) Fluorouracil, Adrucil (Fluorouracil), Efudex (Fluorouracil) or Fluoroplex (Fluorouracil) and (cxi) Methotrexate, Abitrexate (Methotrexate), Folex (Methotrexate), Folex PFS (Methotrexate), Methotrexate LPF (Methotrexate), Mexate (Methotrexate) or Mexate-AQ (Methotrexate) or (cxii) Epirubicin Hydrochloride or Ellence (Epirubicin Hydrochloride). Thus, the CMF and FEC drug combinations may be tested using a personalized medicine approach according to the invention.
  • In some embodiments, the drug is used for treating, preventing or ameliorating symptoms of genetic diseases, metabolic diseases, pathogenic diseases, inflammatory diseases etc, for example including, but not limited to: breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • Target Discovery
  • In some embodiments, the organoids of the invention or cells grown using the culture media and methods of the invention can be used for target discovery. Cells of the organoids originating from healthy or diseased tissue may be used for target identification. The organoids of the invention may be used for discovery of drug targets for: breast cancer, adenoma, mammary duct ectasia, fibrocystic changes (e.g., adenosis, metaplasia, dysplasia, hyperplasia, ductal lesions, lobular lesions, papillomas), mastitis, syphilis of the breast, tuberculosis of the breast and actinomycosis of the breast.
  • Cells and organoids cultured according to the media and methods of the invention are thought to faithfully represent the in vivo situation. For this reason, they can be a tool to find novel (molecular) targets in specific diseases.
  • To search for a new drug target, a library of compounds (such as siRNA) may be used to transduce the cells and inactivate specific genes. In some embodiments, cells are transduced with siRNA to inhibit the function of a (large) group of genes. Any functional read out of the group of genes or specific cellular function can be used to determine if a target is relevant for the study. A disease-specific read out can be determined using assays well known in the art. For example, cellular proliferation is assayed to test for genes involved in cancer. For example, a Topflash assay as described herein, may be used to detect changes in Wnt activity caused by siRNA inhibition. Where growth reduction or cell death occurs, the corresponding siRNA related genes can be identified by methods known in the art. These genes are possible targets for inhibiting growth of these cells. Upon identification, the specificity of the identified target for the cellular process that was studied will need to be determined by methods well known in the art. Using these methods, new molecules can be identified as possible drug targets for therapy.
  • Target and Drug Validation Screens
  • Patient-specific organoids obtained from diseased and/or normal tissue can be used for target validation of molecules identified in high throughput screens. The same goes for the validation of compounds that were identified as possible therapeutic drugs in high throughput screens. The use of primary patient material expanded in the organoid culture system can be useful to test for false positives, etc. from high throughput drug discovery cell line studies.
  • In some embodiments, the expanded stem cell population (for example, organoid of the invention) can be used for validation of compounds that have been identified as possible drugs or cosmetics in a high-throughput screen.
  • Toxicity Assay
  • Said expanded stem cell population (for example, organoid of the invention) can further replace the use of cell lines in toxicity assays of potential novel drugs or of known drugs. Preferably, normal cells or organoids are used for these embodiments. However, it is also envisaged that cancer cells or organoids may be used.
  • Toxicology screens work in a similar way to drug screens (as described above), but they test for the toxic effects of drugs and not therapeutic effects. Therefore, in some embodiments, the effects of the candidate compounds are toxic.
  • Culturing Pathogens
  • Furthermore, said expanded stem cell population (for example, organoid of the invention) can be used for culturing of a pathogen such as mouse mammary tumour virus (MMTV), human mammary tumor virus (HMTV), Epstein Barr virus (EBV), high risk human papillomavirus (HPV), Mycobacterium tuberculosis, Streptococcus uberis, Streptococcus dysgalactiae, Escherichia coli, Staphylococcus aureus, or Streptococcus agalactiae.
  • Therapy Monitoring and Identifying Causes of Drug Resistance
  • Furthermore, said expanded stem cell population (for example, organoid of the invention) can be used for monitoring the progression of a disease in the expanded stem cell population alongside monitoring the progression of a disease in a patient. Thus, for example, an expanded population of breast cancer stem cells could be obtained from a patient with breast cancer, the patient could then be treated with a certain drug or combination of drugs, and if the patient develops resistance to the drug or combination of drugs then a second expanded population of breast cancer stem cells could be obtained from the patient and compared to the first population of breast cancer stem cells, in order to identify causes of drug resistance and an appropriate therapy.
  • Accordingly, said expanded stem cell population (for example, organoid of the invention) can be used for identifying causes of drug resistance. Thus, for example, an expanded population of breast cancer stem cells could be obtained from a patient with breast cancer, the patient could then be treated with a certain drug or combination of drugs, and if the patient develops resistance to the drug or combination of drugs then a second expanded population of breast cancer stem cells could be obtained from the patient and compared to the first population of breast cancer stem cells, in order to identify causes of drug resistance.
  • The causes of drug resistance may be identified by identifying genetic changes in the resistant population of stem cells compared to the population of stem cells that responds to treatment with the drug.
  • Regenerative Medicine and Transplantation
  • Cultures comprising the expanded stem cell population (for example, organoid of the invention) are useful in regenerative medicine, for example in post-radiation and/or post-surgery repair of breast tissue, for example post-mastectomy, or in tissue repair of injured breast tissue. For example, breast epithelial stem cells with BRCA mutations may be obtained from a cancer patient, the BRCA mutations may then be repaired, resulting in the generation of autologous breast epithelial stem cells, which may then be transplanted back into the cancer patient.
  • In an alternative embodiment, the expanded epithelial stem cells are reprogrammed into related tissue fates. It will be clear to a skilled person that gene therapy can additionally be used in a method directed at repairing damaged or diseased tissue. Use can, for example, be made of an adenoviral or retroviral gene delivery vehicle to deliver genetic information, like DNA and/or RNA to stem cells. A skilled person can replace or repair particular genes targeted in gene therapy. For example, a normal gene may be inserted into a nonspecific location within the genome to replace a non-functional gene. In another example, an abnormal gene sequence can be replaced for a normal gene sequence through homologous recombination. Alternatively, selective reverse mutation can return a gene to its normal function. A further example is altering the regulation (the degree to which a gene is turned on or off) of a particular gene. Preferably, the stem cells are ex vivo treated by a gene therapy approach and are subsequently transferred to the mammal, preferably a human being in need of treatment.
  • Since small biopsies taken from adult donors can be expanded without any apparent limit or genetic harm, the technology may serve to generate transplantable epithelium for regenerative purposes. Furthermore, in some embodiments, organoids embedded in, or in contact with, an ECM can be transplanted into a mammal, preferably into a human. In another embodiment, organoids and ECM can be transplanted simultaneously into a mammal, preferably into a human.
  • The skilled person would understand that an ECM can be used as a 3D scaffold for obtaining tissue-like structures comprising expanded populations of cells or organoids according to the invention. Such structures can then be transplanted into a patient by methods well known in the art. An ECM scaffold can be made synthetically using ECM proteins, such as collagen and/or laminin, or alternatively an ECM scaffold can be obtained by “decellularising” an isolated organ or tissue fragment to leave behind a scaffold consisting of the ECM (for example see Macchiarini et al. The Lancet, Volume 372, Issue 9655, Pages 2023-2030, 2008). In some embodiments, an ECM scaffold can be obtained by decellularising an organ or tissue fragment, wherein optionally said organ or tissue fragment is from the breast.
  • As mentioned above, the invention provides an organoid or population of cells of the invention for use in transplantation into a mammal, preferably into a human.
  • Advantageously, the invention enables a small biopsy to be taken from an adult donor and expanded without any apparent limit or genetic harm and so the technology provided herein may serve to generate transplantable epithelium for regenerative purposes.
  • The patient is preferably a human, but may alternatively be a non-human mammal, such as a cat, dog, horse, cow, pig, sheep, rabbit or mouse.
  • Thus, included within the scope of the invention are methods of treatment of a human or non-human animal patient through cellular therapy. Such cellular therapy encompasses the application of the stem cells or organoids of the invention to the patient through any appropriate means. Specifically, such methods of treatment involve the regeneration of damaged tissue. In accordance with the invention, a patient can be treated with allogeneic or autologous stem cells or organoids. “Autologous” cells are cells which originated from the same organism into which they are being re-introduced for cellular therapy, for example in order to permit tissue regeneration. However, the cells have not necessarily been isolated from the same tissue as the tissue they are being introduced into. An autologous cell does not require matching to the patient in order to overcome the problems of rejection. “Allogeneic” cells are cells which originated from an individual which is different from the individual into which the cells are being introduced for cellular therapy, for example in order to permit tissue regeneration, although of the same species. Some degree of patient matching may still be required to prevent the problems of rejection.
  • Generally, the cells or organoids of the invention are introduced into the body of the patient by injection or implantation. Generally, the cells will be directly injected into the tissue in which they are intended to act. A syringe containing cells of the invention and a pharmaceutically acceptable carrier is included within the scope of the invention. A catheter attached to a syringe containing cells of the invention and a pharmaceutically acceptable carrier is included within the scope of the invention.
  • The skilled person will be able to select an appropriate method and route of administration depending on the material that is being transplanted (i.e., population of cells, single cells in cell suspension, organoids or fragments of organoids).
  • As discussed above, cells of the invention can be used in the regeneration of tissue. In order to achieve this function, cells may be injected or implanted directly into the damaged tissue, where they may multiply and eventually differentiate into the required cell type. Alternatively, the organoid can be injected or implanted directly into the damaged tissue. Breast tissues that are susceptible to treatment include all damaged tissues, particularly including those which may have been damaged by disease (e.g. cancer), injury, trauma, an autoimmune reaction, or by a viral or bacterial infection. In some embodiments of the invention, the cells or organoids of the invention are used to regenerate the breast.
  • For example, in one embodiment, the cells or organoids of the invention are injected into a patient using a Hamilton syringe. Accordingly, the invention provides a syringe comprising the cells or organoids of the invention.
  • The skilled person will be aware what the appropriate dosage of cells or organoids of the invention will be for a particular condition to be treated.
  • In one embodiment the cells or organoids of the invention, either in solution, in microspheres or in microparticles of a variety of compositions, will be administered into the artery irrigating the tissue or the part of the damaged organ in need of regeneration. Generally, such administration will be performed using a catheter. The catheter may be one of the large variety of balloon catheters used for angioplasty and/or cell delivery or a catheter designed for the specific purpose of delivering the cells to a particular local of the body. For certain uses, the cells or organoids may be encapsulated into microspheres made of a number of different biodegradable compounds, and with a diameter of about 15 μm. This method may allow intravascularly administered cells or organoids to remain at the site of damage, and not to go through the capillary network and into the systemic circulation in the first passage. The retention at the arterial side of the capillary network may also facilitate their translocation into the extravascular space.
  • In another embodiment, the cells or organoids of the invention may be implanted into the damaged tissue adhered to a biocompatible implant. Within this embodiment, the cells may be adhered to the biocompatible implant in vitro, prior to implantation into the patient. As will be clear to a person skilled in the art, any one of a number of adherents may be used to adhere the cells to the implant, prior to implantation. By way of example only, such adherents may include fibrin, one or more members of the integrin family, one or more members of the cadherin family, one or more members of the selectin family, one or more cell adhesion molecules (CAMs), one or more of the immunoglobulin family and one or more artificial adherents. This list is provided by way of illustration only, and is not intended to be limiting. It will be clear to a person skilled in the art, that any combination of one or more adherents may be used.
  • In another embodiment, the cells or organoids of the invention may be embedded in a matrix, prior to implantation of the matrix into the patient. Generally, the matrix will be implanted into the damaged tissue of the patient. Examples of matrices include collagen based matrices, fibrin based matrices, laminin based matrices, fibronectin based matrices and artificial matrices. This list is provided by way of illustration only, and is not intended to be limiting.
  • In a further embodiment, the cells or organoids of the invention may be implanted or injected into the patient together with a matrix forming component. This may allow the cells to form a matrix following injection or implantation, ensuring that the cells or organoids remain at the appropriate location within the patient. Examples of matrix forming components include fibrin glue liquid alkyl, cyanoacrylate monomers, plasticizers, polysaccharides such as dextran, ethylene oxide-containing oligomers, block co-polymers such as poloxamer and Pluronics, non-ionic surfactants such as Tween and Triton ‘8’, and artificial matrix forming components. This list is provided by way of illustration only, and is not intended to be limiting. It will be clear to a person skilled in the art, that any combination of one or more matrix forming components may be used.
  • In a further embodiment, the cells or organoids of the invention may be contained within a microsphere. Within this embodiment, the cells may be encapsulated within the centre of the microsphere. Also within this embodiment, the cells may be embedded into the matrix material of the microsphere. The matrix material may include any suitable biodegradable polymer, including but not limited to alginates, Poly ethylene glycol (PLGA), and polyurethanes. This list is provided by way of example only, and is not intended to be limiting.
  • In a further embodiment, the cells or organoids of the invention may be adhered to a medical device intended for implantation. Examples of such medical devices include stitches and artificial skin. This list is provided by way of illustration only, and is not intended to be limiting. It will be clear to a person skilled in the art, that the cells may be adhered to the medical device by a variety of methods. For example, the cells or organoids may be adhered to the medical device using fibrin, one or more members of the integrin family, one or more members of the cadherin family, one or more members of the selectin family, one or more cell adhesion molecules (CAMs), one or more of the immunoglobulin family and one or more artificial adherents. This list is provided by way of illustration only, and is not intended to be limiting. It will be clear to a person skilled in the art, that any combination of one or more adherents may be used.
  • Other Uses of Organoids of the Invention
  • The cells or organoids of the invention may be used to define patient groups for drug development. The cells or organoids of the invention may also be used for pre-screening patients in phase I or phase II trials. For example, the cells or organoids of the invention may be used to identify patients on which to perform a clinical trial. The cells or organoids of the invention may also be used in diagnostics after a drug is approved. For example, the cells or organoids of the invention may be used to identify patients who are most likely to benefit from a particular drug, to identify patients likely to be at increased risk for serious side effects as a result of treatment with a particular drug or to monitor the response to treatment with a particular drug for the purpose of adjusting treatment to achieve improved safety or effectiveness.
  • The cells or organoids of the invention may in some embodiments be used for veterinary therapies. For example, the cells or organoids may be used in the treatment of mastitis in cows.
  • Compositions and Other Forms of the Invention
  • The invention provides a pharmaceutical composition comprising the cells or organoids of the invention and further comprising a pharmaceutically acceptable carrier or excipient.
  • The invention provides a composition comprising a culture medium according to the invention and stem cells. The invention also provides a composition comprising a culture medium according to the invention and organoids. Furthermore, the invention provides a composition comprising a culture medium according to the invention and an extracellular matrix.
  • The invention also provides a composition comprising a culture medium of the invention, an extracellular matrix and stem cells of the invention. The invention also provides a composition comprising a culture medium of the invention, an extracellular matrix and one or more organoids of the invention. The invention also provides a culture medium supplement that can be used to produce a culture medium as disclosed herein. A ‘culture medium supplement’ is a mixture of ingredients that cannot itself support stem cells, but which enables or improves stem cell culture when combined with other cell culture ingredients. The supplement can therefore be used to produce a functional cell culture medium of the invention by combining it with other cell culture ingredients to produce an appropriate medium formulation. The use of culture medium supplements is well known in the art.
  • The invention provides a culture medium supplement that comprises an ErbB3/4 ligand according to the invention. The supplement may contain any ErbB3/4 ligand (or combination of ErbB3/4 ligand) disclosed herein. The supplement may also contain one or more additional cell culture ingredients as disclosed herein, e.g. one or more cell culture ingredients selected from the group consisting of amino acids, vitamins, inorganic salts, carbon energy sources and buffers.
  • A culture medium or culture medium supplement may be a concentrated liquid culture medium or supplement (e.g. a 2× to 250× concentrated liquid culture medium or supplement) or may be a dry culture medium or supplement. Both liquid and dry culture media or supplements are well known in the art. A culture medium or supplement may be lyophilised.
  • A culture medium or supplement of the invention will typically be sterilized prior to use to prevent contamination, e.g. by ultraviolet light, heating, irradiation or filtration. A culture medium or culture medium supplement may be frozen (e.g. at −20° C. or −80° C.) for storage or transport. In some embodiments, the culture medium may be stored as a liquid (e.g. at approximately 4° C.). In some embodiments, the culture medium may be split and stored as two components: a frozen component (e.g. at between approximately −20° C. and approximately −80° C.) and a liquid component (e.g. at approximately 4° C.). In particular, temperature-sensitive or time-sensitive degradable material is preferably included in the frozen component, whereas less sensitive material (for example DMEM or FCS) can be stored in the liquid form and thus included in the liquid component for storage and shipping.
  • The invention also provides a hermetically-sealed vessel containing a culture medium or culture medium supplement of the invention. Hermetically-sealed vessels may be preferred for transport or storage of the culture media or culture media supplements disclosed herein, to prevent contamination. The vessel may be any suitable vessel, such as a flask, a plate, a bottle, a jar, a vial or a bag.
  • The invention also provides a kit comprising a culture medium, culture medium supplement and/or a composition of the invention. In some embodiments, the kit further comprises at least one other additional component, for example selected from the list comprising: an ECM (for example, Matrigel™ or Cultrex® Basement Membrane Extract), a population of cells and an organoid.
  • General
  • The term “comprising” encompasses “including” as well as “consisting” e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X+Y.
  • The word “substantially” does not exclude “completely” e.g. a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from the definition of the invention.
  • The term “about” in relation to a numerical value x is optional and means, for example, x±10%.
  • Unless specifically stated, a process comprising a step of mixing two or more components does not require any specific order of mixing. Thus components can be mixed in any order. Where there are three components then two components can be combined with each other, and then the combination may be combined with the third component, etc.
  • Various aspects and embodiments of the invention are described below in more detail by way of example. It will be appreciated that modification of detail may be made without departing from the scope of the invention.
  • Examples Example 1—Tissue Processing and Mammary Organoid Culture Overview
  • 20 breast tumors were sampled, three of which were triple negative (15%, FIG. 1A and FIG. 13). We successfully established three tumor organoid lines (splitting ratio >1:2, passage >7). 8 additional cultures are currently expanding and will, based on our experience, most likely generate successful organoid lines (FIG. 2 and FIG. 13). A selection for a specific subtype is not evident (FIG. 1B).
  • From the 19 normal samples obtained we thus far established three promising cultures (FIG. 2, FIG. 13).
  • Culture Conditions
  • The first 12 samples were used to optimize cell isolation and organoid culture conditions. Samples 13-20 were grown in six different conditions.
  • Alerted Parameters for Cell Isolation
  • For the isolation of viable cells we tested several digestive enzymes (collagenase, dispase, trypsin) at different concentrations (1-5 mg/ml) in different media (AdDF+++, base medium, base-F7/10N medium) for different periods of time (1-2 h, 12 h) with or without agitation. The compositions of these base media are displayed in FIG. 14. The optimal protocol is summarized below.
  • Alerted Parameters for Organoid Culture
  • To allow organoid growth we tested several culture conditions in parallel (6-12 conditions/sample). Based on previous experience with other human tissue we tested several growth factors on top of our base medium (FIG. 14). Other parameters tested included BME (basement membrane extract) density (50-100%) instead of Matrigel, the use of tissue culture treated vs suspension plates, and culturing at 2% vs ambient Oz. An example of the effect of different culture conditions is given for normal and tumor organoid lines obtained from patient W855 (FIG. 3).
  • Protocol for Tissue Processing and Mammary Organoid Culture
  • Upon arrival, tissues were photographed and cut into 1-3 mm3 pieces. Two random pieces were snap frozen and stored at −80° C. for DNA isolation, two random pieces were fixed in formalin for histopathological analysis and immunohistochemistry, and the remainder was processed for the isolation of viable cells. The remaining tissue was minced, washed with 10 ml AdDF+++ and digested in 10 ml base-F7/10N medium containing 1-2 mg/ml collagenase (Sigma-C9407) at 120 rpm and 37° C. for 1-2h. The digested tissue suspension was sequentially sheared using 10 ml and 5 ml plastic and flamed glass Pasteur pipettes. After every shearing step the suspension was strained over a 100 μm filter with retained tissue pieces entering a subsequent shearing step with ˜10 ml AdDF+++. 2% FCS were added to the strained suspension before centrifugation at 400 g. The pellet was resuspended in 10 ml AdDF+++ and centrifuged again at 400. In case of a visible red pellet, erythrocytes were lysed in 2 ml red blood cell lysis buffer (Roche-11814389001) for 5 min at room temperature before the addition of 10 ml AdDF+++ and centrifugation at 400 g. The pellet was resuspended in 10 mg/ml cold Cultrex growth factor reduced BME type 2 (Trevigen-3533-010-02) and 40 μl drops of BME-cell suspension were allowed to solidify on prewarmed 24-well suspension culture plates (Greiner-M9312) at 37° C. for 10-20 min. Upon completed gelation, 400 μl of organoid media (base-F7/10N, base-F7/10N, or base-F7/10N) were added to each well and plates transferred to humidified 37° C./5% CO2 incubators at either 2% or ambient Oz.
  • Medium was changed every 4 days and organoids were passaged every 1-4 weeks: cystic organoids were resuspended in 2 ml cold AdDF+++ and mechanically sheared through flamed glass Pasteur pipettes. Dense organoids were dissociated by resuspension in 2 ml TrypLE Express (Invitrogen-12605036), incubation for 1-5 min at room temperature, and mechanical shearing through flamed glass Pasteur pipettes. Following the addition of 10 ml AdDF+++ and centrifugation at 300 g or 400 g respectively, organoid fragments were resuspended in cold BME and reseeded as above at ratios (1:1-1:6) allowing the formation of new organoids. Single cell suspensions were initially seeded at high density and reseeded at a lower density after ˜1 week.
  • Example 2—Characterization of Established Mammary Tumor Organoid Lines
  • Mammary tumor organoid lines W854T, W855T, and W859T readily expand in base-F7/10N medium and were therefore characterized in more detail.
  • Their tumor status was confirmed through karyotyping revealing aneuploidy at varying degrees (FIG. 4).
  • Line W854T furthermore readily grew in medium containing 5 μM Nutlin-3 indicating the presence of mutant p53. RNA and DNA were isolated and sent to BI for gene expression profiling and exome sequencing.
  • Histological and immunofluorescent analysis confirmed that ER, PR, and HER2 status are conserved between originating tumor and established tumor organoid line after >7 passages (FIGS. 5 and 6).
  • Whereas normal organoid line W855N consists of mutually exclusive basal and luminal cells, tumor organoids solely express luminal cell marker Keratin-8 (FIGS. 6 and 7). E-cadherin expression is present but decreased in tumor compared to normal organoid lines indicating their epithelial origin (FIGS. 6 and 7). However, markers of epithelial to mesenchymal transition (EMT) are mildly upregulated in tumor organoid lines W855T and W859T corresponding to their incoherent phenotype (FIGS. 6 and 7).
  • Example 3—Low Throughput Drug Screen
  • To test the feasibility of mammary tumor organoids for drug screening, lines W854T, W855T, and W859T were plated in duplicate into a 96-well format and exposed to medium containing the EGFR inhibitor Iressa (1 nM-30 μM at 1:3 dilution steps, S1025 Selleck Chemicals) or the p53 stabilizer Nutlin-3 (1 nM-30 μM at 1:3 dilution steps, 10004372 Cayman Chemicals) for 7 days. Organoids were photographed every 3-4 days (FIG. 8) and cell viability measured at day 7 using the CellTiter-Glo 2.0 assay (G9241, Promega) (FIG. 9).
  • All three organoid lines respond to Iressa at concentrations ˜10 μM indicating a dependence on EGFR signaling. With perhaps the exception of W855T, Nutlin-3 does not inhibit growth at concentrations up 30 μM indicating the presence of mutant p53 (FIG. 9). Mammary tumor organoids are suitable for cell viability based low throughput drug screens in a 96-well format. We are currently testing the assay in a 384-well plate format which has been successfully established for colon cancer organoids.
  • Example 4—Culture Medium of Pasic et al. Does not Support Long-Term Growth
  • Normal mammary organoids in Pasic's culture medium slowed expanding at passage 2 and stopped at passage 3; organoids from the same patient grown in base-F7/10E continued expanding up to passage 4 (see FIG. 10). Normal mammary organoids from different patients expand at least up to passage 6 in medium base F7/10 EN.

Claims (37)

1-30. (canceled)
31. A breast organoid comprising a population of breast epithelial stem cells obtained from primary breast cancer or metastatic breast cancer.
32. A breast organoid comprising a population of breast epithelial stem cells in a culture medium comprising an ErbB3/4 ligand, one or more FGFR2b ligands, and a Wnt agonist, wherein the Wnt agonist is an Lgr5 agonist or Wnt3a.
33. The breast organoid of claim 32, wherein the culture medium is suitable for expanding a population of breast epithelial stem cells for at least 4 passages.
34. The breast organoid of claim 32, wherein the breast organoid has been cultured or can be cultured for more than 4 passages.
35. The breast organoid of claim 32, wherein the breast organoid has been cultured for at least 2 months, and wherein the breast epithelial stem cells have been passaged approximately every 1 to 4 weeks.
36. The breast organoid of claim 32, wherein the breast organoid has a cyst-like structure.
37. The breast organoid of claim 36, wherein the cyst-like structure has a central lumen.
38. The breast organoid of claim 37, wherein the central lumen of the cyst-like structure: (a) does not comprise any cells; or (b) comprises breast epithelial stem cells, optionally wherein the central lumen is filled with breast epithelial stem cells.
39. The breast organoid of claim 37, wherein the breast epithelial cells surround the central lumen, optionally wherein the breast epithelial cells surrounding the central lumen are polarized.
40. The breast organoid of claim 32, comprising:
(a) a continuous epithelium and intercellular adhesion, and/or
(b) luminal cells but no basal cells.
41. The breast organoid of claim 32, wherein the breast epithelial stem cells are multipotent, oligopotent, or unipotent organ-specific adult stem cells.
42. The breast organoid of claim 32, wherein the breast organoid consists essentially of epithelial cells.
43. The breast organoid of claim 32, wherein non-epithelial stem cells are absent from the breast organoids.
44. The breast organoid of claim 32, wherein the breast organoid:
(a) has a three dimensional structure, and/or
(b) comprise breast epithelial stem cells which are able to actively divide, optionally wherein the breast epithelial stem cells are able to differentiate to all major differentiated cell lineages present in the corresponding in vivo tissue, and/or
(c) have a section which is formed of multiple layers, and/or
(d) has a monolayer of pseudo-stratified cells folds so that it encloses lumina between the folds.
45. The breast organoid of claim 32, wherein the breast organoid is a breast cancer organoid.
46. The breast organoid of claim 32, wherein the breast organoid is obtained from normal tissue.
47. The breast organoid of claim 32, wherein the ErbB3/4 ligand is a neuregulin polypeptide.
48. The breast organoid of claim 47, wherein the neuregulin polypeptide is encoded by NRG.
49. The breast organoid according to claim 47, wherein the neuregulin polypeptide comprises the amino acid sequence of SEQ ID NO: 27.
50. The breast organoid of claim 32, wherein the one or more FGFR2b ligands is FGF7 and/or FGF10.
51. The breast organoid of claim 32, wherein the Wnt agonist is an Lgr5 agonist.
52. The breast organoid of claim 51, wherein the Lgr5 agonist is a Rspondin or a Rspondin mimic.
53. The breast organoid of claim 32, wherein the culture medium further comprises a BMP inhibitor.
54. The breast organoid of claim 32, wherein the culture medium further comprises one or more components selected from: B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor, and a p38 inhibitor.
55. The breast organoid of claim 32, wherein the culture medium further comprises B27, N-acetylcysteine, Nicotinamide, a ROCK inhibitor, a TGF-beta inhibitor, and a p38 inhibitor.
56. The breast organoid of claim 32, wherein:
(a) the BMP inhibitor is Noggin, and/or
(b) the ROCK inhibitor is Y-27632, and/or
(c) the TGF-beta inhibitor is A83-01, and/or
(d) the p38 inhibitor is SB 202190.
57. The breast organoid of claim 32, wherein the culture medium further comprises one or more components selected from: one or more further receptor tyrosine kinase ligands and a p53 stabilizing agent.
58. The breast organoid of claim 57, wherein:
(a) the one or more further receptor tyrosine kinase ligands are EGF, PDGF and/or amphiregulin, and/or
(b) the p53 stabilizing agent is a member of the Nutlin family.
59. The breast organoid of claim 58, wherein:
(a) the PDGF is PDGF-CC, and/or
(b) the member of the Nutlin family is Nutlin-3.
60. The breast organoid of claim 32 obtained by a method comprising:
providing a population of breast epithelial stem cells;
providing a culture medium comprising an ErbB3/4 ligand, one or more FGFR2b ligands and a Wnt agonist, wherein the Wnt agonist is an Lgr5 agonist or Wnt3a; and wherein the culture medium is suitable for expanding a population of breast epithelial stem cells for at least 6 passages;
contacting the breast epithelial stem cells with the culture medium; and
culturing the breast epithelial stem cells under appropriate conditions to form a breast organoid.
61. The breast organoid of claim 60, wherein the ErbB3/4 ligand is a neuregulin polypeptide.
62. The breast organoid of claim 60, wherein the culture medium comprises an ErbB3/4 ligand, one or more FGFR2b ligands, and a Wnt agonist, wherein the Wnt agonist is an Lgr5 agonist or Wnt3a, and wherein the ErbB3/4 ligand is a neuregulin polypeptide encoded by NRG.
63. A composition comprising a breast organoid of claim 32.
64. The composition of claim 63, wherein the composition further comprises an extracellular matrix.
65. A method for a drug discovery screen; toxicity assay; research of tissue embryology, cell lineages, or differentiation pathways; gene expression study including recombinant gene expression; research of mechanisms involved in tissue injury or repair; research of inflammatory and infectious diseases; studies of pathogenetic mechanisms; or study of mechanisms of cell transformation or aetiology of cancer, comprising utilizing the breast organoid of claim 32.
66. A method of treating a disease in a subject, comprising administering an effective amount of the breast organoid of claim 32, or cells derived therefrom to a subject in need thereof.
US17/182,457 2014-11-27 2021-02-23 Culture Medium for Expanding Breast Epithelial Stem Cells Pending US20210254017A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/182,457 US20210254017A1 (en) 2014-11-27 2021-02-23 Culture Medium for Expanding Breast Epithelial Stem Cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB1421094.2A GB201421094D0 (en) 2014-11-27 2014-11-27 Culture medium
GB1421094.2 2014-11-27
PCT/EP2015/077988 WO2016083612A1 (en) 2014-11-27 2015-11-27 Culture medium for expanding breast epithelial stem cells
US201715529150A 2017-05-24 2017-05-24
US17/182,457 US20210254017A1 (en) 2014-11-27 2021-02-23 Culture Medium for Expanding Breast Epithelial Stem Cells

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/529,150 Division US10961511B2 (en) 2014-11-27 2015-11-27 Culture medium for expanding breast epithelial stem cells
PCT/EP2015/077988 Division WO2016083612A1 (en) 2014-11-27 2015-11-27 Culture medium for expanding breast epithelial stem cells

Publications (1)

Publication Number Publication Date
US20210254017A1 true US20210254017A1 (en) 2021-08-19

Family

ID=52349562

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/529,150 Active US10961511B2 (en) 2014-11-27 2015-11-27 Culture medium for expanding breast epithelial stem cells
US17/182,457 Pending US20210254017A1 (en) 2014-11-27 2021-02-23 Culture Medium for Expanding Breast Epithelial Stem Cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/529,150 Active US10961511B2 (en) 2014-11-27 2015-11-27 Culture medium for expanding breast epithelial stem cells

Country Status (11)

Country Link
US (2) US10961511B2 (en)
EP (2) EP3950937A1 (en)
JP (2) JP7261538B2 (en)
AU (1) AU2015352350B2 (en)
CA (1) CA2968751A1 (en)
DK (1) DK3224351T3 (en)
ES (1) ES2883159T3 (en)
GB (1) GB201421094D0 (en)
HK (1) HK1245324A1 (en)
PT (1) PT3224351T (en)
WO (1) WO2016083612A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023137465A1 (en) * 2022-01-13 2023-07-20 BIOMILQ, Inc. Optimization of live cell constructs for production of cultured milk product and methods of using the same

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2412800A1 (en) 2010-07-29 2012-02-01 Koninklijke Nederlandse Akademie van Wetenschappen Liver organoid, uses thereof and culture method for obtaining them
CA2751332C (en) 2009-02-03 2024-04-02 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
WO2011140441A2 (en) 2010-05-06 2011-11-10 Children's Hospital Medical Center Methods and systems for converting precursor cells into intestinal tissues through directed differentiation
AU2015261380B2 (en) 2014-05-16 2021-04-15 Koninklijke Nederlandse Akademie Van Wetenschappen Improved culture method for organoids
ES2860423T3 (en) 2014-05-28 2021-10-05 Childrens Hospital Med Ct Methods and Systems for Converting Stem Cells to Gastric Tissues by Targeted Differentiation
CA2963704A1 (en) 2014-10-17 2016-04-21 Children's Hospital Medical Center In vivo model of human small intestine using pluripotent stem cells and methods of making and using same
GB201421094D0 (en) 2014-11-27 2015-01-14 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium
GB201421092D0 (en) 2014-11-27 2015-01-14 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium
GB201603569D0 (en) 2016-03-01 2016-04-13 Koninklijke Nederlandse Akademie Van Wetenschappen Improved differentiation method
US11066650B2 (en) 2016-05-05 2021-07-20 Children's Hospital Medical Center Methods for the in vitro manufacture of gastric fundus tissue and compositions related to same
GB201611982D0 (en) 2016-07-11 2016-08-24 Cellesce Ltd Cell culture
WO2018067925A1 (en) * 2016-10-06 2018-04-12 Transgenex Nanobiotech, Inc. Methods for cancer stem cell (csc) expansion
AU2017373767B2 (en) 2016-12-05 2021-09-16 Children's Hospital Medical Center Colonic organoids and methods of making and using same
JP2020521477A (en) * 2017-05-29 2020-07-27 ステムセル テクノロジーズ カナダ インコーポレイテッド Compositions and methods for obtaining organoids
EP3645022A4 (en) * 2017-06-28 2021-07-21 Rutgers, the State University of New Jersey Single breast cell-derived organoids
WO2019133767A1 (en) * 2017-12-28 2019-07-04 Development Center For Biotechnology A method of in vitro diagnostic for prediction of drug efficacy
CA3113675A1 (en) * 2018-09-27 2020-04-02 The Regents Of The University Of California Methods of human retinal progenitor cell isolation and culture
AU2019361030A1 (en) * 2018-10-16 2021-05-27 Board Of Regents, The University Of Texas System Compositions for and methods of producing tumor organoids
GB201903573D0 (en) 2019-03-15 2019-05-01 Koninklijke Nederlandse Akademie Van Wetenschappen Culture method for head and neck organoids
GB201906978D0 (en) 2019-05-17 2019-07-03 Koninklijke Nederlandse Akademie Van Wetenschappen Improved culture method using integrin agonist
EP4031652A4 (en) * 2019-09-19 2023-10-11 Northwestern University Cost effective culture media and protocol for human induced pluripotent stem cells
WO2021072238A1 (en) * 2019-10-10 2021-04-15 University Of Houston System Feeder-based and feeder-free stem cell culture systems for stratified epithelial stem cells, and uses related thereto
CN112779209B (en) * 2019-11-08 2023-01-24 合肥中科普瑞昇生物医药科技有限公司 Primary mammary epithelial cell culture medium, culture method and application thereof
US11629385B2 (en) 2019-11-22 2023-04-18 Tempus Labs, Inc. Tumor organoid culture compositions, systems, and methods
WO2021113821A1 (en) 2019-12-05 2021-06-10 Tempus Labs, Inc. Systems and methods for high throughput drug screening
CN111424015A (en) * 2019-12-10 2020-07-17 重庆康克唯生物科技有限公司 Culture medium and three-dimensional culture method for prostate tumor cells
CN113528425B (en) * 2020-04-15 2023-05-30 合肥中科普瑞昇生物医药科技有限公司 Culture medium and culture method for mammary gland epithelial tumor cells
US11561178B2 (en) 2020-04-20 2023-01-24 Tempus Labs, Inc. Artificial fluorescent image systems and methods
NL2027546B1 (en) * 2021-02-11 2022-09-12 Prinses Maxima Centrum Voor Kinderoncologie B V Method of producing an organoid
EP4323502A1 (en) * 2021-04-16 2024-02-21 Stemcell Technologies Canada Inc. Media and methods for growing mammary organoids
TWI794821B (en) * 2021-05-20 2023-03-01 國立陽明交通大學 Method for in vitro cultivation of primary human pulmonary alveolar epithelial cells
CN113717933B (en) * 2021-07-05 2023-10-13 浙江大学 Application of FGF7 in preparation of stem cell expansion and phenotype maintaining reagent
CN114591887B (en) * 2022-03-14 2022-10-14 杭州艾名医学科技有限公司 Breast cancer organoid culture solution, preparation method and culture method
GB202204266D0 (en) 2022-03-25 2022-05-11 Stichting Hubrecht Organoid Tech Methods for co-culturing genotoxic bacteria and organoids

Family Cites Families (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995030900A1 (en) 1994-05-04 1995-11-16 Mount Sinai Hospital Corporation MODULATORS OF CYTOKINES OF THE TGF-β SUPERFAMILY AND METHODS FOR ASSAYING FOR SAME
EP0953633A1 (en) 1998-04-28 1999-11-03 Livercell L.L.C. Cell culturing method and medium for producing proliferated, normal, differentiated human liver cells
AU3859800A (en) 1999-02-24 2000-09-14 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Artificial salivary gland
US20030003088A1 (en) 2001-05-03 2003-01-02 Board Of Trustees Operating Michigan State University Human pancreatic pluripotential stem cell line
WO2003076564A2 (en) 2001-05-16 2003-09-18 The General Hospital Corporation Tissue-engineered organs
WO2003018752A2 (en) 2001-08-23 2003-03-06 The Wistar Institute Of Anatomy And Biology An organotypic intestinal culture and methods of use thereof
US20030138951A1 (en) 2001-10-18 2003-07-24 Li Yin Conversion of liver stem and progenitor cells to pancreatic functional cells
EP1567639A4 (en) 2002-12-05 2005-12-21 Technion Res & Dev Foundation Cultured human pancreatic islets, and uses thereof
WO2004087896A2 (en) 2003-03-31 2004-10-14 Pfizer Products Inc. Hepatocyte differentiation of stem cells
US20040229355A1 (en) 2003-05-14 2004-11-18 Board Of Regents University Of Texas System Culture medium for long-term culture of hepatocytes
US20070010008A1 (en) 2005-06-29 2007-01-11 Tissuetech, Inc. Ex vivo expansion of primary animal cells
WO2005040391A1 (en) 2003-10-27 2005-05-06 Murdoch Childrens Research Institute Compositions and methods for differentiating stem cells
US20070036769A9 (en) 2004-06-03 2007-02-15 Linheng Li BMP pathway methods and compositions
GB0413005D0 (en) 2004-06-11 2004-07-14 Coletica Ligand
US8492148B2 (en) 2005-02-23 2013-07-23 Foundation For Biomedical Research & Innovation Method for amplification of endothelial progenitor cell in vitro
AU2006202209B2 (en) 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
CA2621161A1 (en) 2005-09-08 2007-03-15 The Government Of The United States Of America As Represented By The Sec Retary Of The Department Of Health And Human Services Methods for promoting stem cell proliferation and survival
CA2643478C (en) 2006-02-23 2019-06-18 Novocell, Inc. Compositions and methods useful for culturing differentiable cells
US7989204B2 (en) 2006-04-28 2011-08-02 Viacyte, Inc. Hepatocyte lineage cells
US8628964B2 (en) 2006-10-11 2014-01-14 Drexel University Fetal pulmonary cells and uses thereof
US8088374B2 (en) 2006-10-20 2012-01-03 Deutsches Krebsforschungszentrum Stiftung Des Offenlichen Rechts Methods for inhibition of angiogenesis and vasculogenesis via rspondin antagonists
US9251637B2 (en) 2006-11-15 2016-02-02 Bank Of America Corporation Method and apparatus for using at least a portion of a one-time password as a dynamic card verification value
US20080182328A1 (en) 2006-12-19 2008-07-31 The Burnham Institute Mammalian extraembryonic endoderm cells and methods of isolation
WO2008101215A1 (en) 2007-02-16 2008-08-21 Varian Medical Systems Technologies, Inc. Preparative regimen for engraftment, growth and differentiation of non-hematopoeitic cells in vivo
EP2022848A1 (en) 2007-08-10 2009-02-11 Hubrecht Institut A method for identifying, expanding, and removing adult stem cells and cancer stem cells
JP2009195142A (en) 2008-02-20 2009-09-03 Institute Of Physical & Chemical Research Method for obtaining liver ovoid cell
US20110191868A1 (en) * 2008-04-10 2011-08-04 Massachusetts Institute Of Technology Methods for identification and use of agents targeting cancer stem cells
AU2009274517B2 (en) * 2008-07-25 2015-03-26 The University Of Georgia Research Foundation, Inc. Compositions for Mesoderm derived ISL1+ Multipotent cells (IMPs), epicardial progenitor cells (EPCs) and multipotent CXCR4+CD56+ cells (C56Cs) and methods of use
WO2010016766A2 (en) 2008-08-08 2010-02-11 Koninklijke Nederlandse Akademie Van Wetenschappen Antibodies recognizing endogenous human lgr5 and/or lgr6
US9464275B2 (en) 2008-08-21 2016-10-11 The Board Of Trustees Of The Leland Stanford Junior University Ex vivo culture, proliferation and expansion of intestinal epithelium
EP2412800A1 (en) 2010-07-29 2012-02-01 Koninklijke Nederlandse Akademie van Wetenschappen Liver organoid, uses thereof and culture method for obtaining them
CA2751332C (en) 2009-02-03 2024-04-02 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
US9752124B2 (en) 2009-02-03 2017-09-05 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium for epithelial stem cells and organoids comprising the stem cells
GB201111244D0 (en) 2011-06-30 2011-08-17 Konink Nl Akademie Van Wetenschappen Knaw Culture media for stem cells
US20120207744A1 (en) 2009-03-19 2012-08-16 Mendlein John D Reprogramming compositions and methods of using the same
SG175367A1 (en) 2009-04-27 2011-11-28 Viacyte Inc Small molecules supporting pluripotent cell growth and methods thereof
US9376664B2 (en) 2010-06-14 2016-06-28 The Scripps Research Institute Reprogramming of cells to a new fate
SG10201508118WA (en) 2010-09-30 2015-11-27 Agency Science Tech & Res Methods and reagents for detection and treatment of esophageal metaplasia
KR102482184B1 (en) 2010-12-22 2022-12-28 페이트 세러퓨틱스, 인코포레이티드 Cell culture platform for single cell sorting and enhanced reprogramming of iPSCs
GB201106395D0 (en) 2011-04-14 2011-06-01 Hubrecht Inst Compounds
EP2725901A4 (en) 2011-06-29 2014-10-29 Penn State Res Found Compositions, methods and kits for treating leukemia
US20130052729A1 (en) 2011-08-29 2013-02-28 Olivier Pourquie Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
CA2849011A1 (en) 2011-10-05 2013-04-11 Genentech, Inc. Methods of treating liver conditions using notch2 antagonists
GB201117469D0 (en) 2011-10-10 2011-11-23 Cell Guidance Systems Ltd Culture media for pluripotent stem cells
JP6238445B2 (en) 2011-10-27 2017-11-29 国立大学法人 東京医科歯科大学 Colorectal epithelial stem cell isolation and culture technology, and colorectal epithelial transplantation technology using this
US20140302491A1 (en) * 2011-10-28 2014-10-09 The Board Of Trustees Of The Leland Stanford Junior University Ex Vivo Culture, Proliferation and Expansion of Primary Tissue Organoids
MX347853B (en) 2011-12-19 2017-05-16 Umc Utrecht Holding Bv A rapid quantitative assay to measure cftr function in a primary intestinal culture model.
US20150284689A1 (en) * 2012-10-26 2015-10-08 The Regents Of The University Of California Strategy for engineering various 3d tissues, organoids and vasculature
EP2956538B1 (en) 2013-02-13 2018-11-14 Wake Forest University Health Sciences Bioengineered liver constructs and methods relating thereto
US20150368616A1 (en) 2013-02-14 2015-12-24 The Cleveland Clinic Foundation Methods for induction of cell fates from pluripotent cells
CN113913366A (en) 2013-02-18 2022-01-11 大学健康网络 Method for generating liver cells and bile duct cells from pluripotent stem cells
US10041046B2 (en) 2013-03-14 2018-08-07 Massachusetts Institute Of Technology Compositions and methods for epithelial stem cell expansion and culture
AU2014233055B2 (en) 2013-03-15 2018-08-09 Board Of Regents, The University Of Texas System Inhibition of pulmonary fibrosis with nutlin-3a and peptides
CN105324391A (en) 2013-04-16 2016-02-10 奥尔布森治疗学有限公司 Medical use of syndecan-2
AU2015261380B2 (en) 2014-05-16 2021-04-15 Koninklijke Nederlandse Akademie Van Wetenschappen Improved culture method for organoids
US11920164B2 (en) 2014-07-30 2024-03-05 Yeda Research And Development Co. Ltd. Media for culturing naive human pluripotent stem cells
CN107075471A (en) 2014-10-08 2017-08-18 新加坡科技研究局 Stem cell is divided into the method for hepatocyte lineage
GB201421092D0 (en) 2014-11-27 2015-01-14 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium
GB201421094D0 (en) 2014-11-27 2015-01-14 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium
GB201603569D0 (en) 2016-03-01 2016-04-13 Koninklijke Nederlandse Akademie Van Wetenschappen Improved differentiation method
GB201610748D0 (en) 2016-06-20 2016-08-03 Koninklijke Nederlandse Akademie Van Wetenschappen Improved diffrentation method

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023137465A1 (en) * 2022-01-13 2023-07-20 BIOMILQ, Inc. Optimization of live cell constructs for production of cultured milk product and methods of using the same

Also Published As

Publication number Publication date
ES2883159T3 (en) 2021-12-07
HK1245324A1 (en) 2018-08-24
DK3224351T3 (en) 2021-08-23
EP3224351B1 (en) 2021-06-30
EP3950937A1 (en) 2022-02-09
PT3224351T (en) 2021-09-03
AU2015352350A1 (en) 2017-06-15
US20170342385A1 (en) 2017-11-30
WO2016083612A1 (en) 2016-06-02
JP2022025124A (en) 2022-02-09
US10961511B2 (en) 2021-03-30
JP7261538B2 (en) 2023-04-20
CA2968751A1 (en) 2016-06-02
AU2015352350B2 (en) 2021-07-29
JP2017536827A (en) 2017-12-14
EP3224351A1 (en) 2017-10-04
GB201421094D0 (en) 2015-01-14

Similar Documents

Publication Publication Date Title
US20210254017A1 (en) Culture Medium for Expanding Breast Epithelial Stem Cells
US20220135952A1 (en) Culture Medium
AU2020200515B2 (en) Culture media for stem cells
US11725184B2 (en) Culture method for organoids
US9074182B2 (en) Differentiated pluripotent stem cell progeny depleted of extraneous phenotypes

Legal Events

Date Code Title Description
AS Assignment

Owner name: KONINKLIJKE NEDERLANDSE AKADEMIE VAN WETENSCHAPPEN, NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SACHS, LARS NORMAN;DROST, JARNO;GRACANIN, ANA;SIGNING DATES FROM 20181001 TO 20190926;REEL/FRAME:055862/0908

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED