Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHOD FOR DIFFERENTIATING EPITHELIAL STEM CELLS
Document Type and Number:
WIPO Patent Application WO/2021/028361
Kind Code:
A1
Abstract:
The subject matter of the present invent ion is a method for differentiating epithelia l stem cells, comprising culturing one or more epithelial stem cells in contact with an extracellular matrix in t he presence of an expansion medium, a bovine pituitary extract, a receptor tyrosine kinase ligand, a supernatant of primary fibroblasts and optionally, a Rho kinase inhibitor.

Inventors:
LLUEL PHILIPPE (FR)
VERGNOLLE NATHALIE (FR)
GAME XAVIER (FR)
ROUGET CÉLINE (FR)
SÉBERT MORGANE (FR)
Application Number:
PCT/EP2020/072318
Publication Date:
February 18, 2021
Filing Date:
August 07, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UROSPHERE (FR)
INST NAT SANTE RECH MED (FR)
UNIV DE TOULOUSE III PAUL SABATIER (FR)
ECOLE NAT VETERINAIRE DE TOULOUSE (FR)
CHU RANGUEIL (FR)
INSTITUT NATIONAL DE RECH POUR L’AGRICULTURE L’ALIMENTATION ET L’ENVIRONNEMENT (FR)
International Classes:
C12N5/071; C12N5/09; G01N33/50
Domestic Patent References:
WO2019006136A12019-01-03
WO2015156929A12015-10-15
WO2010090513A22010-08-12
WO2012014076A22012-02-02
WO2012168930A22012-12-13
Other References:
HARRY HORSLEY ET AL: "A urine-dependent human urothelial organoid offers a potential alternative to rodent models of infection", SCIENTIFIC REPORTS, vol. 8, no. 1, 19 January 2018 (2018-01-19), XP055664527, DOI: 10.1038/s41598-018-19690-7
IGOR VASYUTIN ET AL: "Bladder Organoids and Spheroids: Potential Tools for Normal and Diseased Tissue Modelling", ANTICANCER RESEARCH, vol. 39, no. 3, 1 March 2019 (2019-03-01), GR, pages 1105 - 1118, XP055664530, ISSN: 0250-7005, DOI: 10.21873/anticanres.13219
HAZEM ORABI ET AL: "Optimization of the current self-assembled urinary bladder model: Organ-specific stroma and smooth muscle inclusion", CANADIAN UROLOGICAL ASSOCIATION JOURNAL = JOURNAL DE L'ASSOCIATION DES UROLOGUES DU CANADA NOV 2007, vol. 9, no. 9-10, 9 September 2015 (2015-09-09), CANADA, pages 599, XP055664548, ISSN: 1911-6470, DOI: 10.5489/cuaj.2953
SANLI ET AL., NATURE REVIEWS DISEASE PRIMERS, vol. 3, 2017, pages 17022
BRAY ET AL., CA CANCER J CLIN, vol. 68, 2018, pages 394 - 424
REBILLARD ET AL., PROGRES EN UROLOGIE J. ASSOC FRANCAISE UROL, vol. 4, 2010, pages 211 - 214
MO LI ET AL., THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 380, no. 6, 2019, pages 569 - 579
INOUE ET AL., NATURE REVIEWS UROLOGY, 2017
BYRNE ET AL., NATURE REVIEWS IN CANCER, 2017
FATEHULLAH ET AL., NATURE CELL BIOLOGY, vol. 18, no. 3, 2016, pages 246 - 254
BABJUK ET AL., EUROPEAN UROLOGY, vol. 71, 2017, pages 447 - 461
LEOW ET AL., WORLD JOURNAL OF UROLOGY, 2019
YOSHIDA ET AL., INVESTIGATIVE AND CLINICAL UROLOGY, vol. 59, 2018, pages 149 - 151
LEE ET AL., CELL, vol. 173, 2018, pages 515 - 528
SOUTHGATE ET AL.: "Culture of Epithelial Cells", 2002, WILEY-LISS, INC.
ROUPRET ET AL., PROGRES EN UROLOGIE, vol. 28, 2018, pages S46 - S78
LANG H. ET AL., ONCOTARGET, vol. 7, no. 37, 2016, pages 59336 - 59359
Attorney, Agent or Firm:
ERNEST GUTMANN - YVES PLASSERAUD SAS (FR)
Download PDF:
Claims:
CLAIMS

1. A method for differentiating bladder epithelial stem cells, wherein said method comprises: culturing one or more epithelial stem cells in contact with an extracellular matrix in the presence of an expansion medium, the expansion medium comprising a basal medium for animal or human cells to which is added a bovine pituitary extract (BPE), a receptor tyrosine kinase ligand, a supernatant of bladder primary fibroblasts, and optionally, a Rho kinase inhibitor (rho-associated protein kinase inhibitor or ROCK inhibitor).

2. A method according to claim 1, wherein, in the expansion medium, the receptor tyrosine kinase ligand is selected from the group consisting of FGF, HGF and EGF.

3. A method according to claim 1 or 2, wherein, in the expansion medium, the receptor tyrosine kinase ligand is EGF.

4. The method according to any of claims 1 to 3, wherein, in the expansion medium, the ROCK inhibitor is selected from the group consisting of Fasudil, Ripasudil, Netarsudil, RKI-1447, Y-27632,

GSK429286A, C21H16F4N402 or Y-30141.

5. The method according to any of claims 1 to 4, wherein, in the expansion medium, the ROCK inhibitor is Y-27632.

6. The method according to any of claims 1 to 5, wherein the supernatant of bladder primary fibroblasts is a supernatant of bladder primary fibroblast originating from human bladder tissue.

7. The method of any one of the preceding claims, wherein the epithelial stem cells are from human bladder.

8. An expansion medium, comprising a basal medium for animal or human cells to which is added a bovine pituitary extract (BPE), a receptor tyrosine kinase ligand, a supernatant of bladder primary fibroblasts and optionally, a ROCK inhibitor.

9. The expansion medium of claim 8, wherein the ROCK inhibitor is selected from the group consisting of Fasudil, Ripasudil, Netarsudil, RKI-1447, Y-27632, GSK429286A, C21H16F4N402 or Y-30141. 10. The expansion medium of claim 9, wherein the ROCK inhibitor is Y-27632.

11. The expansion medium of any one of claims 8 to 10, wherein the receptor tyrosine kinase ligand is selected from the group consisting of FGF, HGF and EGF or their combinations.

12. The expansion medium of any one of claims 8 to 1 1, wherein the supernatant of primary fibroblasts is a supernatant of bladder fibroblasts.

13. An organoid obtainable or obtained by a method of any one of claims 1 to 7, wherein the organoid is derived from the bladder, especially from human bladder.

14. An organoid according to claim 13, wherein this organoid originates from non-papillary tumour.

15. An organoid obtainable or obtained by a method of any one of claims 1 to 7, wherein the organoid is derived from a human’ s bladder tumour grafted into the mouse.

16. Use of an organoid as defined in claim 13, or a cell derived from said organoid for drug discovery development; drug screen; target screen; biomarker screen; toxicity assay; gene expression studies including recombinant gene expression; research of mechanisms involved in tissue injury and repair; research of inflammatory and infectious diseases; research on the microbiota and associated dysbiosis and pathologies; studies of pathogenic mechanisms; or studies of mechanisms of cell transformation and aetiology of cancer; precision medicine.

17. An organoid according to claim 13 or 14, or a cell derived from said organoid, for use in treating a disorder, condition or disease.

18. An organoid according to claim 13, or a cell derived from said organoid, for use in treating bladder cancer, bladder inflammation, cystitis, overactive bladder, interstitial cystitis, chemical cystitis, neurogenic bladder, bladder outlet obstruction, underactive bladder or any bladder pathologies.

19. An organoid according to claim 13 or 14, or a cell derived from said organoid, for use in transplantation of the organoid or cell into a patient. 20. A method of diagnosing a disease in a patient, comprising obtaining epithelial cells from the patient and culturing said epithelial cells in vitro according to the method of claim 1 and testing said cultured cells or organoid for the disease.

21. A method of use of an organoid according to any of claim 1 to 7 for the selection of therapeutic compounds, consisting in obtaining a biopsy of bladder cells, culturing said cells in vitro according to the method of claim 1 and testing potential therapeutic molecules.

22. A method of use according to claim 21, for the inclusion of patients in clinical trials, based on the tested therapeutic molecules.

23. A method of preparation of an organoid culture of human bladder tissue, in particular of preparation of an organoid culture of normal bladder tissue or of cancer bladder tissue, comprising the steps of : a. Providing a suspension of cells comprising urothelial cells including epithelial adult stem cells originating from said human bladder tissue in a suspension medium which comprises a basal medium for culture of animal or human cells, a ROCK inhibitor and optionally bovine pituitary extract (BPE) and/or a receptor tyrosine kinase ligand selected among the group of EGF, FGF and HGF and wherein said suspension medium is devoid of supernatant of primary fibroblast and , b. Culturing the cells provided in step above ( a.) in an extracellular matrix and an expansion medium suitable for differentiation of said human epithelial adult stem cells, in conditions allowing cell expansion and cell differentiation of the epithelial adult stem cells originating from said human bladder tissue until a three dimensional tissue is obtained, wherein such step of culturing encompasses one or multiple passages or the cells, advantageously 2, 3, 4, or 5 passages, in particular up to 10 or up to 15 passages, c. Allowing the three dimensional tissue obtained in step above (b.) to grow as an organoid culture and optionally performing cellular and/or molecular characterization of the obtained organoids, wherein the expansion medium comprises at least a basal medium for culture of animal or human cells and a supernatant of primary fibroblast and wherein the composition of the expansion medium is adjusted during cell culture step as disclosed in b. so that in the each passage of the cell culture the expansion medium initially additionally comprises bovine pituitary extract (BPE) and/or a receptor tyrosine kinase ligand selected among the group of EGF, FGF and HGF and, optionally a ROCK inhibitor, when the expansion medium in said each passage is changed or replenished it is devoid of the ROCK inhibitor, in particular the expansion medium is as defined in any one of claims 1 to 12 for replenishment during successive cell passages.

24. The method according to claim 23, wherein the organoid culture is prepared from a bladder tissue previously obtained from a patient bladder tumour wherein the method comprises the following additional steps before first step (a.) of claim 23 : i. providing a sample of bladder tumour tissue of a patient and dissecting said tissue in order to recover urothelium separated from submucosa, and recovering a suspension enriched with urothelial cells in particular as a supernatant following agitation of the medium comprising the dissociated cells; ii. centrifugating the suspension enriched with urothelial cells of i. and recovering the centrifugation pellet that contains urothelial cells.

25. The method according to claim 23, wherein the organoid culture is prepared from a patient derived tumour xenograft (PDX) of a bladder and wherein the method comprises the following additional steps before first step (a.) of claim 23 : i. Providing bladder tumour tissue previously obtained from the xenograft, dissecting said bladder tumour tissue into pieces that may allow cells to be in contact with a dissociation solution, in particular bladder tumour tissue pieces of about 5mm3; ii. Optionally incubating the obtained bladder tumour tissue of i. with a stripping solution to break intercellular junctions between urothelial cells and submucosal cells and recovering a suspension enriched with urothelial cells in particular as a supernatant following agitation of the suspension comprising the dissociated cells; iii. centrifugating the suspension enriched with urothelial cells of ii. and recovering the centrifugation pellet that contains urothelial cells.

26. The method according to claim 23, wherein the organoid culture is prepared from a bladder tissue previously obtained from a healthy bladder tissue of a human subj ect wherein the method comprises the following additional steps before first step (a.) of claim 23 : i. Providing bladder tissue of a human subject and dissecting said tissue in order to recover urothelium separated from submucosa, and optionally incubating the bladder tumour tissue with a stripping solution to break intercellular junctions between urothelial cells and submucosal cells and recovering a suspension enriched with urothelial cells in particular as a supernatant following agitation of the suspension comprising the dissociated cells; ii. centrifugating the suspension enriched with urothelial cells of i. and recovering the centrifugation pellet that contains urothelial cells.

27. The method according to any one of claims 23 to 26 wherein in step a. the expansion medium contains 80% to 40% of volume of a basal medium or of a basal medium supplemented with BPE and receptor tyrosine kinase ligand and receptor tyrosine kinase ligand in particular complete KSFM medium and 20% to 60% of volume of primary fibroblasts supernatant (PFS), especially contains one volume of a basal medium or of a basal medium supplemented with BPE and receptor tyrosine kinase ligand, in particular of complete KSFM medium, and furthermore contains one volume of primary fibroblasts supernatant (PFS) and wherein in culture step b. during 2 to 3 days after the onset of the culture the expansion medium also comprises a ROCK inhibitor and during the subsequent cell culture a fresh expansion medium or a replenished medium is provided that comprises 80% to 40% of volume of a basal medium or of a basal medium supplemented with BPE and receptor tyrosine kinase ligand, in particular complete KSFM medium and 20% to 60% of volume of primary fibroblast supernatant (PFS), especially comprises one volume of a basal medium, in particular a KSFM medium, with one volume of primary fibroblast supernatant (PFS), and said fresh expansion medium or said replenished medium is devoid of a ROCK inhibitor.

28. The method according to any one of claims 23 to 27 wherein the extracellular matrix is provided to the cell suspension prior to the expansion medium and the extracellular matrix is allowed to polymerize in order to allow cells to be embedded therein prior to the addition of the expansion medium.

29. The method of any one of claims 23 to 28, wherein organoids in the culture show viability for at least 10 days, preferably at least 2 weeks, more preferably at least 25 weeks while being maintained in the extracellular matrix, such as matrigel, and in the culture medium.

30. The method according to any one of claims 23 to 29 wherein the organoid culture comprises organoids that have a size in the range of at least 30pm, preferably at least 40pm, more preferably at least 80pm and most preferably at least 200pm. 31. The method according to any one of claims 23 to 30 wherein organoids in the organoid culture express at least one, preferably at least 2 cellular markers selected in the group of CK5, CK17, CK20, GATA3, Upk3 and Ki67, in particular express CK5, CK17, CK20, GATA3, Upk3 and Ki67.

32. An organoid harbouring histological characteristics including cancer subtype and/or cancer stage and/or cytological shape characteristics of parental human bladder tumour tissue and genetic features such as STR fingerprinting of parental human bladder tumour tissue wherein said organoid is obtainable by carrying out the method of any one of claims 23 to 32 and said organoid has a size in the range of at least 30pm, preferably at least 40pm, more preferably at least 80pm and most preferably at least 200pm and expresses CK5, CK17, CK20, GATA3, Upk3 and Ki67 cellular markers.

Description:
METHOD FOR DIFFERENTIATING EPITHELIAL STEM CELLS

The subject matter of the present invention is a method for differentiating epithelial stem cells, comprising culturing one or more epithelial stem cells in contact with an extracellular matrix in the presence of an expansion medium, a bovine pituitary extract, a receptor tyrosine kinase ligand, a supernatant of primary fibroblasts, and optionally, a Rho kinase inhibitor.

The subject of the invention is also a method that further comprises obtaining and isolating an organoid.

In a preferred embodiment, the epithelial stem cells are human adult bladder epithelial stem cells and the supernatant of primary fibroblasts is a supernatant of bladder primary fibroblasts.

Another subject of the invention is the use of this organoid or a cell derived from said organoid, in a drug discovery development; drug screen; target screen; biomarker screen; toxicity assay; gene expression studies including recombinant gene expression; research of mechanisms involved in tissue injury and repair; research of inflammatory and infectious diseases; research on the microbiota and associated dysbiosis and pathologies; research on all epithelial dysfunction, studies of pathogenic mechanisms; or studies of mechanisms of cell transformation and aetiology of cancer and precision medicine.

Bladder related diseases are among the most common and costly diseases. In particular, Bladder cancer is the tenth most common cancer worldwide and the second most common urologic cancer after prostate cancer (GLOBOCAN 2018). Men are more affected than women (3.2:0.9 ratio) and disease incidence increases with age (Sanli et al ., Nature Reviews Disease Primers, 2017; vol 3, pl7022). Thus, the disease ranks higher among men, in whom it is the sixth most common cancer and ninth leading cause of cancer death (Bray et al., Ca Cancer J Clin 2018; 68:394-424). Its incidence is over increasing by 1% each year and yet mortality has remained stable over the past 30 years (Rebillard et al., Progres en Urologie J. Assoc Francaise Urol, 2010 S4: S211-214). Three main risk factors have been identified in the development of bladder tumours, including smoking, genetic factors, and environmental factors (Sanli et al ., 2017). To develop new treatments, the improvement and development of new models for the preclinical study of bladder urothelium is mandatory.

Currently used strategies for disease characterization and therapy development rely on 2D cell culture techniques with subsequent employment of animal models. The in vitro models available routinely for the study of the bladder cancer are essentially tumor cell lines grown in 2D. These tumor lines are easy to handle experimentally, are inexpensive, provide extensive information on cellular pathophysiology and have proven utility in preclinical pharmacology studies. However, these models of 2D tumor lines do not recreate tissue architecture and environmental diversity, nor the difficulties of therapeutic targeting found in vivo. In addition, these cell lines do not reflect tumor heterogeneity between patients and within the tumor itself. Indeed, the cell lines do not allow to faithfully reproduce the genetic and phenotypic diversity of primary tumours and do not fully summarize their complexity because the inflammatory components and stroma are not present in vitro (Mo Li et al, the New England Journal of Medicine, 2019; 380(6):569-579).

Animal models are one of the main tools of cancer research. There are currently three mouse models of bladder cancers: chemically induced tumor models, xenograft models and genetically engineered models (GEM) (Inoue et al., Nature Reviews Urology, 2017). Chemical carcinogens models of tumour and their temporal progression are highly heterogeneous whereas GEM models are relatively genetically homogeneous compared with the heterogeneity of human cancers with the same histology. Thus, these models do not accurately recapitulate the tumour. Conventional cell-line derived xenograft (CDX) models have provided valuable information regarding our understanding of cancer development and metastasis. They have been used for drugs screening and understanding of their mechanism of action. But they have major deficiencies as inappropriate microenvironments and the lack of tumor heterogeneity that are necessary for the study of tumour development and resistance. Patient-derived-xenograft models (PDX) are more indicated than CDX to study the tumour biology as they better reflect the tumour heterogeneity of individual cancers. More, treatment of PDXs reproduce clinical outcomes observed in the individual patient donors (Byrne et al ., Nature Reviews in Cancer, 2017). However, the animal pathophysiology makes delicate the clinical extrapolation of therapeutic tests. In addition, the high costs and the time required to use these models hinder the implementation of large-scale therapeutic tests covering the spectrum of vesical carcinomas in humans.

Other models of preclinical cancer studies are emerging, taking into account both the investigative flexibility of in vitro models and critical elements of in vivo physiopathology such as architecture and tissue microenvironment. These 3D tumor cell cultures contain spheroids and organoids. Among these cultures in 3D, the organoid model seems to be the most relevant for the study of cancers. Indeed, organoids can be developed from primary cells isolated from patients, thanks to the survival and self renewal properties of stem cells, and can differentiate in respect of hierarchy and tissue architecture (Fatehullah et al., Nature Cell Biology, 2016, 18(3):246-254). The organoids closely resemble the in vivo epithelial tissue from which they were obtained. They reproduce the complex spatial morphology of a differentiated epithelium to enable biologically relevant cell-cell and cell-matrix interactions. Ideally, the physical, cellular, and molecular characteristics of organoids mean that they share similar physiological responses with differentiated in vivo epithelia. Organoid culture was established for several tissue types: stomach, colon and pancreas and for prostate, saliva glands and several other tissues (Mo Li et al., the New England Journal of Medicine, 2019, 380(6):569-579).

Increasing evidence suggests that bladder epithelium is a hierarchically organized tissue containing specific stem cells (SCs) that participate in normal homeostasis and response in bladder aggression. Also, in bladder cancers, there would be, among others, cancer stem cells (CSCs), having the ability to self-renew and produce tumor cell heterogeneity by differentiating. Since bladder cancer stem cells are proposed as the drivers of initiation and progression of urothelial cancers, these cells are ideal targets for anticancer therapies. The organoid model would therefore be particularly suitable for the study of normal and cancerous bladder cancer and the development of new treatments. One of the expected perspectives is to define a new preclinical model opening the door of personalized medicine towards which current research tends with the possibility of in vitro tests for the prediction of therapeutic responses. The development of these models can also serve as a support for pathophysiological studies of vesical urothelial diseases (overactive bladder, interstitial cystitis, chemical cystitis, neurogenic bladder, bladder outlet obstruction, underactive bladder, bladder inflammation...) and cellular therapies. Bladder cancer generally originates from the epithelium (urothelium) of the bladder and is referred to as urothelial carcinoma. Non- Muscle Invasive Bladder Cancers that are confined to mucosa or have invaded the lamina propria (submucosa) are classified as Ta and Tl, respectively (Babjuk et al., European Urology, 2017, 71 :447-461). Carcinoma in situ is a flat, poorly differentiated tumour confined to mucosa. Stage 2 tumours have invaded the muscle layer either superficially (T2a) or deeply (T2b). T3 tumours have invaded beyond the bladder wall into perivesical fat (T3a invasion is microscopic, T3b is macroscopic). T4a tumours have invaded the prostate, uterus, vagina and/or bowel, whereas T4b tumours have invaded the pelvic or abdominal walls (Leow et al., World Journal of Urology, 2019).

Patient-derived organoids (PDOs) have recently emerged as robust preclinical models for their abilities to recapitulate patient drug responses in the clinic, and they might therefore be integrated into precision medicine programs. Bladder cancer organoids have been developed but most of the models obtained are from non-muscle invasive forms of bladder cancer called papillary tumours (with a success rate of 80%) whereas the success rate for preparing organoids from non-papillary forms or muscle invasive tumours is less than 30% (Yoshida et al., Investigative and clinical urology, 2018; 59: 149-151). More, it has been observed by Lee et al (cell, 2018; 173 :515- 528) that 64% of the organoid lines they developed switched molecular subtype from that of their parental tumour.

In the present invention, inventors have developed a protocol allowing the development of organoids from normal bladder and bladder cancer including muscle invasive bladder cancer (MIBC). Indeed, the inventors were able to determine that isolated urothelial stem cells needed a fibroblast secretion medium to be able to form organoid structures in a reproducible manner. It is, therefore, an object of the present invention to provide a method, allowing the culture of organoids in a reproducible and controlled manner either from a normal organ or from a pathogenic organ like a cancer organ. More precisely, an object of the present invention is to provide a method, allowing the culture of bladder organoids in a reproducible and controlled manner either from a normal bladder or a pathogenic bladder like a cancer bladder.

Here is provided a method to culture epithelial stem cells and to obtain organoids that show longer-lived maintenance, and are able to differentiate to all major differentiated cell lineages present in the corresponding in vivo tissue. The method is envisaged to be relevant to the culture of all epithelial cell types, in particular bladder cell type.

In particular, the invention provides a method for culturing epithelial stem cells, wherein said method comprises culturing one or more epithelial stem cells in contact with an extracellular matrix in the presence of an expansion medium as described herein.

In some embodiments, the method for culturing or differentiating epithelial stem cells further comprises isolating one or more adult stem cells or obtaining and isolating an organoid. In some embodiments, the epithelial stem cells are from the bladder.

The term organoid is interchangeably used herein with the expression organoid culture and shall be understood as relating to an organoid culture unless when otherwise specified that one organoid of the organoid culture is isolated, for example to be used or when it is used as a singled out biological product and accordingly isolated from the obtained culture, such as in a well of a plaque to carry out assays.

Also provided is a culture medium. In particular, the invention provides an expansion medium, comprising a basal medium for animal or human cells to which is added a bovine pituitary extract (BPE), a receptor tyrosine kinase ligand, a supernatant of primary fibroblasts, and optionally, a Rho kinase inhibitor (rho-associated protein kinase inhibitor or ROCK inhibitor).

In a preferred embodiment the method is directed to the culture or differentiation of human adult epithelial stem cells originating from bladder and the supernatant of primary fibroblasts is obtained from human bladder fibroblasts. Accordingly, in the following where primary fibroblasts are mentioned this should be understood as encompassing the embodiment where fibroblasts are from bladder fibroblasts, especially from human bladder fibroblasts.

The invention also relates to a method of preparation of an organoid culture, of a normal or a pathogenic organ, in particular a cancer organ, wherein the organ is human bladder.

The invention also provides an organoid obtainable by the method of culture of the invention or the method of preparation of an organoid culture. Uses of the organoids described herein and cells derived from the organoids are likewise provided. For example, the invention also provides the use of an organoid of the invention or a cell derived from said organoid in a drug discovery development; drug screen; target screen; biomarker screen; toxicity assay; gene expression studies including recombinant gene expression; research of mechanisms involved in tissue injury and repair; research of inflammatory and infectious diseases; research on the microbiota and associated dysbiosis and pathologies; research on all epithelial dysfunctions, studies of pathogenic mechanisms; or studies of mechanisms of cell transformation, aetiology of cancer and precision medicine. By precision medicine is meant a tool to determine the right treatment at the right moment for a patient but also the possibility to optimize patient recruitment for clinical trials by using an avatar that can be a PDX or an organoid, based on the latter responses to selected drugs or biomarkers expression.

The invention also provides an organoid of the invention, or a cell derived from said organoid, for use in treating a disorder, condition or disease.

The invention also provides an organoid of the invention, or a cell derived from said organoid for use in regenerative medicine, for example, wherein the use involves transplantation of the organoid or cell into a patient.

Methods for culturing epithelial stem cells from a variety of tissues have previously been described in W02010/090513, W02012/014076 and WO2012/168930. However, in the case of bladder, it is very difficult to obtain organoids from muscle invasive bladder cancer as well as reproducible organoids.

The bladder is an organ whose role is to store and evacuate urine excreted by the kidneys. It is subdivided into three main layers: the mucosa, the submucosa, and the muscle. The mucosa membrane is a layer of cells that lines the inside of the bladder. It is also called urothelium or transitional epithelium. The submucosa is a layer of connective tissue that separates the mucosa and the muscularis. It is also called Lamina Propria, sub-urothelial tissue or stroma. The muscularis consists of two layers of muscle tissue, extending to the outside of the submucosa. This third layer is composed by smooth muscle cells divided into muscular longitudinal and muscular external.

The urothelium is an epithelium composed of 3 cellular subtypes: superficial cells, intermediate cells and basal cells. These cell subtypes express different levels of cytokeratin (Ck 5, 7, 8, 10 and 20). The basal cells express a high level of cytokeratin 5 (Ck5), and Ckl7 while the luminal cells, also called umbrellas cells or superficial cells, express high levels of Ck7, Ck20 and Uroplakin 3. Ck 5, 17 and p63 are markers of basal and intermediate cells whereas Ck 7, 8, 18 and 20 as well as uroplakins are markers of cells in umbrella.

The urothelium in homeostatic conditions regenerates in 3 to 6 months but is able in case of aggression to regenerate quickly in a few days. The stem cells are located at the level of the basal cell layer with an estimated proportion of 9%.

The present inventors have surprisingly found that adding a supernatant of fibroblasts to the culture medium allows human epithelial stem cells to be cultured without any defect.

This enables a large number of cells and organoids 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. Similarly, it means that many cells would be available for use in transplants and that multiple patients may be transplanted with cells obtained from a useful donor. Culturing the cells in an expansion medium allows the cells to multiply whilst retaining their stem or progenitor cell phenotype. Organoids are formed comprising these stem or progenitor cells.

Accordingly, there is provided a method for culturing epithelial stem cells, wherein said method comprises culturing one or more epithelial stem cells in contact with an extracellular matrix in the presence of an expansion medium, the expansion medium comprising a basal medium for animal or human cells to which is added a bovine pituitary extract (BPE), a tyrosine kinase ligand, a supernatant of fibroblasts and optionally, a ROCK inhibitor.

As explained above, long-term culture of human cells, in particular bladder cells, was very difficult prior to the present invention. Now, using the present invention, it is possible to culture bladder cells to obtain reproducible organoids. The bladder organoid production is enabled by the presence in the culture of the bladder adult stem cells that are allowed to expand and to differentiate.

The bladder epithelial stem cells to be cultured in the expansion method may be obtained by any suitable method. Preferably they are obtained from a human bladder and so are human bladder epithelial stem cells. In another embodiment, they can also be obtained from patient -derived - xenografts (PDX) from bladder cancer.

The bladder cells for use in the methods of invention are isolated from a bladder biopsy or resection. In some embodiments, bladder cells are isolated by collagenase digestion, for example, as described in Southgate et al ., (Culture of Epithelial Cells, 2002, Second Edition, Wiley-Liss, Inc.).

The bladder fibroblasts may come from biopsy or a resection. The bladder fibroblasts may be obtained from a commercial source such as from Sciencell Research Laboratoires (USA) that provides human bladder stromal fibroblasts under reference 4330 and their fibroblast basal medium under reference 2301. The fibroblast may also be obtained from the ATCC under reference ATCC® PCS-420-013 in a fibroblast basal medium (under ref: ATCC® PCS-201-030) completed by a fibroblast growth kit-low serum (under ref: ATCC® PCS-201-041).

The epithelial stem cells of the invention are epithelial cells from epithelial tissue of bladder or urothelial tissues.

In the expansion medium of the invention, any suitable bovine pituitary extract (BPE) can be used. Preferably, the BPE is used at a concentration comprised between 20 and 100 ng/mL of medium. More preferably, the concentration of BPE in the medium is 50 ng/mL of medium.

Concerning the Receptor Tyrosine Kinase Ligands, growth factors may be used to stimulate urothelial cell proliferation and differentiation, including either Epidermal growth factor (EGF), fibroblast growth factor (FGF) or hepatocyte growth factor (HGF) or their combinations are suitable to be comprised in the expansion medium. Many receptor tyrosine kinase ligands are mitogenic growth factors. In some embodiments, the receptor tyrosine kinase ligand in the expansion medium is selected from the group consisting of: FGF, HGF and EGF. Preferably, the receptor tyrosine kinase ligand used is EGF. Any suitable EGF may be used, for example, EGF obtained from Peprotech. Alternatively, EGF is substituted with an alternative compound that activates the EGF receptor. For example, it is envisaged that IGF (Insulin-like growth factor) may be substituted for EGF.

During culturing of stem cells, said receptor tyrosine kinase ligand is preferably added to the culture medium when required, for example, daily or every other day.

The basal medium may be supplemented with 4 ng/ml to 10 ng/ml EGF. More preferably the basal medium may be supplemented with 5ng/ml of EGF.

ROCK inhibitor is selected from the group consisting of Fasudil, Ripasudil, Netarsudil, RKI-1447, Y-27632, GSK429286A, C21H16F4N402 or Y-30141. Preferably, the ROCK inhibitor is Y-27632. More preferably, the medium may be supplemented with 10 mM of a ROCK inhibitor, in particular by IOmM of Y-27632.

The supernatant of fibroblasts added is preferably a supernatant of bladder primary fibroblasts obtained after 3 days to 3 weeks of culture of the cells (such as cells obtainable under ref ATCC® PCS-420-013) in a fibroblast basal medium (such as available under ref: ATCC® PCS-201-030) completed by a fibroblast growth kit-low serum (such as available under ref: ATCC® PCS-201-041).

Usually, from 20 to 60%, in particular from 20% to 50% or from 30% to 60% or from 30% to 50% of supernatant of fibroblasts in volume with respect to the volume of the basal medium or to the basal medium that additionally comprises BPE and receptor tyrosine kinase ligand (so-called complete basal medium), comprised in the expansion medium (in particular complete KSFM medium) is added to the basal medium or complete basal medium (in particular to the complete KSFM medium). In a particular embodiment, 50% of supernatant of fibroblasts is added to 50% of complete KSFM medium.

Some embodiments of the invention comprise the use of expansion medium for around 8-50, for example, 10-50, 15-50, 20-50, 20-40 passages of the cells. For example, in some embodiments the cells may be split at a 1 : 1 to 1 :3 dilution every 7-10 days for 2, 3, 4, 5 or 6 or more months.

Such dilutions are needed to favor asymmetrical divisions of the stem cells and differentiated epithelial cells. Preferably, during asymmetrical divisions, the medium contains a ROCK inhibitor.

Preferably the cells are human cells. More preferably, the epithelial stem cells are human epithelial stem cells. However, culturing non-human mammalian epithelial stem cells is also envisaged.

In some embodiments, the stem cells are selected from bladder, pancreas, intestine (e.g. small intestine or colon), stomach, prostate, kidney, lung, breast, ovarian, salivary gland, hair follicle, skin, oesophagus and thyroid epithelial stem cells. In a preferred embodiment, the stem cells come from bladder cell.

As described above, the method for culturing epithelial stem cells comprises culturing one or more epithelial stem cells embedded 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 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 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 commercially available extracellular matrices are extracellular matrix proteins (Invitrogen) and basement membrane preparations from Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells (e.g. Matrigel™ (BD Biosciences) which comprises laminin, entactin, and collagen IV.).

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 addition, the presence of an ECM allowed culturing of three- dimensional tissue organoids, which could not be cultured in the absence of an ECM.

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 expansion culture medium is placed on top of the ECM. The expansion culture medium can then be removed and replenished as and when required. In some embodiments, the culture medium is replenished every 1 , 2 or 3 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 method comprises culturing the organoid or a population of epithelial stem cells for at least 2, 4, 6, 8, 10, 12, 14, 16, 18, 20 or 25 weeks.

The expansion culture medium advantageously induces the survival of stem cells and their asymmetrical division. During the phase where stem cell are embedded in the provided extracellular matrix and accordingly when these cells may be provided or found as isolated cells, in order to promote their survival, the expansion medium contains at least the complete basal medium (i.e. a medium that comprises BPE and a receptor tyrosine kinase ligand besides its usual components for cell culture) the supernatant of primary fibroblasts and a ROCK inhibitor such as Y27632. When the expansion medium is then changed or replenished in order to promote cell differentiation, Y27632 (or another ROCK inhibitor) is preferably omitted from the composition of the expansion medium.

The expansion medium preferably induces or promotes the survival and/or proliferation of cells during at least 1 to 2 weeks of culture. Proliferation can be assessed using techniques known in the art such as BrdU staining, Edu staining, Ki67 staining and the use of growth curves assay can be done as well as differentiation labelling assays. Morphological characterization of organoid cultures can also be assessed microscopically and with cell differentiation labelling assays.

Media used according to the invention are capable of expanding a population of stem cells to form organoids for at least 2, at least 4, at least 6, at least 8, or at least 10 passages under appropriate conditions. Preferably, at this stage, the expansion culture medium does not contain a ROCK inhibitor.

In a preferred embodiment, the epithelial stem cells to be cultured in the expansion method 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 tissue.

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.

The epithelial stem cells may be obtained by any suitable method. In some embodiments, cells are isolated by collagenase digestion. In some embodiments, collagenase digestion is performed on a tissue biopsy. 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. 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. Accordingly, the cells may be obtained from a tumour, if required.

In a further aspect, there is provided a method for obtaining an organoid comprising culturing epithelial stem cells in an expansion medium using a 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.

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.

The invention also relates to a method of preparation of an organoid culture of human bladder tissue, in particular of preparation of an organoid culture of normal bladder tissue or of cancer bladder tissue comprising the steps of: a. Providing a suspension of cells comprising urothelial cells including epithelial adult stem cells originating from said human bladder tissue in a suspension medium which comprises a basal medium for culture of animal or human cells, a ROCK inhibitor and optionally bovine pituitary extract (BPE) and/or a receptor tyrosine kinase ligand selected among the group of EGF, FGF and HGF and wherein said suspension medium is devoid of supernatant of primary fibroblast, b. Culturing the cells provided in a. in an extracellular matrix and an expansion medium suitable for differentiation of said human epithelial adult stem cells, in conditions allowing cell expansion and cell differentiation of the epithelial adult stem cells originating from said human bladder tissue until a three dimensional tissue is obtained, wherein such step of culturing encompasses one or multiple passages or the cells, advantageously 2, 3, 4, or 5 passages, c. Allowing the three dimensional tissue obtained in b. to grow as an organoid culture and optionally performing cellular and/or molecular characterization of the obtained organoids, wherein the expansion medium comprises at least a basal medium for culture of animal or human cells and a supernatant of primary fibroblast and wherein the composition of the expansion medium is adjusted during cell culture of step b. so that in each passage of the cell culture the expansion medium initially additionally comprises a ROCK inhibitor and bovine pituitary extract (BPE) and/or a receptor tyrosine kinase ligand selected among the group of EGF, FGF and HGF and, when the expansion medium is changed or replenished in said each passage it is devoid of the ROCK inhibitor.

In a particular embodiment, the expansion medium is as defined in any one of the embodiments and examples of the culture medium disclosed herein for initiation or for replenishment during successive cell passages.

The terms “initially” or “initiation” above refer to the first period of time of each of the cell passage, in particular the first half of the duration of each passage, or preferably the first third of the duration of each passage. A passage usually last 9 to 15 days and the expansion medium is changed or replenished when necessary to promote the survival and differentiation of the cells, in particular is changed or replenished every 2 to 3 days. Accordingly, the ROCK inhibitor may be present in the expansion medium for 2 to 3 days at the beginning of each passage.

The number of cell passages may be adapted according to the cell expansion level and to the cell differentiation activity, to reach a size and maturation of the three-dimensional tissue that enables the use of the obtained organoid culture in screening assays, in particular in screening of drugs or drug candidates.

Using a bladder sample in order to prepare a cell suspension according to step a. above, may require a step of dissection of the sample to eliminate adipose and connective tissue in order to retain, in a first stage, the urothelium and the submucosa. Then a separation is performed to break the intercellular junctions between the urothelial cells and the submucosal cells to dissociate the urothelial cells from the other cells. The obtained urothelial cells may then undergo an enrichment step before being provided as a suspension in order to perform the culture steps.

Culturing of the urothelial cells involves several passages of the cells in culture medium that may have a different composition according to the passages. In particular it is pointed out that ROCK inhibitor (such as Y- 27632) may be one of the variable for adjustment of the composition of the expansion medium such ROCK inhibitor being present to promote the survival of the cells and/or to carry out the early step of the proliferation of the cells and of their asymmetric division.

According to a particular embodiment of this method, the organoid culture is prepared from a bladder tissue previously obtained from a patient bladder tumor wherein the method comprises the following additional steps before step a. : i. Providing a sample of bladder tumor tissue of a patient and dissecting said tissue in order to recover urothelium separated from submucosa, and recovering a suspension of cells comprising urothelial cells in particular as a supernatant following agitation of the suspension comprising the dissociated cells; ii. centrifugating the suspension of cells of i. and recovering the centrifugation pellet that contains urothelial cells.

The step of dissection of the bladder tissue recited herein encompasses a step of cutting the bladder tissue sample in pieces such as pieces of a few mm3 and treating them with a dissociation solution allowing enzymatic digestion of the tissue sample, especially collagenase digestion, in order to provide cells that may be processed according to the methods of the invention. The dissection step may in particular allow breaking intercellular junctions between urothelial cells and submucosal cells the dissection step may optionally comprise treating the sample with a stripping solution to help breaking intercellular junctions between urothelial cells and submucosal cells.

According to a particular embodiment of this method, the organoid culture is prepared from a patient derived tumor xenograft (PDX) of a bladder and the method comprises the following additional steps before the first step in the method of preparation of an organoid culture of human bladder tissue (step a. above): i. Providing bladder tumor tissue previously obtained from the xenograft, dissecting said bladder tumor tissue into pieces that may allow cells to be in contact with a dissociation solution, in particular bladder tumor tissue pieces of about 5mm3; ii. optionally incubating the obtained dissected bladder tumor tissue of i. with a stripping solution to break intercellular junctions between urothelial cells and submucosal cells and recovering a suspension enriched with urothelial cells in particular as a supernatant following agitation of the suspension comprising the dissociated cells; iii. centrifugating the suspension enriched with urothelial cells of ii. and recovering the centrifugation pellet that contains urothelial cells.

The dissociation solution and the stripping solution are solutions known from the person skilled in the art in order to especially break intercellular junctions between the cells of the treated tissue. Dissociation solutions and the stripping solution may be obtained as disclosed in Southgate et ak, 2002, Culture of Epithelial Cells, Second Edition. The dissociation solution comprises in particular enzymes for digestion, such as collagenase, especially collagenase IV.

According to a particular embodiment of this method, the organoid culture is prepared from a bladder tissue previously obtained from a healthy bladder tissue of a human subject wherein the method comprises the following additional steps before the first step in the method of preparation of an organoid culture of human bladder tissue (step a. above): i. Providing bladder tissue of a human subj ect and dissecting said tissue in order to recover urothelium separated from submucosa, and optionally incubating the bladder tumor tissue with a stripping solution to break intercellular junctions between urothelial cells and submucosal cells and recovering a suspension enriched with urothelial cells in particular as a supernatant following agitation of the suspension comprising the dissociated cells; ii. centrifugating the suspension enriched with urothelial cells of i. and recovering the centrifugation pellet that contains urothelial cells; According to a particular embodiment of the method of preparing the organoid culture according to the invention, in step a. of providing a suspension of cells, the expansion medium contains 80% to 40% of volume of a basal medium or of a basal medium supplemented with BPE and receptor tyrosine kinase ligand (such as complete KSFM medium) and 20% to 60% of volume of primary fibroblasts supernatant (PFS), for example contains one volume of a basal medium or of a basal medium supplemented with BPE and receptor tyrosine kinase ligand, in particular of complete KSFM medium, and furthermore contains one volume of primary fibroblasts supernatant (PFS) and, in culturing step b. during 2 to 3 days after the onset of the culture the expansion medium also comprises a ROCK inhibitor whereas during the subsequent culture stages in each passage a fresh expansion medium or a replenished expansion medium is provided that comprises 80% to 40% of volume of a basal medium or of a basal medium supplemented with BPE and receptor tyrosine kinase ligand, and 20% to 60% of volume of primary fibroblasts supernatant (PFS), for example one volume of a basal medium, in particular a complete KSFM medium, with one volume of primary fibroblasts supernatant (PFS) and said fresh expansion medium or a replenished expansion medium is devoid of a ROCK inhibitor.

ROCK inhibitor for use according to the invention is provided to prevent or to compensate stress of isolated cells that are present when embedding the suspension of cells in the extracellular matrix for the culture or when switching to a further passage and accordingly to enable such cells to undergo asymmetric division toward differentiation.

According to a particular embodiment of the method of preparing the organoid culture, the extracellular matrix is provided to the cell suspension in culturing step b. prior to providing the expansion medium and the extracellular matrix is allowed to polymerize in order to allow cells to be embedded therein prior to the addition of the expansion medium.

According to a particular embodiment of the method of preparing the organoid culture, the culture is performed at a temperature of 37°C in an incubator comprising 5% C02 according to conditions well known from the person skilled in the art and the number of passages of the cells is at least 2 or 3, in particular is up to 5 or up to 10 or up to 15 passages According to a particular embodiment of the method or preparing the organoid culture, the method enables production of organoids in the culture that show viability and stability for at least 10 days, preferably at least 2 weeks, more preferably at least 25 weeks while being maintained in the extracellular matrix, such as matrigel, and in the culture medium. More generally it is indicated that organoid cultures obtained after more than 10 especially more than 15 passages may have to be analyzed before use in order to confirm their stability in particular their genetic or proteomics stability.

According to a particular embodiment of the method of preparing the organoid culture the obtained organoid culture comprises organoids that have a size in the range of at least 30pm, preferably at least 40pm, more preferably at least 80pm and most preferably at least 200pm.

In a further embodiment of the method of preparing the organoid culture at least one of the obtained organoids expresses at least one, preferably at least 2 cellular markers selected in the group of CK5, CK17, CK20, GATA3, Upk3 and Ki67, in particular express CK5, CK17, CK20, GATA3, Upk3 and Ki67. Because GATA3, CK20 and Upk3 cellular markers are specific for cells of the luminal compartment of the bladder, their expression on cells of the organoids provides evidence of the advanced stage of maturation of the organoid, especially provides evidence that the organoid is fully differentiated. CK5 and CK17 are cellular markers for the basal cells of the bladder and Ki67 is a cellular marker for cell proliferation.

The organoid culture obtained according to the invention may be cryopreserved for storage before use.

In a preferred embodiment, a cancer organoid obtained from cancer stem cells is grown in a culture medium that is suitable for growth of the corresponding normal tissue organoid obtained from normal 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 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. An organoid is therefore obtainable or obtained by the method of the invention. Preferably, this organoid is derived from the bladder. More preferably, this organoid originates from non-papillary tumour.

Alternatively, an organoid is obtainable or obtained by the method of the invention is derived from a human’ s bladder tumour grafted into a mouse, preferably humanized, previously engrafted with bladder cancer cells from a human and containing a tumor formed from the bladder cancer cells.

It is to be understood that in a preferred expansion organoid, the majority of cells are expanding cells (i.e. dividing cells) that retain an undifferentiated phenotype. Image analysis may be used to assess characteristics of cells in culture such as cell morphology; cell structures 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.

An organoid of the invention preferably has a three-dimensional structure, i.e. the organoid is preferably a three-dimensional organoid. Morphologically, the cells appear like their corresponding in vivo tissue counterpart. The organoid or population of epithelial stem cells may be from any mammalian tissue. In some embodiments, it is from a mouse, rabbit, rat, guinea pig or other non-human mammal. Preferably, the organoid appears like a bladder tissue, more preferably, the cells are human cells.

Within the context of the invention, a tissue fragment is a part of an adult tissue, preferably a human adult tissue, more preferably, a bladder tissue. Preferably an organoid as identified herein is therefore not a tissue fragment.

The invention in particular relates to an organoid harboring histological (including cancer subtype and/or stage and cytological shape) characteristics of parental human bladder tumor and/or genetic features (such as Short Tandem Repeat fingerprinting as disclosed in Table 2) of parental human bladder tumor tissue wherein said organoid is obtainable by carrying out the method of any one of the herein disclosed embodiments and said organoid has a size in the range of at least 30pm, preferably at least 40pm, more preferably at least 80pm and most preferably at least 200pm and expresses at least 2 cellular markers selected in the group of CK5, CK17, CK20, GATA3, Upk3 and Ki67 cellular markers, in particular expresses at least one of CK20, GATA3 and Upk3 markers and at least one of CK5 and CK17 markers.

In a particular embodiment, the organoid originates from non- papillary tumour.

Illustrative examples of organoids generated using the differentiation medium and methods of the invention are given in the accompanying figures. The method of the invention may comprise changing the expansion medium for fresh medium during the course of the culturing because the components of the medium are used up during culturing. It will be clear to the skilled person how often the medium needs to be changed for fresh medium. In some embodiments, the medium is changed every other day, but it is also envisaged that it may be changed every day or every two days or as required.

The methods for culturing epithelial stem cells and/or obtaining an organoid in expansion or differentiation medium are carried out in vitro.

The invention provides an expansion medium, comprising a basal medium for animal or human cells to which is added a bovine pituitary extract (BPE), a receptor tyrosine kinase ligand, a supernatant of primary fibroblasts and optionally a ROCK inhibitor.

Preferably, in the expansion medium of the present invention, the ROCK inhibitor is selected from the group consisting of Fasudil, Ripasudil, Netarsudil, RKI-1447, Y-27632, GSK429286A, C21H16F4N402 or Y-30141. More preferably, the ROCK inhibitor is Y-27632.

Preferably, in the expansion medium, the receptor tyrosine kinase ligand is selected from the group consisting of: a growth factor such as one selected from FGF, HGF and EGF and more preferably is EGF or is selected among LY2156299, PGE2, SB202190, Gastrin, Wnt3a, etc.

The expansion medium of the invention contains the supernatant of primary fibroblasts. Preferably, the supernatant is a supernatant of bladder fibroblasts, especially of human bladder fibroblasts.

The expansion medium used in the method of the invention comprises any suitable basal medium. A basal medium for use in the invention will generally comprises a nutrient solution comprising standard cell culture ingredients, such as amino acids, vitamins, lipid supplements, inorganic salts, a carbon energy source, and a buffer, as described in more detail in the literature and below. In some embodiments, the culture medium is further supplemented with one or more standard cell culture ingredient, for example selected from amino acids, vitamins, lipid supplements, inorganic salts, a carbon energy source, and a buffer. The skilled person will understand from common general knowledge the types of culture media that might be used as the basal medium in the cell culture mediums of the invention. Potentially 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 F 12, Advanced DMEM/Ham's F 12, Iscove's Modified Dulbecco's Media and Minimal Essential Media (MEM), Ham's F-10, Ham's F-12, Medium 199, RPMI 1640 Media and KSFM Media.

The invention provides a composition comprising an expansion medium of the invention, an extracellular matrix and epithelial stem cells of the invention. The invention also provides a composition comprising an expansion medium of the invention, an extracellular matrix and one or more organoids of the invention.

The organoids of the invention are used in drug discovery development; drug screen; target screen; biomarker screen; toxicity assay; gene expression studies including recombinant gene expression; research of mechanisms involved in tissue injury and repair; research of inflammatory and infectious diseases; research on the microbiota and associated dysbiosis and pathologies; studies of pathogenic mechanisms; or studies of mechanisms of cell transformation and aetiology of cancer; precision medicine and/or as ex vivo cell/organ models, such as disease models.

The organoids of the invention or a cell derived from said organoids can be used to treat a disorder, condition or disease. In particular, these organoids or a cell derived from these organoids are useful to treat bladder cancer, bladder inflammation, cystitis, overactive bladder, interstitial cystitis, chemical cystitis, neurogenic bladder, bladder outlet obstruction, underactive bladder or any bladder pathologies.

The organoids of the invention or a cell derived from said organoids are useful for its use in transplantation of the organoid or cell into a patient.

The organoids of the invention can also be grafted in animal 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 of the invention can be used as ex vivo disease models.

Organoids of the invention are also 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 and functional diseases.

The ability to obtain a useful organoid of the invention in short time periods shows that the organoid is highly useful for testing individual patient responses to specific drugs and tailoring treatment according to the responsiveness. Wherein the organoid is obtained from a biopsy or resection from a patient, the organoid is cultured for less than 21 days.

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.

Therefore, the present invention encompasses a method of diagnosing a disease in a patient, comprising obtaining epithelial cells from the patient and culturing said epithelial cells in vitro according to the present invention and testing said cultured cells or organoid for the disease.

Moreover, there is provided a method of use of an organoid of the invention for the selection of therapeutic compounds (specific drugs) for a patient. This method comprises the steps of obtaining a biopsy of bladder cells of a patient, culturing said cells in vitro according to the present invention and testing potential therapeutic molecules for this patient. This method of selection of therapeutic compounds allows the inclusion of patients in clinical trials, based on the tested therapeutic molecules.

The methods of the present invention allow a great number of organoids and epithelial stem cells to be generated in a short period of time, thereby ensuring that sufficient cells are available for use in the application of interest.

The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.

Other subjects will emerge from reading of the description, the examples and the figures which follow.

Figures 1 and 2: describe cultures of healthy bladder organoids in presence of different culture media a) Medium KSFM + BPE + hEGF lOx, b) medium KSFM + BPE + hEGF + fibroblast supernatant, 20x.

Figure 3 : describes tumoural organoids, lOx. a) bilobed organoids, b) organoids in cluster.

Figures 4 and 5 : show a morphological comparison between healthy c) and tumoural d) organoids at D15, magnitude lOx.

Figure 6 describes the IF characterization of organoids from a healthy bladder 1 actin, 2 Ck5, 3 Ckl7, 4 Ck20, 5 UPK3A and 6 Dapi.

Figures 7 to 10: show immunofluorescence performed on BLHOU-40 tissues, 1) healthy tissue, 2) cancer tissue.

Figure 11 : L987 PDX model histological analysis. The analysis of the PDX model was performed in comparison of the patient’ s tumour. The following characteristics were observed

+ Cancer type and subtype: High grade epidermoid urothelial carcinoma + high grade papillary carcinoma; recurrence of a high grade (G3) urothelial carcinoma

+ Tumor stage: pTNM : pT4a + Basal-like phenotype + FGFR3 Mutation : R248C + PPARg mutation : no mutation Figure 12: Pharmacology assay in L987. The evaluation was performed using erlotinib (30 mg/kg) or a vehicle (10 mg/kg) administered per os to mice engrafted with the PDX. No antitumor response of L987 is shown.

Figure 13 : Pharmacology assay in L987. The evaluation was performed using erlotinib (lOOmg/kg) administered per os or with a vehicle (10 mg/kg) administered through intraperitoneal route. An antitumor response is obtained with erlotinib with tumour growth inhibition (TGI) at a level of 62,4% when compared to control.

Figure 14: Study Design for a screening assay (cell viability) on cancer organoid model from PDX

The assay is suitable to establish a proof of concept for using organoid model as a preclinical tool

Read-outs:

Immunofluorescence CK5, CK17, CK20, GATA3, Upk3 and Ki67

- Viability assay reflected by metabolic activity (concentration of ATP by luminescence) in organoids culture: using the kit Cell Titer Glo 3D (Promega).

Pharmacological treatment (triplicates) - 2 Runs Treatment for 5 days

Compound : Erlotinib (Run 1 : 0.04 - 0.2 - 1 - 5 - 10 - 20 mM; Run 2: 0.02-0.04-0.1-0.2-0.5-1 pM)

- Vehicle: DMSO 2%

Figure 15 : Cellular composition of the organoid line BLOU-026: Immunofluorescence staining with the specific marker Uroplakin 3 : Upk3 labeling (orange), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

One confocal stack in the core of one selected organoid from representative z- stack slice showing Upk3 expression.

A- Maximal intensity projection of all z-stack slices

B- One confocal stack in the core of one selected organoid: Representative z- stack slice showing entire structure (nuclei and actin)

C- Merged representation for Upk3, actin and nuclei labeling.

Scale Bar=100pm

Figure 16: Cellular composition of the organoid line BLOU-026: Immunofluorescence staining with the specific luminal marker: GATA3 labeling (red), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

A- One confocal stack in the core of one selected organoid: Representative z- stack slice showing GATA3 expression. Maximal intensity projection of all z-stack slices

B- Merged representation for GATA3, actin and nuclei labeling.

Scale Bar=100pm

Figure 17: Cellular composition of the organoid line BLOU-026: Immunofluorescence staining with the specific markers CK20 (luminal) and CK17 (basal): CK20 labeling (orange), CK17 labelling (red), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

A- One confocal stack in the core of one selected organoid: Representative z- stack slice showing nuclei.

B- One confocal stack in the core of one selected organoid: Representative z- stack slice showing actin labeling.

C- One confocal stack in the core of one selected organoid: Representative z- stack slice showing CK20 expression.

D- One confocal stack in the core of one selected organoid: Representative z- stack slice showing CK17 expression.

E- Merged representation for CK20, CK17, actin and nuclei labeling.

Scale Bar=100pm

Figure 18: Cellular composition of the organoid line BLOU-026: Immunofluorescence staining with the specific marker CK5 (basal) and the proliferation marker Ki67: CK5 labeling (red), Ki67 labelling (orange), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

Scale Bar=100pm

Figure 19: pharmacological assay using the BLOU-026 line Treatment with erlotinib was performed during 5 days - Erlotinib was administered in DMSO 2% as a vehicle and the doses used were as follows: Erlotinib (Run 1 : 0.04 - 0.2 - 1 - 5 - 10 - 20 mM; Run 2: 0.02-0.04-0.1-0.2-0.5-1 pM).

Drug response of organoid line (viability test by reference to ATP activity) is shown on the figure. Dose response curves for BLOU-026 organoids treated with the EGFR inhibitor erlotinib. Each data point corresponds to three biological replicates; error bars correspond to one standard error to the mean.

Example 1 : Studied Population

Organoid cultures of human bladder were performed from bladder specimens collected intraoperatively during cystectomy for urothelial bladder cancer of human patients from 50 to 70 years old.

After extraction of the surgical specimen, a vertical incision was made on the anterior surface of the bladder and two samples were taken: One in a macroscopically "healthy" zone (control group - culture A) and one in a macroscopically "tumoral" zone (tumor organoids - culture B). Two tumors were classified as muscle invasive (at least pT2 or more) and poorly differentiated (high grade) and two were non-invasive (pTl) after cystectomy and anatomo-pathological analysis of the tumour (according to the 1973 and

2004 WHO grading classification).

MVAC = Methotrexate, Vinblastin, Adriamicin, Cisplatin

BCG = Bacille of Calmette and Guerin (0 = no instillation / 1 = a series of 6 instillations in the bladder)

CIS = cisplatine

TR = Transuretral Resection of the bladder

Example 2 Obtention of bladder cells The bladder samples were dissected to eliminate adipose and connective tissue and to retain the urothelium and the submucosa. A "stripping" solution (Southgate et al., 2002, Culture of Epithelial Cells, Second Edition) is injected into the submucosa. The urothelium is incubated in the "stripping solution" at 4 °C overnight, with gentle shaking to break the intercellular junctions. The next day, the urothelium is removed from the muscularis / stroma using dissecting forceps and placed in collagenase IV solution (100U / ml) for 20 minutes at 37 °C. Then, to stop the dissociation, 3ml of KSFM medium (Keratinocyte Serum-Free Medium - Thermo Fischer) supplemented with Y27632 (10 mM - Sigma Aldrich, France) is added. Following strong manual agitation, the supernatant enriched in urothelial cells is centrifuged at 250 x g for 5 minutes at 4 ° C. The supernatant is discarded and the pellet containing urothelial cells resuspended in KSFM supplemented medium containing 50 ng/mL of BPE and 5ng/mL of EGF. After centrifugation and suspension in a volume of Matrigel (Matrigel hESC- qualified Matrix, BD Biosciences) at 4 °C, a concentration of 2 x 10 6 cells / mL of Matrigel is obtained.

Example 3 : Culture cells in slides

8-well Lab-teks (Lab-tek II Chamber Slides system, Dutscher) or 96- well m-angiogenesis-Ibidi plates are used (60 000 cells in 25 pL of Matrigel for 8-well labteck and 25 000 cells seeded in 10 pL of Matrigel for 96-well plate). Matrigel is polymerized for 20 minutes at 37 °C and then the complete medium is added (one volume of complete KSFM medium and one volume of primary fibroblast supernatant (PFS) isolated from the culture of human bladder fibroblasts). The supernatant of fibroblasts is obtained after 3 to 4 days of culture of the fibroblasts (ATCC- PCS-420-013) in a fibroblast basal medium (ATCC- PCS-201-030) supplemented with a fibroblast growth kit- low serum (ATCC- PCS-201-041). During the first 3 days of culture, this complete expansion medium is supplemented with Y27632 (10 mM). The medium is changed every 2-3 days with fresh expansion medium which corresponds to the complete medium without Y27632 (250 pL / well in the 48 well plates and labtek, 100 pL / well in the 96-well plates). Plates are incubated at 37 °C in normoxia condition (5% C02). Example 4: Morphological studies of “healthy” organoids a) Comparison of culture media:

Culture wells of healthy control cells (A cultures) are made with and without PFS in the culture. Then, the cells are monitored and compared at day 14.

A larger number of structures as well as a substantially greater diameter for the organoids subjected to a culture medium supplemented with PFS are observed at D14 (FIG. 2) (approximately 40 organoids per well of 80 pm against a dozen structures of 20 microns in diameter with the KSFMc (Fig 1)). This was observed with all the cultures of organoids performed.

The culture medium supplemented with primary fibroblast supernatant (PFS) thus seemed to promote the proliferation and growth of organoids compared to the KSFM culture medium complemented with EGF and BPE alone. b) Growth monitoring of organoids:

A total of 6 cultures of healthy organoids were carried out with the culture medium supplemented by PFS. We then compared the median organoid areas (solid structures) for these cultures at 14 days. There was a significant increase in the median organoid area on Day 14 (p = 0.0013).

Example 5 : Comparative study of the morphology of organoids from healthy and tumour bladders.

The growth and morphology of organoids derived from patients with healthy control organoids (A cultures) or bladder cancer (B cultures) were monitored at day 7.

The observations were made under a wide-field microscope at lOx magnification (Apotome Zeiss, lOx objective). When acquiring images, the choice of fields was randomly made. The size of the organoids has been evaluated using the image processing software Image J.

In tumor cultures, solid organoids classically observed in healthy cultures but also new morphologies were found. Indeed, the new solid structures for tumor cultures can be classified as full multilobed organoids, "bilobed" (Figure 3a) or "in clusters" (Figure 3b). These different forms have never been observed in healthy cultures. The size of the tumor organoids appeared to be higher than the healthy organoids, as shown between the healthy (A) and tumor (B) cultures (Figures 4 and 5). The comparison was made measuring 340 organoids, 170 healthy and 170 tumours.

Example 6: Immunolabeling of organoids

Immunolabeling on tissue sections

Immunolabeling is performed on frozen bladder sections. Fresh samples were included in OCT (Optimal Cutting Temperature), cut longitudinally, and immediately frozen on dry ice and stored at -80 °C. They are then cut with cryostat in 5 pm sections, deposited on slides (Superfrost Plus, Thermo Scientific) and fixed with 3.7% formaldehyde for 20 minutes at room temperature or with 50% acetone / 50% methanol until evaporation, depending on the antibody used. Blocking of nonspecific antigenic sites and permeabilization were achieved by a solution containing PBS / 1% BSA / 0.5% Triton X100. Samples are incubated with diluted primary antibodies in blocking buffer overnight at 4 °C in a humid chamber. After washing with PBS without calcium / magnesium IX, they are incubated for 1 hour with the secondary antibodies in the dark, at room temperature. The different antibodies used are summarized in Table 1 below. DAPI (4',6-diamidino-2- phenylindole) nuclei labeling (diluted 1/200, incubated 20 min at room temperature) is performed and the slides are mounted with a drop of Prolong gold mounting medium and a coverslip.

Table 1 : Antibodies used for immunolabeling

- Immunolabeling on organoids

Organoids are fixed at culture day 7 or 14 with 3.7% formaldehyde for 40 minutes (labteks) or 20 minutes (96-well) at room temperature. They are permeabilized with 0.5% Triton (in PBS) and the aspecific antigenic sites are blocked for 1.5 hours at 37 °C with a 3% BSA solution in PBS. Then the organoids are incubated with the primary antibodies at 4°C overnight followed by one hour at 37 °C. After washes, the secondary antibodies are incubated for one hour at 37 °C in the dark. DAPI nuclei labeling (dilution l/200e) and actin labeling by Alexa Fluor-488 phalloidin (diluted 1 : 500 in

DAPI solution) is performed. After incubation for 20 minutes in the dark at room temperature, washes are performed and a drop of Vectashield mounting medium (Vector Labs) without DAPI is deposited in each well.

The organoids and slides were observed using the Zeiss LSM 710 confocal microscope with 20x and 40x objectives. For each experiment, sections or control wells are imaged either without any antibodies (control of autofluorescence) and with the secondary antibodies alone. The control images were acquired with the same microscopy parameters and processed identically using the Image J software. The organoids were also imaged with an automated imaging device

(Opera Phenix, Perkin Elmer) with 20x water objectives. The images were processed with the Harmony software that drives the device. Example 7: Characterization of healthy organoids by immunofluorescence (IF)

These structures were observed using a Zeiss LSM 710 confocal microscope at day 14. The epithelial markers Ck5 and Ckl7 are physiologically expressed in undifferentiated basal cells. Ck20 is a predominant epithelial marker on superficial, differentiated cells. There was then a peripheral disposition of undifferentiated cell markers with a predominance of Ck20 at the center of the structures. In addition, Uroplakin- 3A spots are visualized in the center of the structures. Uroplakin-3A (UPK3A) is a protein that plays a major role in urothelial barrier function and is therefore characteristic of differentiated cells. Cell differentiation therefore seems hierarchical, occurring from the outside to the inside of these organoids (see Figure 6).

Example 8: Characterization of tumor tissues and organoids by immunofluorescence

We performed labeling on sections of healthy bladders and tumours with different urothelial markers (Figures 7 and 8). We found a similar distribution of these markers on samples A and B. Indeed, Ckl7 was found preferentially at the level of the basement membrane and Ck20 in contact with the light (here facing upwards images). There was, however, unusual uptake of Ckl 7 in the superficial cells for tumor cutting (indicated by a white arrow). In the same way, there was an unusual fixation of Ck5 at the submucosal level on the tumor section (indicated by a white arrow). Uroplakin-3A was also observed in superficial cells in contact with bladder light. The tumor tissue sections used for labeling Ki67 (cell proliferation marker) and GATA3 (nuclear transcription factor) were probably located at the level of a papillary fringe, as evidenced by the histological aspect and the thickness of the urothelium. Overexpression of Ki67 and GATA3 was observed on these tumor sections, evoking an increase in cell proliferation.

The organoids were then observed on day 14 under a confocal microscope, where IF labeling was performed with the same antibodies as those used on tissue sections (FIG. 9 and 10). We then observed an expression of the different urothelial markers, including Upk3A, showing differentiated cell expression and confirming the urothelial character of these structures. It seems that cell differentiation is from the outside to the inside of the structure. Indeed, the Ckl7 (basal marker) was observed mainly at the periphery of the structure while Ck20 or UPK3A (luminal markers) were expressed towards the center of these organoids whether in healthy cultures or tumours.

There is no specific difference between the IF labeling of organoids according to the healthy or tumor character under a confocal microscope.

Example 9: Pharmacological Responses of organoids following treatments

The organoids were cultured in Matrigel in 96-well black plates for 15 days. Healthy and tumorous organoid cultures were treated with cisplatin and gemcitabine from the 8th day of culture. The concentrations used are 2 mM, 20 mM and 200 pM for cisplatin and 0.05 pM, 0.5 pM and 5 pM for gemcitabine. These molecules were added to the culture medium, the latter being changed every two days until the 15th day of culture. The gemcitabine and cisplatin are chemotherapy medications used to treat bladder cancers (Roupret et al ., Progres en Urologie, 2018, 28: S46-S78).

The cell viability was analyzed using the CellTiter-Glo®3D kit (Promega, # 9683) at the fifteenth day of culture. Briefly, in order to dissociate the organoids, they were incubated with Tryple Express solution (Gibco) for 40 minutes at 37°C by pipetting regularly. After dissociation of the structures, the CellTiter-Glo®3D reagent was added to the culture medium of each well (volume 1 : 1) and incubated for 30 minutes at room temperature, in the dark, with stirring, according to the manufacturer's instructions. Luminescence was measured with a luminometer (Varioskan Flash-Thermo Scientific, Illkirch, France). Three wells per condition were measured and analyzed. Data analyzes were performed using Excel software, and IC50 values were calculated using non-linear regression. Comparisons between groups were made using the paired or unpaired Student t-test. The level of significance retained was p <0.05. Statistical analysis was performed using PRISM (Graph Pad Prism® Version 5.0, Software, Inc., California, USA). The IC50 of cisplatin for tumor culture was 1.75mM while the IC50 for healthy culture was 4.2mM (factor 2.4). These results show a greater effectiveness of treatment on tumor cells for the same concentration. The IC50 of Gemcitabine for tumor culture was 1.2 mM while the IC50 for healthy culture is 2.8 mM, also demonstrating a higher efficiency of chemotherapy on tumor culture (factor 2.3).

Example 10: Organoids from PDX

Organoids are developed from either fresh or frozen PDX obtained from bladder cancer. To obtain the organoids, the PDX tissue is handled as if it was the tumor tissue of a patient according to the protocol of example 2. Then, the same protocol as in examples 3, 5, 6, 8 and 9 is used to derive and obtain the organoids from the PDX.

Example 1 1 : pharmacological results on organoids derived from PDX model (Patient-Derived tumor Xenograft model) from bladder cancer

1.1. Description of the PDX model L987:

L987 is a basal-like model derived from a bladder carcinoma of high grade (G3) infiltrating the muscle of the bladder (pT4a). L987 has been developed as previously described (Lang H. et al, 2016, Oncotarget, Vol.7, No.37:59336-59359). L987 has been characterized at different levels to ensure the conservation of the patient’s tumour characteristics. The different analyses were:

- Histology (H&E staining) and IHC Transcriptomic,

Genomic (Single Tandem Repeat - STR, sequencing)

The histological profile of L987 (figure 11) and its parental tumour correlated with their molecular classification (basal tumour).

STR results (table 2) confirmed the high rate of identity between the PDX model observed after 3 passages (P3) and the patient’s tumour. Both the patient’s tumour and the PDX model harbour an FGFR3 mutation at position R284C Table 2:

1.2. Pharmacology assay with L987

The inventors evaluated the anti-tumour response of an EGFR inhibitor, Erlotinib, on L987 PDX tumor. The PDX was engrafted on the back of Male Mice, Swiss Nude Mice (Charles River Laboratories) that were 4 weeks at delivery.

In a first experiment, mice were treated either with a vehicle per os (p.o .) 5 times a week or with Erlotinib at 30mg/kg p.o., 5 days a week, for 4 weeks. No effect of Erlotinib was observed at that dosing. (Figure 12)

In a second experiment, mice were treated either with a vehicle by intrap eritoneal route 4 times a week or with Erlotinib at lOOmg/kg per os, 6 days a week, for 4 weeks. Results showed that the tumour responded to the treatment. The tumour growth inhibition was of 62,5% (Figure 13)

1.3. Generation and characterization of the organoid line BLOU-026 from the PDX model L987

The organoid line BLOU-026 was obtained following the same protocol as in examples 3, 5, 6, 8 and 9.

After 1 week culture, organoids observed were immature (cystic structures) or differentiated (budding structures).

Organoid dimensions varied from 40 to 500 pm, according to their differentiation state.

Organoid culture can be maintained for a long time, since organoids have been subjected to passaging more than 3 times.

Organoids obtained are representative of human bladder morphology, expressing specific markers of basal (CK5, CK17) and luminal (CK20, Upk3) cells.

The design of the study is shown on figure 14.

The cellular composition of the organoid line BLOU-026 has been examined using various markers. Uroplakin 3 is a marker specific for umbrella cells present in the luminal side of the bladder by contrast to markers expressed on cells that are on the basal side of the bladder. Uroplakin 3 is a marker of the late stage of the differentiation of the organoid showing that the obtained organoid would potentially encompass a lumen compartment. Other markers of the luminal compartment in the baldder were also detected which are GATA-3 and CK20. Additional markers were identified, providing proof for the presence of basal cells and of cells having proliferation capability, i.e., tumor cells. The identification of the markers provides proof of organoid formation with differentiated cells of the various urothelium cellular subtypes: superficial umbrella cells, intermediate cells and basal cells, the umbrella cells of the late differentiation stage showing that a mature organoid has been obtained.

Figure 15 accordingly shows the cellular composition of the organoid line BLOU-26: Immunofluorescence staining with the specific marker Uroplakin 3 : Upk3 labeling (orange), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

A- One confocal stack in the core of one selected organoid: Representative z-stack slice showing CK5 expression B- One confocal stack in the core of one selected organoid: Representative z-stack slice showing Ki67 expression

C- One confocal stack in the core of one selected organoid: Representative z-stack slice showing nuclei.

D- One confocal stack in the core of one selected organoid: Representative z-stack slice showing actin labeling

E- Merged representation for CK5, Ki67, actin and nuclei labeling

Figure 16 discloses cellular composition of the organoid line BLOU-026: Immunofluorescence staining with the specific luminal marker: GAT A3 labeling (red), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

A- One confocal stack in the core of one selected organoid: Representative z-stack slice showing GAT A3 expression.

B- Maximal intensity projection of all z-stack slices C- Merged representation for GAT A3, actin and nuclei labeling.

Figure 17: Cellular composition of the organoid line BLOU-026: Immunofluorescence staining with the specific markers CK20 (luminal) and CK17 (basal): CK20 labeling (orange), CK17 labelling (red), DAPI nuclei labeling (cyan), actin labeling (green), Organoids were analyzed with Opera Phenix (Perkin Elmer) (x20 N.A= 1.20).

A- One confocal stack in the core of one selected organoid: Representative z-stack slice showing nuclei.

B- One confocal stack in the core of one selected organoid: Representative z-stack slice showing actin labeling.

C- One confocal stack in the core of one selected organoid: Representative z-stack slice showing CK20 expression.

D- One confocal stack in the core of one selected organoid: Representative z-stack slice showing CK17 expression.

E- Merged representation for CK20, CK17, actin and nuclei labeling.

The presence of basal (CK5, CK17) and luminal biomarkers (CK20, GATA3 and Upk3) suggested that the experimental conditions allowed correct differentiation of the organoids from BLOU-026 line. GAT A3 was used as a marker of tumoral cells. The presence of Ki67 evidenced proliferation of the organoids.

1.4. Pharmacological assay with BLOU-026 As for the PDX model L987 from which BLOU-026 is derived, a pharmacological assay using the BLOU-026 line has been set-up and validated and a concentration-response curve to Erlotinib has been constructed.

A viability assay reflected by metabolic activity in organoids culture was used based on the commercial kit Cell Titer Glo 3D (Promega), evaluating the concentration of ATP in organoids by luminescence.

Pharmacological treatment (triplicates) - 2 Runs

Treatment for 5 days

- Vehicle: DMSO 2%

- Compound : Erlotinib (Run 1: 0.04 - 0.2 - 1 - 5 - 10 - 20 mM; Run 2: 0.02-0.04-0.1-0.2- 0.5-1 pM)

The results are shown on figure 19

1.5. Conclusion

In our experimental conditions, Erlotinib inhibits metabolic activity of organoid culture in a concentration-dependent manner. This result is in accordance with what has been observed in the corresponding PDX model L987.