WO2018189292A1 - Biomarqueurs de cellules prostatiques résistantes à la castration - Google Patents

Biomarqueurs de cellules prostatiques résistantes à la castration Download PDF

Info

Publication number
WO2018189292A1
WO2018189292A1 PCT/EP2018/059391 EP2018059391W WO2018189292A1 WO 2018189292 A1 WO2018189292 A1 WO 2018189292A1 EP 2018059391 W EP2018059391 W EP 2018059391W WO 2018189292 A1 WO2018189292 A1 WO 2018189292A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
prostate cancer
gene
overexpression
lsc
Prior art date
Application number
PCT/EP2018/059391
Other languages
English (en)
Inventor
Vincent Goffin
Lucila SACKMANN SALA
Jacques-Emmanuel GUIDOTTI
Gaëlle FROMONT
Original Assignee
Institut National De La Sante Et De La Recherche Medicale
Centre National De La Recherche Scientifique
Universite Paris Descartes
Centre Hospitalier Regional Universitaire De Tours
Universite De Tours Francois-Rabelais
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut National De La Sante Et De La Recherche Medicale, Centre National De La Recherche Scientifique, Universite Paris Descartes, Centre Hospitalier Regional Universitaire De Tours, Universite De Tours Francois-Rabelais filed Critical Institut National De La Sante Et De La Recherche Medicale
Publication of WO2018189292A1 publication Critical patent/WO2018189292A1/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the invention relates to the field of medicine. More specifically, the invention relates to the identification of a specific genomic and proteomic signature of castration-resistant prostatic cells. In particular, the invention relates to in vitro methods of prognosing the outcome of prostate cancer comprising identifying prostatic cells exhibiting biomarkers from this specific signature. The invention also relates to the uses of said cells in screening methods for identifying new candidate therapies for prostate cancer and more specifically of hormone refractory prostate cancer.
  • PCa Prostate cancer
  • Treatment options are chosen as a function of the stage of the disease which is determined taking into account notably blood PSA levels, prostate biopsy analyses, and the age of the patient. Management of the disease consequently varies from active surveillance (for low stage PCa) to treatment with curative intention (radical prostatectomy or radiotherapy) in case of clinically localized PCa and ultimately to hormonal therapy and/or chemotherapy.
  • Androgen deprivation therapy aims to block prostate cancerous cells from getting dihydrotestosterone, hormone which is required for the growth and spread of most PCa cells.
  • ADT has been shown to provide rapid and dramatic beneficial effects in the treatment of metastatic PCa and ADT is also used after PCa recurrence following radical prostatectomy or radiotherapy or as an adjunctive therapy for patients with locally advanced disease undergoing radiation therapy or surgery.
  • Androgen deprivation is reached either through orchiectomy or drug based treatments comprising chemical castration using LHRH agonists or antagonists (aiming at lowering the synthesis of testosterone) or antiandrogen therapy (aiming at preventing androgen receptor signal transduction).
  • Metastatic CRPC displays a high inter- and intra- patient heterogeneity by implying a mixture of cells displaying various range of Androgen Receptor (AR) expression levels and dependence thereon. Therefore, clonal selection in the course of treatment is a concern for the long-term use of AR inhibitors, though such mechanisms have not been formally proved. Furthermore, the origin of AR independent cancer cells is even more questioned.
  • WO2013149039 discloses the use of ex vivo organotypic cultures of human prostate cancerous cells and the identification of a group of specific markers of good prognostic regarding the risk of postoperative relapse.
  • LSC med LSC med
  • WT wild-type mouse prostate epithelia
  • prognostic tool for identifying or predicting a risk of prostate cancer relapse and/or androgen deprivation resistance as well as for new therapeutic tools and targets for the prevention of castration-resistant prostate cancer.
  • the present invention provides useful in vitro methods for prognosing the outcome of prostate cancer.
  • the invention stems, inter alia, from the discovery by the Inventors that a particular prostate epithelial cell type is found predominantly in prostate tissue in several in vivo models of prostate tumorigenesis. Furthermore, this cell type population is found notably increased in a premalignant model under androgen deprivation condition thereby highlighting the clinical implication of these cells in castration resistance processes. Last, Inventors also surprisingly identify the tumorigenic properties of these cells in a cancerous context.
  • Inventors have been able to identify a unique gene signature of the cells, thus providing useful, effective, and reliable tools for identifying or predicting a risk of prostate cancer relapse and/or androgen deprivation resistance.
  • an object of the invention relates to an in vitro method of prognosing the outcome of prostate cancer comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying the overexpression of at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl,
  • CD55 Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l, Illrn, Kcnk5, Krt4, Krt23, Krt7, Leng9, Lgals3, Ltf, Lypd2, Meis2, Mgat4a, Mgat5, Myof, Nbea, Ngf, Nt5c2, OsbpB, Pglyrpl, Pparg, Prss22, Psca, Reg3a, Reg3g, Snord73b, SlOOal, Saal, Sbsn, Scara3, Seel, Scnnla, Sdcbp2,
  • a particular object of the invention is an in vitro method of prognosing the outcome of prostate cancer comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying an overexpression of Krt4 gene.
  • a more particular object is the above in vitro method comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying an overexpression of Krt4 gene and an overexpression of at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl
  • Prognostic methods of the invention are particularly suited for identifying or predicting a risk of prostate cancer relapse or progression and/or androgen deprivation resistance from a biological sample of a subject, prostate sample being particularly preferred.
  • any of the above genes within said sample can be easily detected either at the protein or RNA level by methods well known in the art.
  • said prostatic epithelial cells represent at least 10% of cell exhibiting said overexpression.
  • said methods can be implemented by the detection in said sample of an overexpression of any of said genes of at least 1.5-fold change in comparison with non-cancerous prostatic epithelial cells.
  • said method can be implemented by immunohistochemical labelling of Krt4 encoded protein.
  • said methods further comprise a step of detecting, in said prostate sample, loss of basal cells ⁇ e.g. through immunohistochemical labelling of a high molecular weight cytokeratin or p63) and/or a step of detecting cancerous cells (e.g. through immunohistochemical labelling of AMACR protein).
  • prognosing methods of the invention are implemented in at least two samples collected from the same subject at a different time to detect a significant increase of the amount of said prostatic epithelial cells between these at least two samples.
  • prognosing methods of the invention comprise detecting the risk, presence or progression of hormone refractory prostate cancer.
  • Another object of the invention thus resides in a method for selecting an active compound in the prevention and/or the treatment of androgen blockade resistant prostate cancer, said method comprising the use of mouse cancerous prostatic epithelial cells resistant to androgen blockade and displaying the overexpression of Krt4 gene and at least one gene selected from 1700016G22Rik, 4930594C411Rik, AA986860, Acsm3, Aldhla3, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, Cbr2, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll5, Cxcll7, Cyp2f2, D17H6S56E-5, Daf2, Defbl4, Defb2, Dusp4, Ednl, F
  • a particular object of this invention also resides in a method of selecting an active compound in the prevention and/or the treatment of androgen blockade resistant prostate cancer, said method comprising the use of mouse cancerous prostatic epithelial cells resistant to androgen blockade and being Lin Sca-l + /CD49r ed or CK5 CK8 + /CK4 + .
  • Another object of the invention also resides in a method of selecting an active compound for the prevention and/or treatment of androgen blockade resistant prostate cancer, said method comprising the use of human cancerous prostatic cells resistant to androgen blockade and displaying the overexpression of Krt4 and at least one gene selected from : Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3,
  • FIGURES Figure 1 LSC med are the most abundant epithelial cell type in intact and castrated Pten pc / ⁇ prostate tumors. Representative FACS profiles of ⁇ ' prostatic epithelial cells gated as Lin-CD49f + .
  • C) graphical representation of cell type distribution. Bar graphs show mean ( ⁇ SD) percentages of basal/stem (dark grey), LSC med (white bar) and luminal cells (light grey) in n 2-5 independent experiments consisting of 1-10 pooled prostates each.
  • FIG. 2 LSC med are present in prostate of Pb-PRL and Wild Type mice, and proliferate after castration. Representative FACS profiles of Pb-PRL and Wild Type prostatic epithelial cells gated as Lin-CD49f + .
  • Each FACS profile shows gated epithelial populations: basal/stem, LSC med (thick black square), and luminal cells. Percentages are noted under each gate name.
  • Figure 3 Validation of expression of LSC ⁇ -specific genes by qPCR on sorted cell populations from WT, intact and castrated Pb-PRL and intact and castrated Pten IK/ ⁇ mouse prostates. In the three models, a noticeable upregulation of these genes was observed in LSC med compared to basal/stem and luminal cell populations, thereby confirming that LSC med signature is maintained across models.
  • B basal/stem
  • Lm LSC med
  • L luminal cells.
  • Figure 4 Decreased AR signalling in LSCTM* 1 cells. Relative expression level of AR signaling genes as determined by Affymetrix transcriptomic analysis of androgen signaling target genes in the three prostate cell populations enriched from wild type prostate by flow cytometry. (A) representative genes that are up-regulated by androgens, (B) representative genes that are down- regulated by androgens.
  • FIG. 6 LSC med are not enriched in Hi-myc prostates.
  • A Representative FACS profiles of Lin-, CD49f + cells of Hi-Myc mouse prostates, showing gated epithelial populations: basal/stem, LSC med (thick black square), and luminal cells. Percentages are noted under each gate name.
  • B Bar graph showing percentages of basal/stem (dark grey), LSC med (white bar) and luminal cells (light grey) for Hi-Myc prostates in an experiment with 3 pooled prostates.
  • Figure 7 LSC med are detected by CK4 staining and proliferate upon castration.
  • A Representative IHC staining for CK4 in prostates of intact and castrated WT, Pb-PRL and Pten pc / ⁇ mice. Small black arrows indicate CK4 + cells in the four upper panels. A strongly positive region (large black arrow) and a mainly negative region (large white arrow) are shown for intact Pten pc / ⁇ tissue (bottom left panel). Insets show higher magnification images.
  • Figure 8 CK4 staining in proximal vs distal regions of the prostate gland.
  • A Schematic representation of proximal and distal regions of mouse prostate lobes depicted on a 1 ⁇ 2 prostate showing one dorsal, one lateral and one ventral lobe surrounding the urethra.
  • B Representative IHC staining for CK4 in proximal and distal areas of WT and Pb-PRL prostate ventral lobe. Arrows show CK4 + cells. Scale bar in A: 2mm; in B: ⁇ .
  • Figure 9 CK4 staining in human prostate cancer samples.
  • A Representative images of serial slides from PDXs of treatment-naive prostatic tumors in intact or castrated hosts, stained for AMACR/p63 and CK4 by IHC. AMACR-positive tumor areas are marked with dotted lines, and show positive staining for CK4.
  • B Representative image of a tissue slide from a treatment-na ' ive Gleason 4 primary prostate cancer stained for CK4.
  • Figure 10 CK7 staining in Pb-PRL and Pten pc ⁇ ' mouse prostate.
  • A Representative images of slides from Pb-PRL mouse prostatic tissues showing an intense positive staining for CK7 in distal and proximal glands.
  • B Representative images of tissue slides of Pten pc / ⁇ prostate showing an extensive labelling of prostate tissue.
  • the invention relates to methods of prognosing the outcome of prostate cancer said methods comprising identifying epithelial prostatic cells exhibiting biomarkers identified by the inventors as constituting a specific signature of castration-resistant prostate cells that survive and even proliferate upon androgen blockade.
  • the invention also relates to the uses of said cells in screening methods for identifying new candidate therapies for prostate cancer and more specifically of hormone refractory prostate cancer.
  • prostate cancer refers to malignant prostate cancer and more particularly to adenocarcinomas.
  • Subject refers to any mammalian subject, preferably a human subject.
  • prognosing the outcome of prostate cancer means identifying or assessing the risk of presence, recurrence, or progression of prostate cancer, in particular the risk, presence or progression of hormone refractory prostate cancer. More particularly, prognosis methods of the invention can be used to identify disease subtype, to measure the severity, to monitor the progression of the disease or the conversion of a prostate cancer toward a hormone refractory prostate cancer, to qualify the prostate cancer, and/or to assess the responsiveness or likeness of response of a subject to androgen blockade treatments. In a particular embodiment, prognosis methods of the invention can also be used for patients' stratification in the course of clinical trials or to assess the efficacy of a treatment. As explained above "recurrence of prostate cancer” refers to reappearance of the prostate cancer after a first treatment, said treatment being of any form as, for example, chemotherapy, radiotherapy, hormone therapy, orchiectomy, or a combination thereof.
  • hormone refractory prostate cancer hormone resistant prostate cancer
  • broadcastration-resistant prostate cancer androgen blockade resistant prostate cancer
  • gene expression signature refers to one or more gene product (typically RNA or protein) that is specifically differentially expressed in a cell type, cell population, biological sample or tissue as compared to a control cell, control cell population, control biological sample or control tissue. Therefore, the identification of such a signature within a cell, cell population, biological sample or tissue is useful for prognosing the outcome of a disease.
  • Biomarker is also used to design a gene (i.e. any related transcript or protein) from said signature. More specifically, as used herein "LSC med (cell) signature” refers to one or more gene product that have been shown by the inventors as specifically differently expressed in LSC med cells when compared to prostate basal/stem cells and/or luminal cells.
  • Detection of an overexpression of a gene refers to the significant detection of the presence, and/or of an increase in quantity of a product from said gene, i.e. any transcript and/or corresponding protein (i.e. biomarker) in a sample of the subject to be tested as compared to a control. It can also refer to the detection, in a sample of said subject, of cells, or of an increased frequency thereof within a cell population, said cells being characterized by the presence and/or an increase in quantity of product(s) from these genes. In an embodiment, an increase is of at least 5% or even more in comparison with a control sample or reference value or mean value.
  • increases may be of about 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% (or even more). In a particularly preferred embodiment said increase is of at least 50%. In another embodiment said presence or increase is characterized by the detectability of a biomarker in a sample of the subject whereas said biomarker is no to barely detectable in control sample. Said tested sample will be then ranked as "positive" for said biomarker, as for example in immunofluorescence, immunohistochemical or FACS assays.
  • a positive sample for any biomarker of the invention can also be characterized by a change of labelling features for said biomarker as for example, a different localization in (a) particular subcellular region(s), and/or alteration in the quality of the labelling (e.g. from a focused to a diffuse labelling or conversely).
  • subject suffering or suspected of suffering from prostate cancer refers to any mammalian subject for which diagnostic of prostate cancer have been made, and/or presenting symptoms which are considered as making probable such a diagnostic.
  • prostate cancer can be suspected from screening with a prostate-specific antigen (PSA) blood test and/or a digital rectal exam.
  • PSA prostate-specific antigen
  • a PSA serum level above the normal values being considered as an indication of probable prostate cancer, in conjunction with other possible symptoms or analyses.
  • a continuous rise of PSA level in the same subject is also indicative of the progression of the disease.
  • rising of PSA levels signs the recurrence of the disease after a cancer therapy, when occurring after a previous treatment of the cancer.
  • only an analysis of prostatic tissue can lead to an actual diagnosis of prostate cancer.
  • Such analysis is usually based on the assessment of cell morphology within a prostatic tissue and the assignment of a Gleason score to the tissue based on how much the cells looks like normal, tissue being ranking from 1 (normal tissue) to 5 (very abnormal tissue).
  • Morphological assessment can be associated to an immunohistochemical labelling, in order to identify a loss of basal cells which signs cancer.
  • Basal cells are usually labelled with antibodies directed to high molecular weight cytokeratines (CK 903) or CK5/6 or to p63, a loss of labelling thus allowing to diagnose cancer. Nevertheless, being based on absence of labelling, false positives could arise from technical biases or atypical lesions.
  • Cytokeratin and/or p63 labelling can therefore be coupled to the labelling of AMACR (P504S) which has been found specific for tumour tissues (though not specific for prostatic tumour tissue); detection of CK903 or CK5/6 or p63 negative and AMACR positive foci allows to diagnose prostate cancer with certainty.
  • AMACR P504S
  • biological sample comprises any biopsy sample as incisional biopsy, excisional biopsy, or needle biopsy.
  • Biological sample comprises also any autopsy samples, frozen samples dedicated to histologic analyses, fixed or wax embedded sample.
  • biological sample are preferably a prostate biopsy sample, but it can also be metastasis biopsy, or lymph node biopsy from a subject suffering or suspected to suffer from prostate cancer or of prostate cancer relapse.
  • Biological sample can also comprise a prostate resection or a sample thereof, or prostatectomy samples.
  • Bio sample also includes blood and blood fractions or blood-derived products (tumor cells, circulating tumor DNA/RNA, exosomes), urine and urine fractions or urine-derived products, as well as cells or cell lines or organoids or patient- derived xenografts (PDX) derived from patient prostate samples.
  • blood and blood fractions or blood-derived products tumor cells, circulating tumor DNA/RNA, exosomes
  • urine and urine fractions or urine-derived products as well as cells or cell lines or organoids or patient- derived xenografts (PDX) derived from patient prostate samples.
  • PDX patient- derived xenografts
  • prostatic epithelial cells refers to the two main epithelial cell types, basal and luminal, as well as a minor population of neuroendocrine cells found in prostate.
  • prostatic epithelial cells also comprise LSC med cells that are shown herein to share some common features with both basal and luminal cells, though being a very specific cell population.
  • prostatic epithelial cells refers to cells that express cytokeratins and can be separated by cell sorting using cell-surface markers such as CD49f, Sca-1, CD24, CD44 or other cell-specific markers validated in the art.
  • LSC med cells As shown in the experimental section, Inventors have identified LSC med cells as an important therapeutic target for CRPC because they have progenitor properties, they increase proportionally upon disrupted AR-signalling, they withstand androgen blockade in vivo and they massively proliferate after castration in a relevant model of CRPC (Pten pc / ⁇ ). Inventors have surprisingly found that LSC med exhibit a unique gene expression signature that distinguishes them from prostate cell populations designated as luminal and basal/stem cells (Table 1) and defines these cells as a third distinct epithelial prostatic cell entity.
  • Gm6166 predicted gene 6166 nd nd nd
  • Igsfll superfamily member IGSF11 YES NO
  • LSC med cells represents the most abundant epithelial cell type in a model of invasive, castration- resistant, adenocarcinoma (Pten pc / ), and that this prevalence is not affected by castration. Furthermore, the LSC med part within prostate epithelium is significantly increased in WT mice and in Pb-PRL premalignant mouse model, under androgen deprivation condition. Furthermore, LSC med from Pten pc /" mice were found by the Inventors to exhibit tumorigenic properties. Altogether these results highlight the clinical implication of these cells in castration resistance and prostate cancer relapse.
  • the above genes i.e. any of their transcript or related protein
  • the detection of these cells and/or the detection of a significant increase of frequency of these cells through their specific expression of at least one of these genes within the prostate cell population of a subject is efficient in prognosing the risk for, the presence of, or the evolution of a prostate cancer toward a hormone resistant prostate cancer.
  • said subject is a human subject.
  • An object of the invention thus relates to an in vitro method of prognosing the outcome of prostate cancer, said method comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying the overexpression of at least one gene selected from Clorfl 16, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2f 1, Defb4A, Defdl 14, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of at least one gene selected from Aldhla3, Anxal, Anosl, Apod, Areg, Arll4, Aspa, AtplOb, B4galnt2, Bace2, Btc, C15orf62, Clorfl 16, C3, C7, Cadps, Capnl3, Capsl, Cd55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Dusp4, Ednl, F5, Ffar4, Gprl37b, Gprc5a, Gsdmc, Gsta4, Igsfl 1, Illrn, Kcnk5, Krt23, Krt4, Krt7, Lgals3, Ltf, Mgat4a, Mgat5, Myof, Nbea, Nt5c2, OsbpB, Pglyr
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of at least one gene selected from, Cyp2fl, Krt4, Saal, CxcL17, Ednl, Gsdmc, Pglyrpl, Reg3a, Reg3g and Psca.
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of at least one gene selected from Saal, Pparg, Aldhla3, Tspan8, Illrn, Ctse, Cdk6, Clcal, Ltf, Areg, Ednl, Krt4, Krt7, Clu, Ngf, Lgals3, F5, Psca, C3, Scnnla and Anxal.
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of at least one gene selected from Pglyrpl, Tspan8, Kcnk5, Ffar4, Gprl37b, Smiml5, Clcal, Psca, Gprc5a, Scnnla, Anxal, Bace2, Usp6nl, Seel, Sultlc2, Gsta3, Illrn, Apod, Capsl, Ltf, ATPIOB, Myof, Scara3, C7, Igsfl l, Sdcbp2, C3.
  • at least one gene selected from Pglyrpl, Tspan8, Kcnk5, Ffar4, Gprl37b, Smiml5, Clcal, Psca, Gprc5a, Scnnla, Anxal, Bace2, Usp6nl, Seel, Sultlc2, Gsta3, Illrn, Apod, Capsl, Ltf, ATPIOB, Myof, Scar
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of at least one gene selected from Krt7, Spns2, Gfpt2.
  • a combination of at least two biomarkers of the above lists is preferred.
  • cytokeratin 4 (CK4 protein, encoded by Krt4 gene) represent a reliable biomarker for LSC med cells being specific for these cells and providing results consistent with FACS profiles obtained in the different models (including human tumour xenografts) and conditions used in the experimental section.
  • another object of the invention relates to an in vitro method of prognosing the outcome of prostate cancer comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying an overexpression of Krt4 gene.
  • said in vitro method comprises identifying prostatic epithelial cells displaying the overexpression of Krt4 gene and of at least one gene selected from ClorfH6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATPIOB, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l, Illrn, Kcnk5, Krt23, Krt7, Leng9, Lgal
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of Krt4 gene and at least one gene selected from Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, AtplOb, B4galnt2, Bace2, Btc, C15orf62, Clorfl l6, C3, C7, Cadps, Capnl3, Capsl, Cd55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Dusp4, Ednl, F5, Ffar4, Gprl37b, Gprc5a, Gsdmc, Gsdmc, Gsdmc, Gsta4, Igsfl l, Dim,
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of Krt4 gene and at least one gene selected from, Cyp2f 1, Saal, CxcL17, Ednl, Gsdmc, Pglyrpl, Reg3a, Reg3g and Psca.
  • the prostatic epithelial cells detected in the above in vitro method display an overexpression of Krt4 gene and of at least one gene selected from Saal, Pparg, Aldhla3, Tspan8, Dim, Ctse, Cdk6, Clcal, Ltf, Areg, Ednl, Krt7, Clu, Ngf, Lgals3, F5, Psca, C3, Scnnla and Anxal.
  • Krt7 aka CK7
  • Spns2 or Gfpt2 labelling of Pb-PRL and Pten pc /" mouse prostate display the same features as CK4 labelling. Consequently, Krt7, Spns2, Gfpt2 are particularly suitable for the detection and identification of LSC med .
  • a particular embodiment of the invention relates to an in vitro method of prognosing the outcome of prostate cancer comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying an overexpression of Krt7 gene.
  • said in vitro method comprises identifying prostatic epithelial cells displaying the overexpression of Krt7 gene and of at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2f 1, Defb4A, Defdl 14, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l, Dim, Kcnk5, Krt23, Krt4, Leng9, Lgals3, L
  • the invention relates to an in vitro method of prognosing the outcome of prostate cancer comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying an overexpression of Spns2 gene.
  • said in vitro method comprises identifying prostatic epithelial cells displaying the overexpression of Spns2 gene and of at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa,
  • ATP10B B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l, Illrn, Kcnk5, Krt23, Krt4, Leng9, Lgals3, Ltf, Lypd2, Meis2, Mgat4a, Mgat5, Myof, Nbea, Ngf, Nt5c2, Osbpl3, Pglyrpl, Pparg, Prss22, Psca, Reg3a, Reg3g, Snor
  • the invention relates to an in vitro method of prognosing the outcome of prostate cancer comprising identifying, in a biological sample of a subject suffering or suspected of suffering from prostate cancer, prostatic epithelial cells displaying an overexpression of Gfpt2 gene.
  • said in vitro method comprises identifying prostatic epithelial cells displaying the overexpression of Gfpt2 gene and of at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2f 1, Defb4A, Defdl 14, Dusp4, Ednl, F5, Ffar4, Krt7, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l, Illrn, Kcnk5, Krt23, Krt4, Leng9, L
  • product of the gene biomarker of the LSC signature as mentioned in Table 1 can be detected by means well known in the art such as nucleic acid probe, RNA or protein aptamer, antibody and the like.
  • the transcript level can be determined by quantitative Reverse Transcription PCR, northern blotting assays, nucleic acid microarray, in situ hybridization, fluorescent labelling and the like.
  • the level of any protein or peptide encoded by said gene biomarker of the invention can be determined using western blotting assays, 2-dimensional electrophoresis, immunofluorescence, immunohistochemistry and the like.
  • high pressure chromatography either coupled or not with mass spectrometry methods (e.g.
  • MS/MS, Maldi/TOF, Maldi TOF/MS can be used to detect said gene product.
  • Said means and methods can be used alone or in combination in the methods of the invention.
  • the overexpression of a gene biomarker of the LSC med signature in the biological sample of the subject is determined by comparison with the expression level of said gene in a control sample.
  • said control sample is from a subject not suffering from prostate cancer.
  • said control sample is from a subject not suffering from castration-resistant prostate cancer.
  • said control sample has been previously collected from the same subject.
  • said control sample has been previously collected from the same subject before triggering an androgen deprivation or an hormonal therapy on this subject.
  • a significant presence of LSC med cells in a biological sample of a subject can be detected in an unsorted preparation of prostatic cells on the basis of their gene signature as described above, by detecting an increase quantity of the corresponding transcripts in said sample.
  • the methods of the invention comprise determining an increased quantity of RNA level of at least one gene of LCS med signature.
  • said increased RNA level of at least one gene is of at least 1.5 fold change or even more in comparison with non-cancerous prostatic cells or non-cancerous biological sample.
  • said increase corresponds to 2, 3, 4 times the RNA level found in noncancerous prostatic cells or non-cancerous biological sample.
  • the methods of the invention comprise the detection of epithelial cell positive, as described above, for at least one of the protein encoded by Pglyrpl, Tspan8, Kcnk5, Ffar4, Gprl37b, Smiml5, Clcal, CK4, Psca, Gprc5a, Scnnla, Anxal, Bace2, Usp6nl, Seel, Sultlc2, Gsta3, Dim, Apod, Capsl, Ltf, AtplOB, Myof, Scara3, C7, Igsf 11, Sdcbp2, and C3.
  • the methods of the invention comprise the detection of epithelial cell positive, as described above, for at least one of the protein encoded by a gene selected from Krt7, Spns2, Gfpt2.
  • said detection is performed either by a FACS analysis or by an immunohistochemical staining for said at least one protein.
  • the methods of the invention comprise the detection of cytokeratin 4 positive (CK4 + ) cells in the biological sample of the subject, Cytokeratin 4 (CK4) protein being known as encoded by the Krt4 gene.
  • said detection comprises an immunohistochemical staining for CK4 protein.
  • the methods of the invention comprise the detection of cytokeratin 7 positive (CK7 + ) cells in the biological sample of the subject, Cytokeratin 7 (CK7) protein being known as encoded by the Krt7 gene.
  • said detection comprises an immunohistochemical staining for CK7 protein.
  • the methods of the invention comprise the detection of Glutamine- fructose-6-phosphate transaminase 2 positive (GFPT2 + ) cells in the biological sample of the subject, GFPT2 protein being known as encoded by the Gfpt2 gene. In an even more preferred embodiment said detection comprises an immunohistochemical staining for GFPT2 protein.
  • the methods of the invention comprise the detection of spinster homolog 2 positive (SPNS2 + ) cells in the biological sample of the subject, SPNS2 protein being known as encoded by the Spns2 gene. In an even more preferred embodiment said detection comprises an immunohistochemical staining for SPNS2 protein.
  • the methods of the invention comprise the detection of Scnnla positive (SCNN1A + ) cells in the biological sample of the subject.
  • said detection comprises an immunohistochemical staining for SCNNla protein.
  • the methods of the invention comprise the detection of Psca positive (PSCA + ) cells in the biological sample of the subject.
  • said detection comprises an immunohistochemical staining for PSCA protein.
  • the methods of the invention comprise the detection of Cxcll7 positive (CXCL17 + ) cells in the biological sample of the subject.
  • said detection comprises an immunohistochemical staining for CXCL17 protein.
  • methods according to the invention further comprise immunohistochemical diagnostic test for cancer prostate comprising detecting in vitro in a biological sample from said subject:
  • in vitro methods according to the invention comprise:
  • AMACR positive cells sign(s) a castration-resistant prostate cancer, a progression of prostate cancer and/or recurrence.
  • in vitro methods according to the invention comprise:
  • a step of detecting AMACR positive cells in said sample wherein the presence of more than 10% of the cells being found CK4 + , a loss of basal cell and/or the presence of AMACR positive cells sign(s) a castration-resistant prostate cancer, a prostate cancer progression and/or recurrence.
  • in vitro immunohistochemical diagnostic test for cancer prostate is performed together or separately, simultaneously or at different time interval, with the detection of cells overexpressing at least one gene of the LSC med signature, on the same sample or on a different sample of the subject.
  • said detections are made simultaneously on the same biological sample of the subject.
  • said detection is made by using an immunohistochemical labelling cocktail, including a basal cell marker (e.g. p63) and a cancer cell marker (e.g. AMACR).
  • an object of the invention is a kit comprising a mean for detecting the overexpression of at least one gene product of LSC med signature as exposed above, selected from Clorfl 16, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl 1, Illrn, Kcnk5, Krt4, Krt23, Krt7,
  • an object of this invention is a kit comprising a mean for detecting the overexpression of Krt4 gene and of at least one gene selected from Clorfl 16, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl 1, Illrn, Kcnk5, Krt23, Krt7, Leng9, L
  • an object of this invention is a kit comprising a mean for detecting the overexpression of Psca gene and of at least one gene selected from Clorfl 16, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl 1, Illrn, Kcnk5, Krt23, Krt4, Krt7, Leng9,
  • kits comprising a mean for detecting the overexpression of Scnnla gene and of at least one gene selected from Clorf 116, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l, nirn, Kcnk5, Krt23, Krt4, Krt7, Leng9
  • kits comprising a mean for detecting the overexpression of Cxcll7 gene and of at least one gene selected from Clorfl 16, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsf 11, Illrn, Kcnk5, Krt23, Krt4, Krt7, Leng9, Lgals3, Ltf, Ly
  • an object of the invention resides in the use of a kit as above for prognosing the outcome of prostate cancer in a mammalian subject, preferably a human subject.
  • an object of this invention is the use of a kit comprising at least a mean for detecting CK4 + cells for prognosing the outcome of prostate cancer in mammalian subject, preferably a human subject.
  • kits comprising at least a mean for detecting PSCA + cells for prognosing the outcome of prostate cancer in mammalian subject, preferably a human subject.
  • a particular object of this invention is the use of a kit comprising at least a mean for detecting SCNNAl "1" cells for prognosing the outcome of prostate cancer in mammalian subject, preferably a human subject.
  • a further object of this invention is the use of a kit comprising at least a mean for detecting
  • CXCL17 "1" cells for prognosing the outcome of prostate cancer in mammalian subject, preferably a human subject.
  • a more particular object of this invention is any of the use of a kit as above in prognosing the risk of relapse or of developing an androgen blockade resistant prostate cancer in a mammalian subject, preferably a human subject.
  • any of the above-mentioned methods or kits can be used in a method for determining, adapting or modifying the treatment of prostate cancer in a subject suffering or suspected of suffering from said cancer.
  • experimental data show the clinical implication of LSC med cells in castration resistance and prostate cancer relapse, these cells having been found by the Inventors as increased upon androgen deprivation conditions and as having tumorigenic properties.
  • androgen deprivation therapy orchiectomy, chemical castration or antiandrogen therapy
  • alternative therapeutic strategies should be considered instead.
  • an object of the invention is a method for determining, adapting or modifying the treatment of prostate cancer in a subject suffering or suspected of suffering from said cancer, said method comprising not applying androgen deprivation therapy in a patient shown as having prostatic epithelial cells exhibiting an overexpression of any of the gene of the LSC med signature, or combination thereof, as described above elsewhere in this specification.
  • the method for determining, adapting or modifying the treatment of prostate cancer comprises not applying androgen deprivation therapy when said patient is shown as having prostatic epithelial cells exhibiting an overexpression of at least Krt4 gene or of Krt4 gene with at least one gene selected from Clorf 116, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3,
  • the method for determining, adapting or modifying the treatment of prostate cancer comprises not applying androgen deprivation therapy when said patient is shown as having prostatic epithelial cells exhibiting an overexpression of at least Krt7 gene or of Krt7 gene with at least one gene selected from Clorf 116, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3, G
  • the method for determining, adapting or modifying the treatment of prostate cancer comprises not applying androgen deprivation therapy when said patient is shown as having prostatic epithelial cells exhibiting an overexpression of at least Spns2 gene or of Spns2 gene with at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Gfpt2, C15orf62, Gprl37b, Gprc5a, Gsdmc, Gsta3,
  • the method for determining, adapting or modifying the treatment of prostate cancer comprises not applying androgen deprivation therapy when said patient is shown as having prostatic epithelial cells exhibiting an overexpression of at least Gfpt2 gene or of Gfpt2 gene with at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Defb4A, Defdl l4, Dusp4, Ednl, F5, Ffar4, Krt7, C15orf62, Gprl37b, Gprc5a, Gsdmc, G
  • an object of the invention relates to a method of selecting an active compound in the prevention and/or the treatment of androgen blockade resistant prostate cancer, said method comprising the use of mouse prostatic epithelial cells resistant to androgen blockade and displaying the overexpression of at least one gene selected from 1700016G22Rik, 4930594C411Rik, AA986860, Acsm3, Aldhla3, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, Cbr2, CD55, Cdc42ep5, Cdk6, Clcal, Clca3, Clu, Ctse, Ctsh, Cx
  • the above method of selecting an active compound in the prevention and/or the treatment of androgen blockade resistant prostate cancer comprises the use of mouse prostatic epithelial cells resistant to androgen blockade and displaying the overexpression of at least one gene selected from AA986860, Aldhla3, Anxal, Apod, Areg, Arll4, Aspa, AtplOb, B4galnt2, Bace2, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, CD55b, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2f2, Daf2/, Dusp4, Ednl, F5, Ffar4, Gml4137, Gprl37b, Gprc5a, Gsdmc2, Gsdmc3, Gsdmc4, Gsta4, Igsf 11, Illrn, Kcnk5, Krt23
  • Another object of the invention is a method of selecting an active compound in the prevention and/or the treatment of androgen blockade resistant prostate cancer, said method comprising the use of mouse prostatic epithelial cells resistant to androgen blockade, said mouse prostatic epithelial cells being found Lin Sca-l + /CD49r ed and/or CK57CK87CK4 + in FACS analysis.
  • Another object of the invention is a method of selecting an active compound in the prevention and/or treatment of androgen blockade resistant prostate cancer, said method comprising the use of human prostatic cells resistant to androgen blockade and displaying the overexpression of at least one gene selected from Clorfl l6, Acsm3, Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, ATP10B, B4galnt2, Bace2, Bexl, Btc, C3, C7, Cadps, Capnl3, Capsl, CD55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2f 1, Defb4A, Defdl 14, Dusp4, Ednl, F5, Ffar4, Gfpt2, Gml4137, Gprl37b, Gprc5a, Gsdmc, Gsta3, Gsta4, Igsfl l,
  • said method comprises the use of human prostatic cells resistant to androgen blockade and displaying the overexpression of least one gene selected from Aldhla3, Anosl, Anxal, Apod, Areg, Arll4, Aspa, AtplOb, B4galnt2, Bace2, Btc, C15orf62, Clorfl l6, C3, C7, Cadps, Capnl3, Capsl, Cd55, Cdc42ep5, Cdk6, Clcal, Clu, Ctse, Ctsh, Cxcll3, Cxcll7, Cyp2fl, Dusp4, Ednl, F5, Ffar4, Gprl37b, Gprc5a, Gsdmc, Gsta4, Igsfl l, fllrn, Kcnk5, Krt23, Krt4, Krt7, Lgals3, Ltf, Mgat4a, Mgat5, Myof, Nbea, Nt5c2, O
  • Compounds for the prevention and/or the treatment against androgen blockade resistant prostate cancer can be selected using in vitro assays well known in the art.
  • Such assays comprise, for example, MTT cytotoxicity assays whereby cell proliferation can be measured as a function of compound concentration in the culture medium.
  • Assays can be done on culture plates or, for example, in soft agar to follow the formations of 2D and 3D colonies respectively.
  • Such in vitro cell toxicity/proliferation assays are particularly convenient for high throughput screening of compounds libraries.
  • Xenografts of LSC med cells containing tumours and the subsequent follow up of tumour growth in the animals subjected to a treatment with candidate compound(s) are further tests that can be implemented in the methods of the invention.
  • Pten pc / ⁇ mice (pten loxP/loxP mice crossed with Pb-Cre4 transgenic males) were generated as described previously (2) and maintained on a C57BL/6 and Sv/129 mixed genetic background. Hi- Myc (ARR2/Pb-MYC) mice were maintained on a pure FVB/N background.
  • Pb-PRL mice prolactin transgene driven by the short probasin promoter were generated on the C57BL/6J background (>20 backcrosses), as previously described (3).
  • mice were used as controls and are referred to as WT animals.
  • Pb-PRL pre-malignant
  • Pten pc / ⁇ and Hi-Myc aggressive malignant
  • mice were housed in controlled conditions, on a 12/12-hour light/dark cycle with normal food and water provided ad libitum. Where indicated, mice were surgically castrated at the age of 2 (WT and Pb-PRL) or 3 weeks (Pten pcV ⁇ ) and for the latter, BrdU (1 mg/ml) was administered in the drinking water during 4 days before sacrifice. Prostate samples were obtained by microdissection immediately after sacrifice by cervical dislocation.
  • BD FACS Aria III BD Biosciences, San Jose, CA.
  • Prostate cells were isolated from freshly dissected mouse prostates by tissue mincing and collagenase digestion, trypsin and DNase treatment, followed by homogenization with syringe and needle and passing through a 40 ⁇ nylon mesh.
  • Cell suspensions from all genotypes were subjected to differential centrifugation using Histopaque-1119 (Sigma- Aldrich) to reduce the prostate secretion contents. Isolated cells were then stained on ice for 20 minutes.
  • Antibodies included FITC-coupled lineage "Lin” antibodies (anti-CD31, CD45 and Terl l9, 11-0311-85, 11-0451-85, 11-05921-85, respectively), PE-coupled anti-CD49f (integrin alpha-6; 12-0495-83) and APC-coupled anti-Sca-
  • lymphocyte antigen 6A-2/6E-1 (lymphocyte antigen 6A-2/6E-1; 17-5981-82) all from eBioscience, San Diego, CA. Dead cells were stained with SYTOX Blue (S34857, Life Technologies, Carlsbad, CA).
  • Sorted cells were loaded onto cytospin slide chambers of a Shannon Cytospin 2 (Thermo Scientific) and centrifuged at 500 rpm at room temperature for 10 min. They were collected in DMEM medium, supplemented with 50% FBS, glutamine, and penicillin-streptomycin, or in RA1 Lysis Buffer (Macherey-Nagel, Diiren, Germany) to perform RNA extraction.
  • RNA extraction and amplification RNA was extracted from FACS sorted cells with a Nucleospin RNA XS kit (Macherey-Nagel) according to the manufacturer's instructions. RNA quality and concentration measurements were performed using an Agilent RNA 6000 Pico kit on a BioAnalyzer (Agilent Technologies, Santa Clara, CA). RNA samples were amplified using an Ovation PicoSL WTA System V2 (NuGEN Technologies, San Carlos, CA), according to manufacturer's instructions. Resulting cDNA was purified using Agencourt RNAClean XP Beads (Beckman Coulter, Brea, CA).
  • Gene expression analysis was performed using GeneChip® Mouse Transcriptome Arrays 1.0 (Affymetrix, Santa Clara, CA). Prior to hybridization, cDNA was fragmented and biotin-labeled using the Encore Biotin Module (NuGEN). Biotinylated DNA fragments were hybridized onto the array chips using the Hybridization Wash Stain kit (Affymetrix). The chips were washed, stained, and scanned using the Affymetrix Model 450 Fluidics Station, the Affymetrix Model 3000 scanner and the Command Console software for piloting the GeneChip systems. For data analysis, raw data CEL files were imported in R/Bioconductor using the Oligo package [http://www.r- project.org/].
  • Expression levels were normalized using the RMA algorithm from the Affymetrix package. Expression levels and background noise were computed using a custom algorithm within R as follows. Assuming that a maximum of 80% of genes are expressed on any given microarray, 20% of probes were tagged with the lowest intensity for each microarray as background. A threshold was fixed at two standard deviations over the mean of the background. All probes for which normalized intensities were lower than the computed threshold were designated as background for each array. When comparing gene expression levels between two groups, a probe was included in the analysis if its intensity exceeded the background in at least 80% of the samples from at least one group. Group comparisons were done using Student's t-test and lists were filtered at P ⁇ 0.05 and fold change > 1.5. Cluster analysis was performed by hierarchical clustering using the Spearman correlation similarity measure and average linkage algorithm.
  • Gsdmc4 TGAGGAGCCTGCCAATCTAAA ATGTGGGGTGCTAGAATCCTT
  • PDX patient-derived xenografts
  • IHC/ICC Immunohisto/cy to chemistry
  • IF immunofluorescence
  • mice anti-CK4 MA1-35558; ThermoFisher Scientific, Waltham, MA
  • rabbit anti-Krt7 HPA007272; Sigma-Aldrich, Saint Louis, USA
  • rabbit anti-p63 clone 7JUL, Novocastra, Leica Biosystems
  • rabbit anti-AMACR clone 13H4, Dako, Santa Clara, CA
  • mouse anti-BrdU ref. 11170376001, Sigma Aldrich
  • antibody diluent MM- France, Francheville, France was used as negative control.
  • Fluorescent-labelled secondary antibodies used for IF were goat anti-mouse IgG-CFL 594 (sc-362277; Santa Cruz Biotechnology), goat anti-rabbit Alexa Fluor 488 (A-11070; Invitrogen) and goat anti-rat IgG- CFL 647 (sc-362293 Santa Cruz Biotechnology). 8. In vivo prostate regeneration assay
  • the in vivo prostate regeneration assay was performed as previously described (16). Briefly, prostates from 3 Pten pc /" mice were digested. 8 10 4 FACS-sorted LSC med cells or Basal/stem cells were mixed with 1.5 10 5 urogenital sinus mesenchymal (UGSM) cells mixed with growth factor reduced matrigel (Corning, Corning, NY) (1: 1, v/v) in a final volume of 150 ⁇ , and transplanted subcutaneously in immunodeficient male SCID mice for 10 weeks. Regenerated tissues were collected, fixed in 4% buffered formalin and embedded in paraffin for histological, morphological and/or IHC analysis.
  • UGSM urogenital sinus mesenchymal
  • Tumours of Prostate-specific Pten-deficient mice exhibit a significant increase in LSC med cells.
  • Pro state- specific Pten-deficient mice ⁇ Pten pc / ⁇ are known to develop invasive prostate adenocarcinomas. This model has been shown to mimic the progression of the disease toward invasive carcinoma and subsequent metastasis, as seen in humans (7).
  • Wild Type mice exhibit a very low prevalence of LSC med cells ( ⁇ 10%, Fig. 2A and C) in intact prostates and a significant increase of LSC med cells population after castration (>20%, Fig. 2B and C).
  • LSC med cells are found significantly increased in prostate cancerous epithelium in several in vivo models for prostate cancer and that, furthermore, these cells are resistant and even proliferate under androgen deprivation conditions, thereby suggesting a role for these cells type in prostate tumorigenesis and in the progression to castrate resistant prostate cancer (CRPC): some LSC med subsets may be more tolerant to castration than others, leading to clonal expansion.
  • LSC med cells exhibit a specific gene expression signature
  • Microarray analyses of the above WT FACS-sorted LSC med , basal/stem, and luminal cell populations were performed.
  • WT prostate were chosen to circumvent any bias due to prostate pathology.
  • LSC cells signature is conserved across mouse models LSC med signature is detected in Pten pc , ⁇ mouse prostates
  • LSC med represent the vast majority of epithelial cells in Pten pc / ⁇ mouse prostates (Fig. 1).
  • LSC med signature of Table 1 was used to query two independent transcriptomic datasets of Pten pc / ⁇ mouse prostate whole tissue (i.e. unsorted cells) relative to WT (GSE46799 and GSE46473) (8,9).
  • Both analyses revealed a strong overlap of LSC med -specific genes with those differentially expressed in Pten pc / ⁇ mice vs WT, most of the genes being concordantly up- or downregulated in both unsorted Pten pc / ⁇ prostates and sorted WT LSC med (Table 1, not shown for down-regulated genes).
  • the gene expression profile of unsorted Pten pc / ⁇ mouse prostates do reflect an enrichment of LSC med -specific genes, thereby showing that the signature can be detected using microarrays without any sorting of the cells.
  • LSC med signature is confirmed by qPCR in WT, premalignant Pb-PRL and malignant Pten pc , ⁇ mouse prostates sorted cells.
  • LSC med survive and are proportionally enriched upon castration thereby demonstrating their androgen-independence.
  • inventors observed that androgen signalling was markedly lower in LSC med , with AR-activated genes significantly downregulated and AR-repressed genes, upregulated or unchanged relative to luminal cells (representative examples are given in Fig. 4, ENTREZ gene ID in Table 3 below).
  • the intrinsically low androgen signalling of LSC med thus explains why they tolerate androgen-deprivation.
  • AR-activated genes Representative example of AR-repressed genes Gene symbol Gene ID mouse/human Gene symbol Gene ID mouse/human
  • LSC are enriched in low androgen signalling contexts
  • Cytokeratin 4 and 7 are protein markers for LSC
  • CK4 (Krt4 gene product, table 1), whose expression was validated by qPCR as overexpressed in LSC med only (Fig. 3), was tested as a marker to identify LSC med on tissue slides. To confirm that CK4 protein expression was specific for LSC med , all the three sorted cell populations (LSC med , luminal, basal/stem cells) were immunostained for CK4.
  • CK4 + cells are observed with an increased frequency from WT, to Pb-PRL, to Pten pc /" tissues (Fig. 7A, left column), consistent with FACS analyses (Fig. 1).
  • Castration is found to result in an increase of the frequency of CK4 + cells in WT and Pb-PRL prostates while their prevalence remained high in Pten pc /" mice (Fig. 7A, right column), which is also consistent with the proliferation of LSC med cells observed upon castration (Fig. l).
  • CK4 + cells were found to be restricted to the proximal regions of the gland (Fig. 8), where stem/progenitor cells have been shown to reside (13).
  • the CK4 + cells beside an increased frequency, were found distributed throughout the proximal and distal ductal regions (Fig. 8).
  • such a distribution is also observed for CK7 + cells, with an increased labelling of luminal epithelial cells within proximal as well as distal areas of the gland (Fig. 10A, circled areas).
  • genes of the LSCmed signature as identified herein by the inventors provide valuable tools for use for detecting the risk, presence or progression of hormone refractory prostate cancer.
  • LSCmed ofPterf mice are tumor initiating cells. Inventors tested the tumorigenicity of LSC med cells in the in vivo prostate regeneration as exposed in the Protocol section A). The evolution of grafts made of LSC med cells or basal/stem cells of Pten pc /" mice mixed with UGSM cells was assayed 10 weeks after the subcutaneous cells engraftment into SCID mice.
  • the basal/stem cells grafts were found to form tumoral glandular structures: high grade Prostatic Intraepithelial Neoplasia (PIN) lesions with some foci of carcinoma in situ (not shown). They were characterized by epithelial cell stratification, nuclear atypia, cribriform gland. Noteworthy, immuno staining of CK5, CK4 and CK8 showed presence of basal cells, luminal and LSC med cells in the formed tumoral structures.
  • PIN Prostatic Intraepithelial Neoplasia
  • Inventors made the observation that grafts from Pten pc /" LSC med cells were able to also form tumoral prostate epithelial structures (not shown). Indeed, up to now only basal/stem cells grafts were found to form tumoral glandular structures in this assay (17). Predominantly the histomorphology of these structures were found comparable to that of carcinoma in situ with focal micro invasion, PIN and extended atrophic glands. The tissue grafts were characterized by glandular structures with multiple atrophic, cystic glands, as well as less differentiated irregular arranged smaller glands. Focal non-atrophic irregular glands with loss of CK5 positive basal cell layer were found. Those glands showed micro-invasion through the basal membrane and cribriform luminal structures and were composed exclusively of CK4/CK8 positives cells as found for LSC med cell (not shown).
  • CK4/BrdU staining was used to address whether Pten pc /" LSC med play a role in cancer relapse.
  • CK4 + /BrdU + cells proliferation capacity was assayed three weeks after castration.
  • castrated Pten pc /" prostates exhibited large clusters of CK4 + /BrdU + cells located adjacent to negative regions (Fig. 7B), again suggesting clonal expansion.
  • This analysis proved that LSC med of Pten pc /" mice not only survive castration, but massively proliferate under these conditions of androgen depletion.
  • the identification of an enrichment in LSC med cells and/or the detection of an LSC med signature from a sample of a subject could help in predicting the responsiveness to androgen therapy and the risk of relapse and therefore in adapting the treatment of the subject.
  • CK4-positive cells are present in androgen-deprived patient-derived xenografts
  • FIG. 9 shows dual localization of AMACR + (Alpha-methylacyl-CoA racemase, a prostate cancer marker, Ref. 15), p63 " and CK4 + cells on serial sections (encircled regions) of both specimens.
  • AMACR + Alpha-methylacyl-CoA racemase, a prostate cancer marker, Ref. 15
  • p63 " and CK4 + cells on serial sections (encircled regions) of both specimens.
  • CK4 + cells were found present in primary tumor tissues (a representative section from a Gleason 4 prostate cancer is shown in Fig. 9B).
  • CK4 + cells are present in human prostatic tumors and are castrate- tolerant, consistent with a key role in the progression to castration-resistant prostate cancer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne l'identification d'une signature génomique et protéomique spécifique de cellules prostatiques résistantes à la castration. L'invention concerne, en particulier, des méthodes in vitro permettant de pronostiquer l'issue d'un cancer de la prostate, consistant à identifier des cellules prostatiques présentant des biomarqueurs à partir de cette signature spécifique.
PCT/EP2018/059391 2017-04-13 2018-04-12 Biomarqueurs de cellules prostatiques résistantes à la castration WO2018189292A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17166591 2017-04-13
EP17166591.2 2017-04-13

Publications (1)

Publication Number Publication Date
WO2018189292A1 true WO2018189292A1 (fr) 2018-10-18

Family

ID=58548620

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/059391 WO2018189292A1 (fr) 2017-04-13 2018-04-12 Biomarqueurs de cellules prostatiques résistantes à la castration

Country Status (1)

Country Link
WO (1) WO2018189292A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3674421A1 (fr) * 2018-12-28 2020-07-01 Asociación Centro de Investigación Cooperativa en Biociencias - CIC bioGUNE Procédés de pronostic du cancer de la prostate

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020119463A1 (en) * 2000-07-28 2002-08-29 Mary Faris Prostate cancer markers
WO2013149039A1 (fr) 2012-03-29 2013-10-03 YU, Winston, Chung-Yuan Marqueurs moléculaires pour la prédiction de pronostic du cancer de la prostate, procédé et trousse associés
WO2014004931A1 (fr) * 2012-06-27 2014-01-03 Berg Pharma Llc Utilisation de marqueurs dans le diagnostic et le traitement du cancer de la prostate
WO2016094425A1 (fr) * 2014-12-08 2016-06-16 Berg Llc Utilisation de marqueurs comprenant de la filamine a dans le diagnostic et le traitement du cancer de la prostate

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020119463A1 (en) * 2000-07-28 2002-08-29 Mary Faris Prostate cancer markers
WO2013149039A1 (fr) 2012-03-29 2013-10-03 YU, Winston, Chung-Yuan Marqueurs moléculaires pour la prédiction de pronostic du cancer de la prostate, procédé et trousse associés
WO2014004931A1 (fr) * 2012-06-27 2014-01-03 Berg Pharma Llc Utilisation de marqueurs dans le diagnostic et le traitement du cancer de la prostate
WO2016094425A1 (fr) * 2014-12-08 2016-06-16 Berg Llc Utilisation de marqueurs comprenant de la filamine a dans le diagnostic et le traitement du cancer de la prostate

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
BROWNE TJ; HIRSCH MS; BRODSKY G; WELCH WR; LODA MF; RUBIN MA.: "Prospective evaluation of AMACR (P504S) and basal cell markers in the assessment of routine prostate needle biopsy specimens", HUM PATHOL., vol. 35, 2004, pages 1462 - 8, XP004684399, DOI: doi:10.1016/j.humpath.2004.09.009
CARVER BS; CHAPINSKI C; WONGVIPAT J ET AL.: "Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer", CANCER CELL, vol. 19, 2011, pages 575 - 586, XP055039150, DOI: doi:10.1016/j.ccr.2011.04.008
CHEN Y; CHI P; ROCKOWITZ S ET AL.: "ETS factors reprogram the androgen receptor cistrome and prime prostate tumorigenesis in response to PTEN loss", NAT MED, vol. 19, 2013, pages 1023 - 1029
ELLWOOD-YEN K; GRAEBER TG; WONGVIPAT J ET AL.: "Myc-driven murine prostate cancer shares molecular features with human prostate tumors", CANCER CELL, vol. 4, 2003, pages 223 - 238
LAWRENCE, M.G.; TAYLOR, R.A.; TOIVANEN, R.; PEDERSEN, J.; NORDEN, S.; POOK, D.W.; FRYDENBERG, M.; PAPARGIRIS, M.M.; NIRANJAN, B.;: "A preclinical xenograft model of prostate cancer using human tumors", NAT PROTOC, vol. 8, 2013, pages 836 - 848
LAWSON DA; ZONG Y; MEMARZADEH S; XIN L; HUANG J; WITTE ON.: "Basal epithelial stem cells are efficient targets for prostate cancer initiation", PROC NATL ACAD SCI USA., vol. 6, 2010, pages 2610 - 5
MULHOLLAND DJ; TRAN LM; LI Y ET AL.: "Cell autonomous role of PTEN in regulating castration-resistant prostate cancer growth", CANCER CELL, vol. 19, 2011, pages 792 - 804, XP028232339, DOI: doi:10.1016/j.ccr.2011.05.006
OUDARD, S.; BANU, E.; BEUZEBOC, P.; VOOG, E.; DOURTHE, L.M.; HARDY-BESSARD, A.C.; LINASSIER, C.; SCOTTE, F.; BANU, A.; COSCAS, Y.: "Multicenter randomized phase II study of two schedules of docetaxel, estramustine, and prednisone versus mitoxantrone plus prednisone in patients with metastatic hormone-refractory prostate cancer", J CLIN ONCOL, vol. 23, 2005, pages 3343 - 3351
PENCIK J; SCHLEDERER M; GRUBER W ET AL.: "STAT3 regulated ARF expression suppresses prostate cancer metastasis", NAT COMMUN, vol. 6, no. 7736, 2015
ROUET V; BOGORAD RL; KAYSER C ET AL.: "Local prolactin is a target to prevent expansion of basal/stem cells in prostate tumors", PROC NATL ACAD SCI USA, vol. 107, 2010, pages 15199 - 15204
SACKMANN-SALA L; CHICHE A; MOSQUERA-GARROTE N ET AL.: "Prolactin-Induced Prostate Tumorigenesis Links Sustained Stat5 Signaling with the Amplification of Basal/Stem Cells and Emergence of Putative Luminal Progenitors", AM J PATHOL, vol. 184, 2014, pages 3105 - 3119
TOIVANEN R; FRYDENBERG M; MURPHY D ET AL.: "A preclinical xenograft model identifies castration-tolerant cancer-repopulating cells in localized prostate tumors", SCI TRANSL MED, vol. 5, 2013, pages 187ral71
TOIVANEN, R.; FRYDENBERG, M.; MURPHY, D.; PEDERSEN, J.; RYAN, A.; POOK, D.; BERMAN, D.M.; TAYLOR, R.A.; RISBRIDGER, G.P.: "A preclinical xenograft model identifies castration-tolerant cancer-repopulating cells in localized prostate tumors", SCI TRANSL MED, vol. 5, 2013, pages 187ral71
TSUJIMURA A; KOIKAWA Y; SALM S ET AL.: "Proximal location of mouse prostate epithelial stem cells: a model of prostatic homeostasis", J CELL BIOL, vol. 157, 2002, pages 1257 - 1265
WANG G; LUNARDI A; ZHANG J ET AL.: "Zbtb7a suppresses prostate cancer through repression of a Sox9-dependent pathway for cellular senescence bypass and tumor invasion", NAT GENET, vol. 45, 2013, pages 739 - 746, XP055176011, DOI: doi:10.1038/ng.2654
WANG S; GAO J; LEI Q ET AL.: "Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer", CANCER CELL, vol. 4, 2003, pages 209 - 221, XP008118654, DOI: doi:10.1016/S1535-6108(03)00215-0
XIN L; IDE H; KIM Y; DUBEY P; WITTE ON.: "In vivo regeneration of murine prostate from dissociated cell populations of postnatal epithelia and urogenital sinus mesenchyme", PROC NATL ACAD SCI USA., vol. 100, no. 1, 2003, pages 11896 - 903

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3674421A1 (fr) * 2018-12-28 2020-07-01 Asociación Centro de Investigación Cooperativa en Biociencias - CIC bioGUNE Procédés de pronostic du cancer de la prostate
WO2020136281A1 (fr) * 2018-12-28 2020-07-02 Asociación Centro De Investigación Cooperativa En Biociencias-Cic Biogune Procédés pour le pronostic du cancer de la prostate

Similar Documents

Publication Publication Date Title
Leja et al. Novel markers for enterochromaffin cells and gastrointestinal neuroendocrine carcinomas
Banzola et al. Expression of indoleamine 2, 3-dioxygenase induced by IFN-γ and TNF-α as potential biomarker of prostate cancer progression
Wang et al. BCAT1 expression associates with ovarian cancer progression: possible implications in altered disease metabolism
Li et al. Oral cancer-associated tertiary lymphoid structures: gene expression profile and prognostic value
Chen et al. Insulin‐like growth factor II mRNA‐binding protein 3 expression predicts unfavorable prognosis in patients with neuroblastoma
Soutto et al. Loss of TFF1 is associated with activation of NF-κB–mediated inflammation and gastric neoplasia in mice and humans
US7811778B2 (en) Methods of screening for gastrointestinal cancer
Mimeault et al. Molecular biomarkers of cancer stem/progenitor cells associated with progression, metastases, and treatment resistance of aggressive cancers
Romero et al. Assessment of topoisomerase II α status in breast cancer by quantitative PCR, gene expression microarrays, immunohistochemistry, and fluorescence in situ hybridization
JP5150909B2 (ja) 食道癌を診断する方法
Russo et al. SOX2 boosts major tumor progression genes in prostate cancer and is a functional biomarker of lymph node metastasis
WO2013006495A2 (fr) Méthodes de pronostic prédictif du cancer
US20110182881A1 (en) Signature and determinants associated with metastasis and methods of use thereof
Baněčková et al. Immunohistochemical and genetic analysis of respiratory epithelial adenomatoid hamartomas and seromucinous hamartomas: are they precursor lesions to sinonasal low-grade tubulopapillary adenocarcinomas?
Besso et al. FXYD5/Dysadherin, a biomarker of endometrial cancer myometrial invasion and aggressiveness: its relationship with TGF-β1 and NF-κB pathways
Shimada et al. Establishment of an immortalized cell line from a precancerous lesion of lung adenocarcinoma, and genes highly expressed in the early stages of lung adenocarcinoma development
Bommi et al. The transcriptomic landscape of mismatch repair-deficient intestinal stem cells
Zhao et al. TWIST2: A new candidate tumor suppressor in prostate cancer
Liu et al. TREM-1 expression in craniopharyngioma and Rathke's cleft cyst: its possible implication for controversial pathology
US20220170105A1 (en) Methods for diagnosis and prognosis of prostate cancer
US20190316205A1 (en) Mmp1 gene transcript for use as a marker for diagnosis of ovarian cancer prognosis, and test method
Dobi et al. ERG expression levels in prostate tumors reflect functional status of the androgen receptor (AR) as a consequence of fusion of ERG with AR regulated gene promoters
WO2018189292A1 (fr) Biomarqueurs de cellules prostatiques résistantes à la castration
Meng et al. Correlations of TOP2A gene aberrations and expression of topoisomerase IIα protein and TOP2A mRNA expression in primary breast cancer: a retrospective study of 86 cases using fluorescence in situ hybridization and immunohistochemistry
Landreville et al. Identification of differentially expressed genes in uveal melanoma using suppressive subtractive hybridization

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18717055

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18717055

Country of ref document: EP

Kind code of ref document: A1