Method for the Molecular Diagnosis of Prostate Cancer and Kit for Implementing Same

Information

  • Patent Application
  • 20100227317
  • Publication Number
    20100227317
  • Date Filed
    February 15, 2007
    17 years ago
  • Date Published
    September 09, 2010
    13 years ago
Abstract
The invention relates to a method for the molecular diagnosis of prostate cancer, comprising the in vitro analysis of the overexpression or underexpression of combinations of genes that can distinguish, with high statistical significance, tumorous prostate samples from non-tumorous prostate samples. The invention also relates to a kit for the molecular diagnosis of prostate cancer, which can perform the above-mentioned detection.
Description
FIELD OF THE INVENTION

The invention falls within the biotechnology sector and specifically within the field of methods for the diagnosis of prostate cancer. Accordingly, the present invention relates to a method for the molecular diagnosis of prostate cancer, comprising the in vitro analysis of the overexpression and underexpression of combinations of genes capable of differentiating between carcinomatous and noncarcinomatous prostate samples with high statistical significance. In particular, the present invention relates to a kit for the molecular diagnosis of prostate cancer capable of carrying out the aforementioned detection.


BACKGROUND OF THE INVENTION

Prostate cancer (PC) is a neoplasia having one of the highest rates of mortality and morbidity in industrialized countries and has therefore considerable socioeconomic impact [1], for which reason it is the subject of intensive study. Despite this effort, and in contrast to other types of neoplasia, comparatively little of substance is known about the molecular factors determining its initiation, maintenance, and malignant progression. On the other hand, the most singular characteristic of PC—its high androgen dependence—can provide important keys to understanding some of the molecular mechanisms underlying the biology of this cancer.


As in other cancers, there exists a genetic susceptibility to PC, which is why so many studies have sought to discover the link between genetic loci and susceptibility to PC. These studies have yielded a multiplicity and diversity of genetic loci [2]. None of these loci and genes explains more than a small proportion of PC familial clusters and, which is more striking, none have been confirmed in independent replication studies. This could be explained by the great genetic heterogeneity of PC, such that several high-penetrance genes can be associated with different familial PC pedigrees, as well as by the high frequency of phenocopies, i.e. sporadic PCs that have found themselves included in familial PC studies, owing to their characteristics being indistinguishable. Alternately, it could be that no single gene is associated with susceptibility to PC, but that instead many genes are involved, each of them being of relatively low penetrance. An additional characteristic of familial PC is that it is not associated to any significant degree with other cancer types, with the possible exception of breast cancer and tumors of the CNS (central nervous system) in specific family clusters, which indicates that the gene or genes involved do not participate in generalized neoplastic syndromes but seem instead to be “organ-specific”. However, PC has been used to study alterations in genes often associated with other neoplasias, such as TP53, BRCA1, PTEN, or repair genes affected, for example, in HNPCC (hereditary nonpolyposis colon cancer), and in fact few alterations or, as in the case of TP53 or PTEN, mutations that appear only at a late stage of tumor development have been found.


The fact that the PC susceptibility genes identified to date have been found altered in very few individuals and families stands in the way of an effective preventive approach to the problem. A related, though separate, question relates to early detection of PC. Determining the serum levels of PSA (prostate-specific antigen) in its various forms remains the most relevant reference for the detection and clinical follow-up of PC. Doubts arise when a differential diagnosis is required, or in cases where the PC is not accompanied by elevated PSA levels. This protein is a tissue marker and an androgen receptor signaling mechanism and not really a marker of malignity, so that, strictly speaking, its serum levels merely indicate the total mass of prostatic epithelial glands having the capacity to produce and secrete it. Elevated PSA levels are therefore observed not only in PC, but also in BPH (benign prostatic hyperplasia) and other benign prostatic processes, while, on the other hand, its production can sometimes be compromised in highly undifferentiated PCs, in which neoplastic prostate epithelial cells lose the capacity to express PSA.


This is why many laboratories are searching for new molecules that will offer greater specificity and sensitivity than PSA as a marker for the detection and follow-up of PC. The application of high-throughput (HT) techniques to the study of PC has allowed molecules to be identified that had previously not been associated with PC and which have shown themselves to be excellent malignity markers having a far superior differentiating capacity and specificity than PSA when detected in tissue [3]. Of these markers, the ones that stand out are alpha-methylacyl-CoA racemase (AMACR), hepsin (HPN), and fatty-acid synthase (FASN), which are expressed in large amounts in the majority of cases of PC, whereas, in contrast to PSA, its expression levels in normal prostate epithelium are minimal. Moreover, most malignant cells in PC lose their ability to express glutathione-S-transferase π (GSTP1) through hypermethylation of its promoter. Then again, as carcinomatous prostate glands have no basal cells, in PC there is decreased expression of genes and proteins characteristic of these cells, such as the high-molecular-weight keratins (e.g. CK5 or CK14) or the nuclear protein p63, a homolog of the cancer suppressor gene p53, which is expressed in the basal layers of several epithelia, including prostatic epithelium.


The availability of good reagents has allowed the use of some of these markers in clinically relevant applications such as determining levels in punch biopsy samples, thus demonstrating its usefulness in the diagnosis of doubtful cases of PC [4]. However, despite its great tumor specificity, none of the proteins mentioned is physiologically secreted by the prostate epithelium, which means that their determination in serum and other fluids—one of the greatest assets of PSA as an indicator of the mass of active prostate epithelium—does not give results that are fully consistent with their tissue determination. High-throughput studies are helping identify other secreted molecules that are expressed in anomalous quantities in PC. Determination of one or more of these proteins, even if they are not tissue-specific, in conjunction with the determination of PSA levels, is a promising avenue for developing tests of greater specificity and sensitivity.


There is therefore a need to identify subsets of markers for the diagnosis and prognosis of prostate cancer that are a significant improvement over existing ones. In this invention, new methods are provided for the molecular diagnosis of PC, having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, based on the detection of the expression of a series of gene subsets described in the present invention, as well as kits capable of performing said methods and the uses of said kits for the diagnosis and prognosis of the disease. The use of expression levels of sets of two or more genes to differentiate between carcinomatous and noncarcinomatous samples makes it possible to achieve levels of statistical significance in such differentiation that is often not achievable with the determination of the expression level of a single gene.


DESCRIPTION OF THE INVENTION
BRIEF DESCRIPTION OF THE INVENTION

The present invention relates to a method for the molecular diagnosis of prostate cancer, comprising the in vitro analysis, in a test sample, of the expression level of at least one gene or subsets of at least two genes selected from the group of 60 genes comprising: TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4.


In addition, the present invention relates, but is not limited to, kits for performing the aforementioned methods, as well as the uses for said kits.





DESCRIPTION OF THE FIGURES


FIG. 1. Clusters of samples analyzed on HGF (Human Genome Focus) arrays by means of FADA [13]. Samples were clustered automatically into carcinomatous (circle in the lower part of the Figure), normal (circle at top-right of the Figure), cell lines (circle on the left of the Figure), and stromal samples (circle at top left of the Figure).



FIG. 2. Eisen representation, after analysis by FADA and hierarchical clustering (HC), of the 318 genes over- and underexpressed in prostate samples that differentiate more significantly between normal prostate tissue samples and samples of carcinomatous prostate (Table 2). The expression values used to generate the hierarchical clusters are those corresponding to Table 6. The hierarchy is established by the so-called hierarchical clustering method. It is a standard method used in applied statistics and therefore any person skilled in the art can derive the result obtained in the present invention from the numerical values in Table 6. In the upper part of the image: N, samples of normal prostate; T, samples of prostatic adenocarcinoma; S, samples of pure prostatic stroma; C, culture cells. On the right are indicated the compartments to which the different groups of genes predominantly correspond. This is a post hoc interpretation, i.e. arrived at on the basis of the expression profiles observed for these genes.



FIG. 3. Hierarchical clustering of the 30 samples analyzed on Affymetrix HGF arrays, using the 45 genes included on Diagnostic Chip 1 (Table 3). The expression values used to generate the hierarchical clusters are those corresponding to Table 6. The resulting sample clusters are denoted as described for FIG. 2: N, normal prostate tissue; T, carcinomatous tissue; S, stroma; C, culture cells.



FIG. 4. Eisen diagram corresponding to the analysis by hierarchical clustering of the expression patterns in carcinomatous prostate, normal prostate, pure prostatic stroma, and cell lines, obtained on Affymetrix HGF arrays from 22 genes selected and validated by real-time RT-PCR (Table 4). The values used for the hierarchical clustering of these 22 genes were taken from Table 6. The resulting sample clusters are denoted as described for FIG. 2: N, normal prostate tissue; T, carcinomatous tissue; S, stroma; C, culture cells.



FIG. 5. Eisen diagram corresponding to the analysis by hierarchical clustering of the expression patterns in carcinomatous prostate, normal prostate, pure prostatic stroma, and cell lines, obtained on Affymetrix HGF arrays from 14 genes selected and validated by real-time RT-PCR (Table 5). The values used for the hierarchical clustering of these 14 genes were taken from Table 6. The resulting sample clusters are denoted as described for FIG. 2: N, normal prostate tissue; T, carcinomatous tissue; S, stroma; C, culture cells.



FIG. 6. Differentiation between samples of carcinomatous prostate (T) and normal prostate (N) by determining transcription levels using Affymetrix arrays of the MYO6 gene in combination with determining the transcription levels of the following genes: ABCC4, AMACR, BIK, BNIP2, CDK5, CSTA, DST, EIF3S2, EPHA2, ETS2, GJB1, HPN, NIT2, PYCR1, ROR2, TACSTD1, and TP73L. The expression values used for the hierarchical clustering of these genes were taken from Table 6.



FIG. 7. Discrimination between samples of carcinomatous prostate (T) and normal prostate (N) by determining transcript levels with Affymetrix arrays of the ABCC4 gene in combination with determining the transcription levels of the following genes: CSTA, GJB1, GSTP1, HOXC6, HPN, LAMB3, MYO6, PRDX4, and TP73L. The expression values used for the hierarchical clustering of these genes were taken from Table 6.



FIG. 8. Immunohistochemical detection of MYO6 protein in a tissue sample from a prostate cancer patient containing carcinomatous glands (T) and normal glands (N). The staining is clearly more intense in the carcinomatous epithelial cells than in the normal epithelial cells. The staining for MYO6 exhibits a cytoplasmic pattern having submembranous reinforcement.



FIG. 9. Immunohistochemical detection of EPHA2 protein in a tissue sample from a prostate cancer patient containing carcinomatous glands (T) and normal glands (N). The staining is exclusively in normal glands, specifically in basal layer cells. The staining exhibits a cytoplasmic pattern having membranous reinforcement.



FIG. 10. Immunohistochemical detection of CX3CL1 protein in various prostate tissue samples. FIG. 10 A: Sample comprising carcinomatous glands (T) and normal glands (N), wherein the CX3CL1 levels are clearly lower in carcinomatous cells than in normal cells. FIG. 10 B: Samples comprising carcinomatous epithelial cells, wherein the staining for CX3CL1 is intense in most of the carcinomatous cells. FIG. 10 C: Sample with PIN (P) and normal glands (N), wherein the staining is significantly more intense in the PIN cells than in the normal epithelial cells.





DETAILED DESCRIPTION OF THE INVENTION

For the realization of the present invention, a total of 31 prostate samples were analyzed by hybridization on Affymetrix Human Genome Focus arrays (FIG. 1).


The raw hybridization signals were normalized by the method of Irizarry et al. (2003) and subjected to unsupervised analysis using the FADA algorithm [13]. Genes were considered to be differentially expressed between normal and carcinomatous groups when their associated q-value [17] was less than 2.5×10−4. This analysis allowed samples to be clustered automatically, such that all the cancer samples, except one, were clustered in one clade and all the normal samples were clustered in another clade (FIG. 1). Established prostate cell lines and cells from primary explants obtained from samples of human prostate were also included in this analysis. In the FADA analysis, the cultured cells were clustered separately from the 2 aforementioned clades. From this analysis it was possible to deduce which genes are able to differentiate with the highest statistical significance (with p≦10−4 in Student's t-test with multiple correction) between carcinomatous samples and normal samples; a total of 318 genes were identified in this way, whereof 134 were found to be significantly over-represented (overexpressed) in cancers and 184 significantly under-represented in cancers (Table 2 gives a list of genes capable of differentiating between samples of carcinomatous prostate and normal prostate, analyzed according to their expression profiles obtained by hybridization on Affymetrix HGF microarrays).


Some of these genes and their relevance in PC are described in rather greater detail in the following chapters.


The previously studied genes overexpressed in PC (FIG. 2) were analyzed first. The identification of these genes served as external validation for the study. Genes in this category include the much investigated HPN and AMACR and, to a lesser extent, genes such as SIM2 and HOXC6. HPN has been extensively characterized [19-21], and it has been shown very recently that its overexpression can lead to a transformed phenotype in mouse models of prostate cancer [22]. AMACR has also been studied in many laboratories as a malignity marker in PC [23-27], and its clinical use has recently been expanded [26-29]. SIM2 has also been found, to a more limited extent, associated with PC [29], though it has already been studied as a possible therapy target with siRNA and antisense oligonucleotides in cell models [29, 31]. HOXC6 has been studied both as a malignity marker [32-35] and in its role in the survival of cultured prostate cancer cells [36].


Next, genes overexpressed in PC were analyzed that had not previously been unequivocally associated with prostate cancer. Among these genes there are many that appear in “lists” of genes from studies using microarray analysis, but none of these studies place any special emphasis on their biological characterization or make any special efforts in that direction. Among these genes there are transcription factors of very great interest in this context (FOXA1, NONO, ZNF278, ZNF85), vesicle transport protein genes (MYO6, RAB17, SYNGR2, RABIF), membrane transport genes (ABCC4, TMEM4, SLC19A7), fatty acid metabolism and nucleic acid metabolism-related enzyme genes.


The third group to be analyzed corresponded to the genes underexpressed in PC. Among the 184 genes detected as being significantly underexpressed in cancers, there is a relatively large number of genes that are expressed in stromal cells, so that it is suspected that, despite the care taken in selecting the samples to ensure a balance of the stromal component in carcinomatous and normal samples, the stromal component is more strongly represented in normal samples. However, there are also a large number of genes that appear to be typical of normal prostate epithelium and which are the ones that allow unsupervised clustering of normal samples in one and the same phyletic branch, separated from the stromal samples (FIG. 1). Some of these genes have already been described as exhibiting decreased expression in PC. Two examples are GSTP1 and LOH11CR2A, and it has already been shown that the absence of expression in tumors of these two genes is due to CpG island hypermethylation in their promoter regions [37, 38]. Another interesting gene is TP73L, which codes for p63 and of which several isoforms (principally ΔN and TA) are involved in the effector function of the p53 cancer suppressor gene [39]. It has been shown, in addition, that p63 expression is associated with basal epithelial cells of the normal prostate gland, and that deletion of this gene in mice impedes the formation of a normal prostate [40].


Furthermore, primary cultures of prostate epithelial cells, as well as prostate cell lines immortalized with HPV-16, but not tumorigenic ones (e.g. RWPE1), express TP73L, while prostate cells established from tumors do not express this gene. Other underexpressed genes in cancers are transcription factors of the FOX family (FOXO1A, FOXF1) and other transcription factors, potential cancer suppressors (TACC1, SLIT2), transmembrane receptors and their ligands (TGFBR3, TGB3, FGFR1, FGF2, FGF7, IL6R), or cell adhesion proteins (DDR2, CADH9, ITGA5, GJA1).


Additionally, the expression levels of some of the proteins corresponding to genes overexpressed or underexpressed in PC in the present study as well as in previous studies [13] were validated by immunohistochemistry on paraffin-embedded samples (in Tissue Microarray format).


One of the genes found overexpressed in the transcription studies, and whose protein was studied by immunohistochemistry, was MYO6. The present immunohistochemical study validated the transcription data, showing that the MYO6 protein is also overexpressed in the majority of cancers. A clear example of overexpression of the MYO6 protein in prostate cancer, by comparison with normal prostate glands, is shown in FIG. 8, which corresponds to a sample containing both carcinomatous prostate epithelium and normal prostate epithelium, having been stained with an MYO6-specific monoclonal antibody. This protein is an atypical myosin with endocytosis and vesicular transport functions and which previously had been shown to be expressed in large amounts in ovarian cancer, principally in association with invasive edges [41].


An analysis was also conducted of the in situ expression of several of the genes underexpressed in the present invention, in particular those whose underexpression represent a novelty in this neoplasia, such as the tyrosine kinase receptor EPHA2, the transcription regulator SNAI2, or the chemokine CX3CL1. These results are worth highlighting, especially in relation to EPHA2 and SNAI2, as both EPHA2 [42-59] and SNAI2 [59-62] have been associated in numerous publications with overexpression rather than underexpression in many types of cancer, including prostate adenocarcinoma.


An example of the absence of EPHA2 protein expression in carcinomatous prostate epithelium is shown in FIG. 9, wherein it is observed that while the normal prostate glands (in cells of the basal layer) express high levels of EPHA2, the adjacent carcinomatous prostate epithelial cells completely lack any reactivity and therefore express no detectable levels of this protein.


In the case of the CX3CL1 chemokine (also called fractalkine), the expression determined by real-time RT-PCR indicated a tendency for the carcinomatous epithelium to exhibit lower expression levels than the normal epithelium. Immunohistochemical staining for the corresponding protein, however, revealed variable profiles depending on the case, so that in some samples there was a significant decrease in CXC3L1 expression in carcinomatous epithelium, while in other cases the carcinomatous prostate epithelium gave high levels of said protein (FIG. 10). Finally, various cases of prostatic intraepithelial neoplasia (PIN) showed variable levels of staining for CX3CL1, being in some cases of greater intensity than in the adjacent normal epithelium (FIG. 10).


In the context of PC, therefore, our results indicate that, contrary to what has been generally accepted, the possible overexpression of these molecules should not be used as an indicator of malignity or serve as a therapeutic target in cancers of this type. Our data indicate, in fact, that the level of expression of these molecules in malignant prostate epithelium is low or nonexistent.


As a consequence of the foregoing analyses, a set of genes has been identified and defined, corresponding to the group of 318 genes, and also several subsets of genes on the basis of the former, useful for the molecular diagnosis of prostate cancer and having a high capacity for differentiating between carcinomatous and noncarcinomatous samples, wherein the determination of the levels of mRNA and/or protein represents a diagnostic signature of prostate cancer that constitutes a significant improvement over existing methods for the diagnosis of said cancer.


With the aim of designing a method for the diagnosis of prostate cancer in a format that is smaller than the set of 318 genes, more practical, and more akin to clinical practice (e.g. by means of RT-PCR analysis on a microarray or diagnostic chip), a smaller group of genes included in this first set was selected (see Example 2). This selection of a subset of 60 genes represents one of the many alternatives that can be obtained from the analysis of the original group of genes and should not be regarded as limiting the scope of the present invention. A person skilled in the art could come up with groups of genes different from those described in the present invention.


The first of the subsets contains a carefully selected set of 45 genes, validated by real-time RT-PCR, having a high capacity to differentiate between normal and carcinomatous samples (Table 3, FIG. 3). Another generated version, for the analysis of a still smaller number of genes, validated by real-time RT-PCR, retains virtually the same capacity to differentiate between normal and carcinomatous samples as the foregoing. Said subset of genes included in this design corresponds to the 22 validated genes shown in Table 4 and FIG. 4, or an even smaller subset of genes that corresponds to the 14 genes shown in Table 5 and FIG. 5. Other generated versions of gene subsets having a high capacity to differentiate between carcinomatous and noncarcinomatous samples and falling within the scope of the present invention are shown in FIGS. 6 and 7. The identification of the expression levels of all these gene subsets serves as the basis for the development of a relatively low-cost and high-performance prostate cancer diagnostic kit or device for quantifying multiple transcripts, in real time, on a platform that allows a diverse and high number of samples to be analyzed simultaneously. Preferably, when the diagnostic kit is based on the quantitation of transcripts, less than 1 ng of total RNA is required per sample. It is equally possible to develop a prostate cancer diagnostic kit or device based on the determination of the protein levels of said genes in cancer samples.


Therefore, in an initial aspect, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer comprising the in vitro analysis, in a test sample, of the expression level of at least one gene selected from the group of 60 genes consisting of: TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4.


In another aspect, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer comprising the in vitro analysis, in a test sample, of the expression level of at least two genes selected from the group of 60 genes consisting of: TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4, wherein the capacity to discriminate between carcinomatous and noncarcinomatous samples when the expression levels of two or more genes from said group are determined together is greater than the discriminating capacity of the same genes separately.


In particular, the discriminating capacity when the expression levels of two or more genes are determined together is 1%, preferably 10%, more preferably 25%, more preferably still 50% greater than the differentiating capacity of at least one of the genes separately.


In the context of the present invention, “discriminating capacity” is defined as the capacity to discriminate between carcinomatous and noncarcinomatous samples when applying a method for classifying samples based on the set of data obtained from expression analysis experiments for one gene or for a subset of at least two genes from the group of 60 genes that is the object of the present invention.


For example, when applying a given classification method to the set of samples described in Table 6, the capacity of the genes MYO6 and CDK5 to discriminate between carcinomatous and noncarcinomatous samples determined individually was 93.6% and 87.1%, respectively, whereas the discriminating capacity of both genes determined together was 96.8%. In another example, the discriminating capacity of the genes ABCC4 and FOXO1A determined individually was 87.1% and 83.9%, respectively, whereas the discriminating capacity of both genes determined together was 96.8%.


The expression “test sample” as used in the description refers, but is not limited to, biological tissues and/or fluids (blood, urine, saliva, etc.) obtained by means of biopsies, curettage, or any other known method serving the same purpose and performed by a person skilled in the art, from a vertebrate liable to have prostate cancer, where said vertebrate is a human.


In a preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer comprising the in vitro analysis, in a test sample, of the expression level of at least two genes selected from the group of 22 genes consisting of TACSTD1, HPN, AMACR, APOC1, GJB1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, MYO6, and ABCC4, wherein the capacity to discriminate between carcinomatous and noncarcinomatous samples when the expression levels of two or more genes from said group are determined together is greater than the discriminating capacity of the same genes separately.


In another preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer comprising the in vitro analysis, in a test sample, of the expression level of at least two genes selected from the group of 14 genes consisting of: TACSTD1, HPN, AMACR, APOC1, CX3CL1, SNAI2, GSTP1, KRT5, DST, LAMB3, CSTA, EPHA2, MYO6, and ABCC4, wherein the capacity to discriminate between carcinomatous and noncarcinomatous samples when the expression levels of two or more genes from said group are determined together is greater than the discriminating capacity of the same genes separately.


In another preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer comprising the in vitro analysis, in a test sample, of the expression level of at least two genes selected from the group of 7 genes consisting of: TACSTD1, HPN, DST, CSTA, LAMB3, EPHA2, and MYO6, wherein the capacity to discriminate between carcinomatous and noncarcinomatous samples when the expression levels of two or more genes from said group are determined together is greater than the discriminating capacity of the same genes separately.


In a third aspect, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the expression level of at least two genes selected from Table 3, wherein at least one of said selected genes is MYO6 or ABCC4.


In a preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the expression level of the MYO6 gene in combination with the analysis of the expression level of at least one gene from the group consisting of: ABCC4, AMACR, BIK, BNIP2, CDK5, CSTA, DST, EIF3S2, EPHA2, ETS2, GJB1, HPN, NIT2, PYCR1, ROR2, TACSTD1, and TP73L.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer with a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of the MYO6 gene in combination with the analysis of the overexpression of at least one gene from the group consisting of: ABCC4, AMACR, BIK, CDK5, EIF3S2, GJB1, HPN, NIT2, PYCR1, and TACSTD1.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of the MYO6 gene in combination with the analysis of the underexpression of at least one gene from the group consisting of: BNIP2, CSTA, DST, EPHA2, ETS2, ROR2, and TP73L.


In another preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer with a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the expression level of the ABCC4 gene in combination with the analysis of the expression level of at least one gene from the group consisting of: CSTA, GJB1, GSTP1, HOXC6, HPN, LAMB3, MYO6, PRDX4, and TP73L.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of the ABCC4 gene in combination with the analysis of the overexpression of at least one gene from the group consisting of: GJB1, HOXC6, HPN, MYO6, and PRDX4.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of the ABCC4 gene in combination with the analysis of the underexpression of at least one gene from the group consisting of: CSTA, GSTP1, LAMB3, and TP73L.


In another preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of MYO6, TACSTD1, or HPN genes or the analysis of the underexpression of DST, CSTA, LAMB3, or EPHA2 genes.


In another preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of MYO6, ABCC4, TACSTD1, HPN AMACR, or APOC1 genes or the analysis of the underexpression of the CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, or EPHA2 genes.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, or GJB1, or analysis of the underexpression of genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, or ETS2.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression of MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, GOLPH2, TRIM36, POLD2, CGREF1, or HSD17B4, or analysis of the underexpression of genes PRDX4 CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, or SCHIP1.


“Overexpressed gene” as used in the present invention should be understood to mean, in general, the abnormally high expression of a gene or of its transcription or expression products (RNA or protein) in cells coming from tumorigenic prostate tissue, when compared to the expression of said gene or its transcription or expression products (RNA or protein) in normal cells of the same nontumorigenic tissue. In the case of determining expression levels by hybridization on Affymetrix microarrays, any gene in a prostate cancer sample whose expression levels are at least 2.0 times as high as the expression levels of the corresponding noncarcinomatous prostate tissue sample is defined as “overexpressed”. When the determination is performed by quantitative RT-PCR, the term “overexpression” applies when the expression level of the gene in question in the cancer sample is at least 1.5 times the expression level in the corresponding normal prostate sample. However, when several cancer samples are being analyzed, a gene is considered to be “generally overexpressed” or “overexpressed in such prostate cancers when said gene is overexpressed in at least 70% of the cancer samples studied, comparing the normalized levels of said gene, determined in carcinomatous prostate tissue samples, with the arithmetic mean of the normalized levels of at least five samples of noncarcinomatous prostate tissue, the “overexpression” levels being quantitatively defined as described above for determinations on microarrays or by quantitative RT-PCR.


“Underexpressed gene” as used in the present invention should be understood to mean, in general, the abnormally low expression of a gene or of its transcription or expression products (RNA or protein) in cells coming from tumorigenic prostate tissue, when compared to the expression of said gene or its transcription or expression products (RNA or protein) in normal cells of the same nontumorigenic tissue. In the case of determining expression levels by hybridization on Affymetrix microarrays, any gene in a prostate cancer sample whose expression levels are one-half or less of the expression levels of the corresponding noncarcinomatous prostate tissue sample is defined as “underexpressed.” When the determination is performed by quantitative RT-PCR, the term “underexpression” applies when the expression level of the gene in question in the cancer sample is 0.75 times or less the expression level in the corresponding normal prostate sample. However, when several cancer samples are being analyzed, a gene is considered to be “generally underexpressed” or “underexpressed” in such prostate cancers when said gene is underexpressed in at least 70% of the cancer samples studied, comparing the normalized levels of said gene, determined in carcinomatous prostate tissue samples, with the arithmetic mean of the normalized levels of at least five samples of noncarcinomatous prostate tissue, the “underexpression” levels being quantitatively defined as described above for determinations on microarrays or by quantitative RT-PCR.


It was considered that a sample exhibited overexpression or underexpression of a protein with respect to another sample when the percentage difference in epithelial staining between the two samples was greater than 20% and/or the intensity differed by at least one point.


And, finally, in a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the overexpression or underexpression of the 318 genes indicated in Table 2.


In a fourth aspect, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to differentiate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the expression level of at least one gene or subsets of two genes selected from Table 3, wherein the analysis of the expression level of said genes is performed by determining the level of mRNA derived from their transcription and/or by determining the level of protein encoded by the gene or fragments thereof.


In a preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the expression level of at least one gene or subsets of two genes selected from Table 3, wherein the analysis of the expression level of said genes is performed by determining the level of mRNA derived from their transcription where the analysis of the mRNA level can be performed, by way of illustration and without limiting the scope of the invention, by PCR (polymerase chain reaction) amplification, RT-PCR (retrotranscription in combination with polymerase chain reaction), RT-LCR (retrotranscription in combination with ligase chain reaction), SDA, or any other nucleic acid amplification method; DNA chips produced with oligonucleotides deposited by any mechanism; DNA chips produced with oligonucleotides synthesized in situ by photolithography or by any other mechanism; in situ hybridization using specific probes labeled by any labeling method; by gel electrophoresis; by membrane transfer and hybridization with a specific probe; by NMR or any other diagnostic imaging technique using paramagnetic nanoparticles or any other type of detectable nanoparticles functionalized with antibodies or by any other means.


In another preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, of the expression level of at least one gene or subsets of two genes selected from Table 3, wherein the determination of the expression level of said genes is performed by determining the level of protein encoded by the gene or fragments thereof, by incubation with a specific antibody (wherein the analysis is performed by Western blot and/or by immunohistochemistry); by gel electrophoresis; by protein chips; by ELISA or any other enzymatic method; by NMR or any other diagnostic imaging technique.


The term “antibody” as used in the present description includes monoclonal antibodies, polyclonal antibodies, recombinant antibody fragments, combibodies, Fab and scFv antibody fragments, as well as ligand binding domains.


In a fifth aspect, the present invention relates, but is not limited to, a prostate cancer molecular diagnostic kit. Said kit may comprise primers, probes, and all the reagents necessary to analyze the variation in the expression level of at least one gene or subset of two genes of any of the aforementioned methods. The kit can additionally include, without any kind of limitation, the use of buffers, polymerases, and cofactors to ensure optimal activity thereof, agents to prevent contamination, etc. Furthermore, the kit can include all the media and containers necessary for start-up and optimization.


Accordingly, another object of the present invention is a device for the molecular diagnosis of prostate cancer, hereinafter called ‘diagnostic device of the invention,’ which comprises the necessary elements for analyzing the variation in the expression levels of at least one gene or subsets of two genes of any of the foregoing methods.


A preferred embodiment of the present invention consists in a diagnostic device of the invention for the detection of mRNA expression levels using a technique, by way of illustration and without limiting the scope of the invention, belonging to the following group: Northern blot analysis, polymerase chain reaction (PCR), real-time retrotranscription in combination with polymerase chain reaction (RT-PCR), retrotranscription in combination with ligase chain reaction (RT-LCR), hybridization, or microarrays.


Another preferred embodiment of the invention consists in a diagnostic device of the invention for the detection of mRNA expression levels comprising, by way of illustration and without limiting the scope of the invention, a DNA microarray, a DNA gene chip, or a microelectronic DNA chip, including gene probes.


Another preferred embodiment of the invention consists in a diagnostic device of the invention for the detection of protein expression levels using a technique, by way of illustration and without limiting the scope of the invention, a DNA microarray, belonging to the following group: ELISA, Western blot, and a protein biochip or a microarray-type device that includes specific antibodies.


In a sixth aspect, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, wherein the overexpression of the genes MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, or analysis of the underexpression of the genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMBr, or EPHA2 is used for the diagnosis of the presence of prostate cancer or of a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.


In a preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, wherein the overexpression of MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, or GJB1, or analysis of the underexpression of the genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, or ETS2 is used for the diagnosis of the presence of prostate cancer or of a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, wherein overexpression of the genes MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, or FOXA1, or analysis of the underexpression of the genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, or FOXF1 is used for the diagnosis of the presence of prostate cancer or of a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.


In a still more preferred embodiment, the present invention relates, but is not limited to, a method for the molecular diagnosis of prostate cancer having a high capacity to discriminate between carcinomatous and noncarcinomatous samples, comprising the in vitro analysis, in a test sample, wherein overexpression of the 318 genes indicated in Table 2 is used for the diagnosis of the presence of prostate cancer or of a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.


Unless otherwise defined, all technical and scientific terms used herein have the same meanings as those commonly understood by a person skilled in the art to which the invention belongs. Throughout the description and claims the word “comprises” and its variants do not seek to exclude other technical characteristics, components, or steps. To persons skilled in the art, other objects, advantages, and characteristics of the invention will be apparent, partly from the description and partly in the practice of the invention. The following examples and drawings are provided by way of illustration and do not be regarded as in any way limiting the present invention.


EXAMPLES OF THE INVENTION
Example 1
Identification of the Genes Associated with a Cluster Identifying a Prostate Cancer Tumor Pattern

For the realization of the present invention, a series of 31 human prostate samples were analyzed by hybridization on Affymetrix Human Genome Focus arrays (FIG. 1):


I. 20 samples enriched with carcinomatous epithelium.


II. 7 samples enriched with normal epithelium (<1% of cancer cells).


III. 1 sample comprising a group of 5 normal samples (POOL N).


IV. 3 samples consisting exclusively of stromal tissue.


The collected tissues were embedded in OCT, frozen in isopentane, and stored at −80° C. The samples were assessed histologically and selected for analysis in accordance with the following criteria: (a) minimum 90% of pure normal or carcinomatous epithelium in the normal and carcinomatous samples, respectively; (b) absence or minimal presence of foci of inflammation or atrophy. All the samples except three (one normal and two carcinomatous) come from the peripheral region, including the stroma samples. The estimated mean epithelial content in the carcinomatous samples was 70%, an average 90% of which exhibited neoplastic characteristics. The estimated mean epithelial content in normal samples was 40%, with no carcinomatous glands. The stroma samples contained less than 1% of epithelium. For extracting total RNA from the tissues, 20-30 cryosections were used, each 20 μm thick. To confirm the diagnosis and the quality of the samples, the first and last section of every sample was stained with hematoxylin-eosin. Table 1 describes the clinico-pathological characteristics corresponding to the samples used.









TABLE 1





Clinico-pathological characteristics corresponding to the samples used in


the study



















STROMA SAMPLES









1E



1E



18E







CARCINOMATOUS





SAMPLES
GLEASON SCORE
GRADE







3T
8
T2



4T
7
T3a



5T
7
T3a



6T
7
T3a



7T
6
T2



8T
9
T3a



9T
9
T3a



10T
7
T3a



11T
7
T3a



12T
6
T3a



13T
7
T2



14T
7
T2



1ST
5
T2



16T
7
T3a



17T
9
T2



18T
7
T2c



19T
8
T2



20T
6
T2



21T
7
T3a



22T
7
T3a







NORMAL SAMPLES







7N



9N



12N



13N



14N



17N



21N







PRIMARY CULTURES
ORIGINAL SAMPLE







PC17
17T



PC23
23T







*Clinical and pathological staging according to the international TNM classification of prostate adenocarcinoma.



The Gleason score goes from 2 to 10 and describes the aggressiveness of the cancer cells and, therefore, the likelihood of the tumor spreading. The lower the score, the lower the likelihood of the tumor spreading.






The cell lines HeLa and RWPE-1 (obtained from the American Type Culture Collection) were cultured in DMEM (PAA, Ontario, Canada) supplemented with 10% of serum (FBS) and KSFM (Gibco, Carlsbad, Calif.), respectively, with the aim of using them as controls. The primary cultures (PC17 and PC23) were derived from radical prostatectomies from patients having clinically localized prostate cancer, in which the adenocarcinoma had been detected macroscopically. The tissue explants were washed in PBS, ground, and cultured in KSFM (Gibco, Carlsbad, Calif.) supplemented with 5-α-dihydrotestosterone at a concentration of 10−11 M. After 4-5 weeks of culturing and two passes, the cultures were morphologically assessed to ensure absence of fibroblasts and used to obtain total RNA.


The tissue samples were laser-microdissected. 8 μm cryosections were mounted on plastic membrane-covered glass slides (PALM Mikrolaser Technology, Bernried, Germany), fixed for 3 minutes in 70% ethanol, stained with Mayer's hematoxylin (SIGMA, St. Louis, Mo.), dehydrated in a series of alcohols, left to dry for 10 minutes and stored at −80° C. until used. The samples were microdissected using the PALM MicroBeam system (PALM Mikrolaser Technology). Approximately 1.2 mm2 of normal or carcinomatous epithelium was collected for each sample and estimated to be 99% homogeneous by microscopic visualization.


Total RNA from the tissue samples and cell lines was extracted using the RNeasy Mini Kit (Qiagen, Valencia, Calif.). Total RNA from the microdissected samples was extracted with the RNeasy Micro Kit (Qiagen). In all cases there was a DNase I digestion step (Qiagen), and the RNA quality and concentration was assessed with the 2100 Bioanalyzer (Agilent Technologies, Palo Alto, Calif.).


For the gene expression analysis by microarray hybridization, RNA was used that had been isolated from 7 samples of normal prostate tissue with its corresponding pair (i.e. same patient and same surgical resection) of carcinomatous prostate sample, one sample comprising a mixture of equal parts of the RNA extracted from 5 samples of normal prostate tissue (normal pool), 13 unpaired carcinomatous samples (i.e. without a corresponding sample of normal prostate tissue from the same patient), 3 samples of pure normal prostate stroma (without epithelial tissue), two established epithelial cell lines (HeLa and RWPE-1), and two primary prostate cultures (PC17 and PC23). cDNA was synthesized from 2 μg of total RNA, using a primer having a promoter sequence for RNA polymerase T7 added at the 3′ end (Superscript II Reverse Transcriptase, Invitrogen, Carlsbad, Calif.). After synthesis of the second chain, an in vitro transcription was performed using the BioArray High Yield RNA Labeling Kit (Enzo, Farmingdale, N.Y.) to obtain biotin-labeled cRNA.


Prior to hybridization, washing, and scanning of the microarrays, the cRNA (15 μg) were heated at 95° C. for 35 min to provide fragments 35-200 bases long. Each sample was added to a hybridization solution [100 mM 2-(N-morpholino)ethanesulfonic acid, 1 M Na+, and 20 mM EDTA] in the presence of 0.01% Tween-20 at a final concentration of cRNA of 0.05 μg/mL. 5 μg of fragmented cRNA was hybridized on a TestChip (Test3, Affymetrix, Santa Clara, Calif.) by way of quality control. 10 μg of each fragmented cRNA were hybridized on Affymetrix Human Genome Focus Arrays at 45° C. for 16 h, washed and stained in the Affymetrix Fluidics Station 400, and scanned at 3 μm resolution in an Agilent HP G2500A GeneArray scanner (Agilent Technologies, Palo Alto, Calif.).


Computer analysis was then performed and the results obtained were normalized. The raw hybridization signals were normalized in accordance with the normalization method described by Irizarry et al. using the RMA algorithm [14], available as part of the Bioconductor package from Affymetrix. The first step in the RMA normalization procedure is to subtract the background signal; this is achieved taking into account that the observed PM probes can be modeled as a signal component that follows a normal distribution. The distribution parameters are adjusted on the basis of the data and the noise component is then eliminated. Normalization between arrays is then performed by quantile-quantile normalization at probe level, using the method proposed by Bolstad et al. [15]. The goal is for all the chips to have the same empirical distribution. Finally, the observed intensities of the groups of probes are summarized to obtain the measurement of the expression of each gene using the median polish algorithm [16], which is adapted to this model in a robust manner.


Prior to selecting the differentially expressed genes and to modeling the gene networks or the groups of genotypically consistent samples (see below), the genotypic consistency of the samples belonging to each of the groups was checked. The normalized expression data were analyzed using the FADA program [13]. This program applies a Q-Mode Factor Analysis, a multivariate tool related to PCA, coupled to clustering algorithms in sample space. Genes were considered to be differentially expressed between the normal and carcinomatous groups when their associated q-value [17] was less than 2.5×10−4. The q-values were calculated from the p-values obtained from the t-test using the Benjamini-Hochberg step-down false-discovery rate (FDR) algorithm [18], as implemented in the Bioconductor multitest package. This algorithm adjusts the p-values upward to eliminate the effects of multiple testing.


In the context of the present invention it is understood that the values of a parameter discriminate between two classes or categories of samples (in our case, carcinomatous samples and normal samples) with high significance when the value of p in a statistical comparison (by applying e.g. the t-test) between the two categories is <0.001. Table 6 shows the numerical data corresponding to the expression levels of the genes shown in the first column for the samples shown in the first row. Samples ending in T correspond to carcinomatous prostate and those ending in N correspond to normal prostate. Table 6 also shows the expression values for the cell lines HeLa (originating in a human cervical cancer) and RWPE-1 (human prostate epithelium transformed with the herpes virus HPV16), and for two primary explants derived from prostate cancers, designated PC17 and PC23. The digits are values of the signals obtained by hybridization of labeled cRNA on Affymetrix HGF microarrays, normalized by the MRA method [14].


This analysis enabled samples to be clustered automatically, such that all the carcinomatous samples, except one, were clustered in one clade and all the normal samples were clustered in another clade (FIG. 1). At the same time, the cultured cells and the stroma samples were clustered separately from the 2 aforementioned clades (FIG. 1).


From this analysis it was possible to identify the genes that were able to discriminate with the highest significance level (with p≦10−4 in Student's t-test with multiple correction) between carcinomatous samples and normal samples; a total of 318 genes were identified in this way, whereof 134 were found to be significantly over-represented (overexpressed) in cancers and 184 significantly under-represented (underexpressed) in cancers (Table 2).









TABLE 2







List of 318 genes capable of discriminating between samples of


carcinomatous prostate and normal prostate, analyzed according to the


expression profiles obtained for them by hybridization on Affymetrix HGF


microarrays












Genes overexpressed in

Genes underexpressed in




carcinomatous prostate

carcinomatous prostate













UniGene

UniGene



Gene symbol
cluster
Gene symbol
cluster







ALBCC4
Hs.508423
ACTB
Hs.520640



ACAT1
Hs.232375
ACTC
Hs.118127



ACY1
Hs.334707
ADAMTS5
Hs.58324



ADSL
Hs.75527
ALDH1A2
Hs.435689



AKR1A1
Hs.474584
ALDH2
Hs.436437



AMACR
Hs.508343
ANK2
Hs.137367



AP1M2
Hs.18894
ANXA2
Hs.511605



AP1S1
Hs.489365
APG1/HSPA4L
Hs.135554



APOC1
Hs.110675
ARHE
Hs.6838



APRT
Hs.28914
ARL7
Hs.111554



ATP5G1
Hs.80986
ASC/PYCARD
Hs.499094



ATP5G2
Hs.524464
ATP1A2
Hs.34114



ATP6V1F
Hs.78089
ATP2B4
Hs.343522



ATP6V1G1
Hs.388654
B4GALT5
Hs.370487



B4GALT3
Hs.321231
BHMT2
Hs.114172



BIK
Hs.475055
BIN1
Hs.193163



C15orf2
Hs.451286
BNIP2
Hs.283454



TRIB3
Hs.516826
BPAG1/DST
Hs.485616



CAMKK2
Hs.297343
CALM1
Hs.282410



CDK5
Hs.166071
CAPG
Hs.516155



CGREF1
Hs.546335
CAV1
Hs.74034



COX5A
Hs.401903
CAV2
Hs.212332



COX7A2L
Hs.339639
CD59
Hs.278573



CSTF3
Hs.44402
CES1
Hs.499222



CYB561D2
Hs.149443
CHST2
Hs.8786



DECR2
Hs.513233
CLIC4
Hs.440544



DHPS
Hs.79064
CLU
Hs.436657



DKC1
Hs.4747
CNN1
Hs.465929



DOM3Z
Hs.153299
CNN2
Hs.169718



DXS9879E
Hs.444619
COL13A1
Hs.211933



ECHS1
Hs.76394
COL17A1
Hs.117938



EIF3S2
Hs.530096
COL18A1
Hs.517356



ENTPD5
Hs.131555
CORO1C
Hs.330384



EPB41L4B
Hs.269180
CRYAB
Hs.408767



EPB42
Hs.368642
CSRP1
Hs.108080



ERP70
Hs.93659
CSTA
Hs.518198



ETFA
Hs.39925
CX3CL1
Hs.531668



FARSLA
Hs.23111
CYB5R2
Hs.414362



FBP1
Hs.494496
CYP27A1
Hs.516700



FKBP4
Hs.524183
CYP4B1
Hs.436317



FLJ10458
Hs.85570
DDR2
Hs.275757



FOXA1
Hs.163484
DES
Hs.471419



GABRD
Hs.113882
DF
Hs.155597



GALNT7
Hs.127407
DMPK
Hs.546249



GRL
Hs.29203
DNAJB4
Hs.380282



GJB1
Hs.333303
DPYSL3
Hs.519659



GOLPH2
Hs.494337
DVS27/C9orf26
Hs.348390



GTF3C2
Hs.75782
EDNRB
Hs.82002



GUSH
Hs.255230
EFEMP2
Hs.381870



HEBP2
Hs.486589
EFS
Hs.24587



HOXC6
Hs.820
ELF4
Hs.271940



HPN
Hs.182385
EMILIN1
Hs.63348



HRI/EIF2AK1
Hs.520205
EMP3
Hs.9999



HSD17B4
Hs.406861
ENIGMA/PDLIM7
Hs.533040



HSPD1
Hs.113684
EPAS1
Hs.468410



HYPK
Hs.511978
EPHA2
Hs.171596



ICA1
Hs.487561
ETS2
Hs.517296



KPTN
Hs.25441
EVA1
Hs.116651



LASS2
Hs.285976
FCGRT
Hs.111903



LIM/PDLIM5
Hs.480311
FEM1B
Hs.362733



MDH2
Hs.520967
FER1L3
Hs.500572



METTL3
Hs.168799
FEZ1
Hs.224008



MARCKSL1
Hs.75061
FGF2
Hs.284244



MRPL17
Hs.523456
FGF7
Hs.122006



MYO6
Hs.149387
FGFR1
Hs.264887



NDUFA7
Hs.515112
FGFRZ
Hs.533683



NDUFB4
Hs.304613
FLJ10539
Hs.528650



NDUFV2
Hs.464572
FLNA
Hs.195464



NFS1
Hs.194692
FLNC
Hs.58414



NIT2
Hs.439152
FLRT3
Hs.41296



NME1
Hs.118638
FOXF1
Hs.155591



NME2
Hs.463456
FOXO1A
Hs.370666



NONO
Hs.533282
FZD7
Hs.173859



NT5M
Hs.513977
GABRP
Hs.26225



P24B/TMED3
Hs.513058
GAS1
Hs.65029



P2RX4
Hs.321709
GATM
Hs.75335



P4HR
Hs.464336
GBPZ
Hs.386567



PCSK6
Hs.498494
GJA1
Hs.74471



PAFAH1B3
Hs.466831
GNAZ
Hs.555870



PAICS
Hs.518774
GPR161
Hs.271809



PCCB
Hs.63788
GPR87
Hs.58561



PDCD8
Hs.424932
GPRC5B
Hs.148685



PDE3B
Hs.445711
GRK5
Hs.524625



PDIR
Hs.477352
GSTM4
Hs.348387



PECI
Hs.15250
GSTP1
Hs.523836



PGLS
Hs.466165
HEPH
Hs.31720



PLEKHB1
Hs.445489
CFH
Hs.363396



POLD2
Hs.306791
HLF
Hs.196952



PP3111
Hs.514599
HSD11B1
Hs.195040



PPA2
Hs.480452
HSPB8
Hs.400095



PPIH
Hs.256639
IL6R
Hs.135087



PRDX4
Hs.83383
ILK
Hs.5158



PYCR1
Hs.458332
ISYNA1
Hs.405873



RAB11A
Hs.321541
ITGA5
Hs.505654



RAB17
Hs.44278
ITGB4
Hs.370255



RABIF
Hs.90875
KCNJ8
Hs.102308



RAP1GA1
Hs.148178
KCNMB1
Hs.484099



REPIN1
Hs.521289
KRT14
Hs.355214



REPS2
Hs.186810
KRT15
Hs.80342



RGS10
Hs.501200
KRT17
Hs.2785



RPL12
Hs.408054
KRT5
Hs.433845



RPL39
Hs.300141
KRT7
Hs.411501



RPL7A
Hs.499839
LAMB3
Hs.497636



RPS15
Hs.406683
LAPTM4B
Hs.492314



RUSC1
Hs.226499
LMCD1
Hs.475353



SERP1
Hs.518326
LMNA
Hs.491359



SERPINB6
Hs.519523
LOH11CR2A
Hs.152944



SFRS9
Hs.555900
MAOB
Hs.46732



SFRS9
Hs.555900
MAP1B
Hs.535786



SIM2
Hs.146186
MAPRE1
Hs.472437



SLC19A1
Hs.84190
MBNL2
Hs.125715



SLC25A10
Hs.511841
MCAM
Hs.511397



SND1
Hs.122523
MEF2C
Hs.444409



SNRPD2
Hs.515472
ZNF258
Hs.554935



SSR2
Hs.74564
MYH11
Hs.460109



STK16
Hs.153003
NAB1
Hs.107474



STX3A
Hs.530733
NT5E
Hs.153952



SYNGR2
Hs.464210
OPTN
Hs.332706



TACSTD1
Hs.692
PALM2-AKAP2
Hs.259461



TIMM13
Hs.75056
PCDH9
Hs.407643



TM4SF13
Hs.364544
PDE2A
Hs.503163



TM9SF2
Hs.130413
PDE4A
Hs.89901



TMEM4
Hs.8752
PDLIM4
Hs.424312



TRAP1
Hs.30345
PER2
Hs.58756



TREM2
Hs.435295
PFKFR3
Hs.195471



TRIM36
Hs.519514
PGRMC1
Hs.90061



TRIP13
Hs.436187
PLEKHA1
Hs.287830



TROAP
Hs.524399
PLN
Hs.170839



TXN
Hs.435136
PLP2
Hs.77422



UCK2
Hs.458360
POPDC2
Hs.16297



WDR23
Hs.525251
PPP1R12A
Hs.49582



ZMPSTE24
Hs.132642
PPP1R12B
Hs.444403



ZNF278
Hs.517557
PRNP
Hs.472010



ZNF85
Hs.37138
PSIP1
Hs.493516





PTBP2
Hs.269895





PTGER2
Hs.2090





PTGIS
Hs.302085





RARGEF1
Hs.530053





RARRES2
Hs.521286





RASL12
Hs.27018





RBBP7
Hs.495755





RIBM9
Hs.282998





RBMS1
Hs.470412





RBP1
Hs.529571





RBPMS
Hs.334587





ROCK2
Hs.58617





ROR2
Hs.98255





S100A6
Hs.275243





SART2
Hs.486292





SCHIP1
Hs.134665





SEC23A
Hs.272927





SERPINB1
Hs.381167





SERPINE2
Hs.3 8449





SLCZSA12
Hs.470608





SLC8A1
Hs.468274





SLIT2
Hs.29802





SMARCD3
Hs.444445





SMTN
Hs.149098





SNAI2
Hs.360174





SNX7
Hs.197015





SRD5A2
Hs.458345





SRF
Hs.520140





ST5
Hs.117715





STAT5B
Hs.132864





SVIL
Hs.499209





TACC1
Hs.279245





TAZ
Hs.409911





TCF7L1
Hs.516297





TCIRG1
Hs.495985





TGFB3
Hs.2025





TGFBR3
Hs.482390





TMG4/PRRG4
Hs.471695





TP73L
Hs.137569





TPM2
Hs.300772





TRIM29
Hs.504115.





TRPC1
Hs.250687





TU3A
Hs.8022





VAMP3
Hs.66708





VCL
Hs.500101





WDR1
Hs.128548





WFDC2
Hs.2719





ZFHX1B
Hs.34871










Example 2
Validation of the Genes Identified as Most Relevant in the Microarray Hybridization Experiments by Means of Real-Time RT-PCR Using the TaqMan LDA Format

This was done by performing real-time RT-PCR on the genes of greatest interest biologically and as markers from the complete panel of 318 genes identified previously, using both non-microdissected samples and samples laser-microdissected using the PALM instrument. The object of the RT-PCR analysis is to determine the expression levels of these genes in a diagnostic chip-type format, which is smaller and more akin to clinical practice.


Real-time RT-PCR was carried out for each replica of prostate tissue (in triplicate) or of microdissected samples (in quadruplicate), whether of carcinomatous or normal tissue. Thus, 1 ng of starting total RNA was used for the synthesis of cDNA using the reverse transcriptase Superscript II (Invitrogen) and random hexamers at 42° C. for 50 min, followed by treatment with RNase at 37° C. for 20 min. The resulting cDNA were used to perform real-time PCR in an ABI PRISM 7900HT instrument (Applied Biosystems, Foster City, Calif.), using a specially designed TaqMan Low Density array (Applied Biosystems) containing primers and probes specific for 45 genes of interest and the RPS18 gene for calibration, and designated as Diagnostic Chip 1 (see Table 3). The Thermocycler conditions were established in accordance with the manufacturer's specifications. The data obtained were analyzed using the SDS 2.1 software (Applied Biosystems) applying the ΔΔCt relative quantification method.









TABLE 3







List of the 45 genes that best discriminate between carcinomatous and


normal prostate samples








Genes overexpressed in
Genes underexpressed in


carcinomatous prostate
carcinomatous prostate










Gene symbol
UniGene cluster
Gene symbol
UniGene cluster













PP3111
Hs.514599
DDR2
Hs.275757


CAMKK2
Hs.297343
CLU
Hs.436657


ZNF85
Hs.37138
TP73L
Hs.137569


MYO6
Hs.149387
SNAI2
Hs.360174


SND1
Hs.122523
ETS2
Hs.517296


NONO
Hs.533282
KRT5
Hs.433845


ICA1
Hs.487561
TGFBR3
Hs.482390


ABCC4
Hs.508423
GSTP1
Hs.523836


PYCR1
Hs.458332
ROR2
Hs.98255


ZNF278
Hs.517557
LAMB3
Hs.497636


TACSTD1
Hs.692
BPAG1/DST
Hs.485616


APOC1
Hs.110675
CSTA
Hs.518198


BIK
Hs.475055
CX3CL1
Hs.531668


HOXC6
Hs.620
GJA1
Hs.74471


CDK5
Hs.166071
BNIP2
Hs.283454


AMACR
Hs.508343
PER2
Hs.58756


LASS2
Hs.285976
EPHA2
Hs.171596


HPN
Hs.182385
FOXO1A
Hs.370666


NME1
Hs.118638
FOXF1
Hs.155591


PRDX4
Hs.83383


GJB1
Hs.333303


SYNGR2
Hs.464210


SIM2
Hs.146186


EIF3S2
Hs.530096


NIT2
Hs.439152


FOXA1
Hs.163484









For these determinations, the microdissected material consisted exclusively of pure epithelial cells, taken either from tumors or from normal prostate tissue.


This first carefully selected subset of 45 genes provided a high capacity to discriminate between normal and carcinomatous samples. The selection of these genes was based on three criteria: (1) the capacity of each gene to discriminate between normal and carcinomatous samples in the expression analysis on Affymetrix HGF microarrays (values from Table 6), i.e. genes having the most significant p values; (2) the biological interest thereof, based on functional and expression data previously described in the scientific literature; and (3), as far as possible, the existence of commercial antibodies specific for the corresponding proteins, for subsequent validation of expression by means of immunoassays, including immunohistochemical determinations.


In fact, this subset of genes correctly includes within the group of carcinomatous samples a sample that had been incorrectly grouped together with global transcriptomic analysis by means of FADA (FIG. 2).


More specifically, in the case of microdissected samples it was found by this method that, of the 26 genes included in Diagnostic Chip 1 that were considered to be overexpressed in tumors according to Affymetrix HGF microarray analysis, 13 genes (50%) also exhibited higher levels in tumors than in noncarcinomatous tissue in quantitative determination by real-time RT-PCR. In the case of the 19 genes found underexpressed in tumors by microarray determination, of the 18 genes that were detectable, 18 (95%) were found underexpressed by real-time RT-PCR in the analysis of non-microdissected samples. When the quantitative determination was performed on microdissected samples (i.e. comparing carcinomatous pure epithelia with normal pure epithelia from the same individuals), it emerged that, of the 26 genes selected as overexpressed in tumors, only 9 (34.6%) were also found overexpressed in the majority of samples by means of transcript quantification by real-time RT-PCR. In this determination on microdissected samples, of the 19 genes considered as underexpressed in tumors following the microarray analyses, 18 were assessable and, of these, 15 (83.3%) were also found underexpressed in most microdissected samples using quantitative determination by real-time RT-PCR. Therefore, of the 45 assessable genes on Diagnostic Chip 1 (26 overexpressed and 19 underexpressed), 24 (9 overexpressed and 15 underexpressed) had their respective expression profiles validated by real-time RT-PCR on laser-microdissected pure epithelia. Taking into account the results obtained in the validations with non-microdissected samples and with microdissected samples, genes that had been validated in both analyses were selected, resulting in a set of 22 genes (7 overexpressed and 15 underexpressed; see Table 4). Taking the expression data from the Affymetrix HGF microarray analysis corresponding to these 22 genes, it was found that this small subset of expression data allows perfect differentiation between carcinomatous and normal samples with high statistical significance (FIG. 4).









TABLE 4







List of the 22 genes that best discriminate between carcinomatous and


normal prostate samples








Genes overexpressed in
Genes underexpressed in


carcinomatous prostate
carcinomatous prostate










Gene symbol
UniGene cluster
Gene symbol
UniGene cluster





TACSTD1
Hs.692
FOXO1A
Hs.370666


ABCC4
Hs.508423
TGFBR3
Hs.482390


MYO6
Hs.149387
CLU
Hs.436657


GJB1
Hs.333303
ROR2
Hs.98255


HPN
Hs.182385
SNAI2
Hs.360174


AMACR
Hs.508343
GSTP1
Hs.523836


APOCi
Hs.110675
BPAG1/DST
Hs.485616




KRT5
Hs.433845




CSTA
Hs.518198




LAMB3
Hs.497636




EPHA2
Hs.171596




ETS2
Hs.517296




CX3CL1
Hs.531668




GJA1
Hs.74471




PER2
Hs.58756









Using even stricter real-time RT-PCR validation criteria for selecting genes overexpressed or underexpressed in tumors, and taking into account the compartments in which it had been deduced from their expression profiles that each gene was expressed, it was possible to identify an even smaller subset of 14 genes (6 overexpressed in tumors and 8 underexpressed; see Table 5). Again taking the expression data corresponding to these 14 genes obtained for all the starting samples on Affymetrix HGF microarrays, it was found that this smaller subset was also able to discriminate with high statistical significance between carcinomatous samples and normal prostate samples (FIG. 5).









TABLE 5







List of the 14 genes that best discriminate between carcinomatous and


normal prostate samples








Genes overexpressed in
Genes underexpressed in


carcinomatous prostate
carcinomatous prostate










Gene symbol
UniGene cluster
Gene symbol
UniGene cluster





TACSTD1
Hs.692
SNAI2
Hs.360174


ABCC4
Hs.508423
GSTP1
Hs.523836


MYO6
Hs.149387
BPAG1/DST
Hs.485616


HPN
Hs.182385
KRT5
Hs.433845


AMACR
Hs.508343
CSTA
Hs.518198


APOC1
Hs.110675
LAMB3
Hs.497636




EPHA2
Hs.171596




CX3CL1
Hs.531668









One of the applications for the gene sets whose expression profiles are capable of discriminating between carcinomatous samples and their normal counterparts is that of predicting whether a prostate tissue sample is carcinomatous or not, a diagnosis that could not have been known in advance. A prerequisite for being able to apply this type of predictive analysis is that said gene set must be capable of discriminating between carcinomatous and noncarcinomatous samples, not only on the basis of the experimental data themselves, but also on the basis of the experimental data of others. In order to discover what was the minimum set of genes, from among the set of 14 genes described above, having sufficient capacity to discriminate between carcinomatous and noncarcinomatous samples, a linear discriminant analysis (LDA) was performed [64]. This is a statistical technique that allows objects to be exhaustively classified into mutually exclusive groups, based on sets of measurable characteristics of such objects. In this case, the point was to classify samples into carcinomatous and noncarcinomatous, using the expression levels of given sets of genes as measurable variables. The ultimate objective was to optimize the set of genes most useful for discriminating between carcinomatous and normal samples. In order to extend the usefulness of this classifying set beyond the 27 experimental samples, data corresponding to another microarray analysis carried out on 57 samples, published by Liu et al. [65], were obtained. In order to be able to apply statistical analysis equally to all the samples, expression data from the 84 samples (27 own samples and the 57 of Liu et al.) were normalized using the RMA method of Irizarry et al. [14], followed by quantile normalization. Next, the samples were randomly distributed into two groups: a training group of 63 samples (75% of all the samples) and a validation, or test, group of 21 samples. Using the training group, all the possible gene-pair combinations from among the 14 genes described above were applied in a cross-validation of the LOOCV type (“leave-one-out cross-validation”), which quantitates the capacity to discriminate between carcinomatous and normal samples when applying LDA as implemented in the R MASS package [66]. From this LOOCV analysis it was found that the gene pair comprising TACSTD 1 and LAMB3 was capable of classifying samples correctly as carcinomatous or normal in 98% of cases. Accordingly, this gene pair was used as the starting point for increasing, in increments of one, the number of genes (from among the 14-gene set or mini-signature), keeping those that gave the best results in the LOOCV test. This process led to a minimum set of seven genes from the mini-signature of 14, which allowed carcinomatous and normal samples to be classified with complete accuracy in an LOOCV analysis, and this worked equally well with data relating to our own samples and to the data of Liu et al. These genes are, from among those overexpressed in tumors: TACSTD1, MYO6, and HPN, and from among those underexpressed in tumors: LAMB3, EPHA2, DST, and CSTA.









TABLE 6







LDA weights for each of the seven genes in the minimum classifying set














Gene
TACSTD1
MYO6
EPHA2
DST
HPN
CSTA
LAMB3





Weight
1.0737
−0.1341
0.5108
−0.0248
0.7580
0.2182
−1.9292









Cut-off point: 7.93


Similarly, a series of 27 paired human prostate samples—i.e. carcinomatous samples and the corresponding normal samples from the same patient—were analyzed by hybridization on 60-mer oligonucleotide microarrays in which the entire human transcriptome was represented. The grading of the carcinomatous samples according to the Gleason scoring system was as follows: 5 samples in Grade 5, 2 samples in Grade 6, 15 samples in Grade 7, 2 samples in Grade 8, and 2 samples in Grade 9. At the same time, 3 samples of stromal tissue were also analyzed. The paired samples were cohybridized after labeling with different fluorochromes. The stroma samples were cohybridized against a pool of normal samples.


This analysis made it possible to identify a set of 15 genes, in addition to the 45 genes identified previously, that would also make it possible to discriminate between carcinomatous samples and normal samples. In particular, this set was made up of the genes CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4. Of these, the genes GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4 were overexpressed, while the genes CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, and SCHIP1 were underexpressed.


In this way, a set of 60 genes was defined that exhibited a high capacity to discriminate between carcinomatous samples and normal samples.


Example 3
Immunohistochemical Technique Used on Tissue Microarrays

The tissue microarrays were constructed using a Beecher instrument (Beecher Instruments) and a 1 mm-diameter needle. Three different microarrays were constructed, containing selected zones of samples of normal prostate, carcinomatous prostate, and PIN tissue, all previously embedded in paraffin. Blocks of lung tissue previously stained with three different colors and placed in different zones of the microarray were used as orientation markers for the samples within the arrays. Complete sections of the microarrays were taken and stained with hematoxylin-eosin to confirm quality. 2 μm thick sections were taken and mounted on xylene-coated glass slides (Dako, Carpinteria, Calif.) for the immunohistochemical stainings. These were done with the Techmate 500 system (Dako), using the Envision system (Dako) for the detection. Briefly, the sections were deparaffinized and rehydrated in graded alcohol series and water. For the detection of MYO6, antigen unmasking was performed in a pressure cooker with citrate buffer (pH 6) for 5 min. This treatment was not done for the EPHA2 and CX3CL1 antigens. Next, the microarrays were incubated for 30 min with the primary antibodies (1:100 dilution for MYO6, mouse monoclonal antibody from Sigma, St. Louis, Mo.; 1:50 dilution for EPHA2, mouse monoclonal antibody from Sigma; and 1:200 dilution for CX3CL1, goat polyclonal antibody from R&D Systems, Minneapolis, Minn.) and washed in ChemMate buffer solution (Dako). The endogenous peroxidase was blocked for 7.5 min in ChemMate peroxidase-blocking solution and then incubated for 30 min with a peroxidase-labeled polymer. After washing in ChemMate buffer solution, the microarrays were incubated with the chromogenic substrate solution diaminobenzidine, washed in water, counterstained with hematoxylin, dehydrated, and mounted.


The results were analyzed by a pathologist. Two aspects of the immunohistochemistries were analyzed: firstly, the percentage of epithelial staining, assessed as between 0 and 100%, and secondly, the intensity of the staining, assessed as none (0), weak (1), moderate (2), or intense (3). The expression patterns of each of the proteins were also analyzed. A sample was considered to exhibit overexpression or underexpression of a protein by comparison with another sample when the percentage difference in epithelial staining between the two samples was greater than 20% and/or the intensity was different by at least one grade.









TABLE 7







Numerical data corresponding to the expression levels of the genes shown in


the first colunm for the samples indicated in the first row














Gene









symbol
PC17
17N
17T
HeLa
RWPE.1
6T
18S





ABCC4
7.01894144
10.4613757
11.6230748
8.37260766
7.50159596
10.8634026
7.09213398


AMACR
8.68108886
7.96926986
12.8351796
8.39624468
7.34379933
10.2225514
7.5281908


APOC1
7.98525042
7.80815015
9.06457735
8.1481063
8.24110628
8.46054204
8.10230296


BIK
8.72048074
9.26375117
9.64501802
7.62614321
7.90914969
10.4011472
7.55835806


BNIP2
8.16566105
7.80079673
7.18051408
8.2173358
8.89602213
7.06987928
8.07765999


BPAG1
10.8672916
7.30166451
6.01919471
5.20003457
8.72091247
6.00368215
5.08879459


CAMKK2
9.737646
9.70184651
10.370831
10.0657809
9.73526508
11.1595365
9.76286829


CDK5
8.568005
7.70409083
8.60235141
9.23623118
8.94813497
8.14872131
7.9070929


CLU
6.51221674
10.8940133
8.93030445
9.9730063
6.84752844
8.4818287
11.5912177


CSTA
11.7195251
8.14146382
6.60991459
5.88939882
10.3088495
6.33161278
5.8571188


CX3CL1
8.88988776
10.0243526
9.00472634
7.4931274
7.37948345
8.76551159
10.3847154


DDR2
7.41767027
8.95529175
8.33788988
8.71221281
7.47395843
8.72937805
9.96868172


EIF3S2
10.7992799
10.561861
10.9027349
10.8741758
11.3019077
10.7176234
10.476417


EPHA2
9.7082Q466
7.49934765
7.16421139
8.11270183
9.75777822
7.38790777
7.47322296


ETS2
9.47553159
9.77888956
8.42741684
9.18178376
8.92309412
7.68369387
8.3554048


FOXA1
9.02905242
10.0313732
10.8553978
7.4232805
6.49892159
10.8676969
6.42851047


FOXF1
7.0642855
9.85003934
8.75526704
7.00736919
6.94349969
8.58135979
10.8835825


FOXO1A
8.91078537
9.03902761
8.24237988
7.92187059
8.23887082
8.23140389
9.00659083


GJA1
11.2901588
11.2772941
9.54483514
5.26350546
7.36129935
9.89041451
11.3069706


GJB1
7.64303997
7.69738283
8.01763073
7.87662152
7.67073411
9.38503063
7.5918705


GSTP1
12.0075611
10.130881
9.04306561
7.89947839
12.2896721
8.76328398
10.3019874


HOXC6
7.18359621
7.06346651
10.8797062
8.99023293
7.79906711
9.02764744
6.93338044


HPN
7.67694465
8.13029835
9.17676877
7.9509903
7.77658452
9.75933907
6.42802676


ICA1
7.40640467
7.8223028
8.27952589
7.02354553
7.24834786
8.32881152
7.28397149


KRTS
13.6426559
10.2949882
8.80835698
7.92292386
13.7036552
9.01522843
7.71264456


LAMB3
12.0274355
7.00889594
6.61191666
7.35030303
8.8988928
6.50613336
6.06671194


LASS2
9.92813099
10.5139364
10.8480268
10.5626413
10.4168201
10.8673543
10.6740624


MYO6
8.88009387
8.08434501
9.99425853
6.03962017
6.67780748
10.2684098
6.92339771


NIT2
9.61978598
8.75482274
9.29367533
10.4865837
10.1578698
8.82951705
8.46774918


NME1
10.8911876
10.0882175
11.0335317
12.907781
11.9095752
10.6540593
8.81423984


NONO
11.8163895
12.0151783
12.2094843
12.5281615
12.0816142
12.5257706
12.0667089


PER2
7.84799328
9.89444514
8.58175271
7.03180711
8.36908903
8.98743391
8.73764612


PP3111
8.44084113
8.38662752
9.02985592
9.17312402
8.62674245
9.27000731
7.96829081


PRDX4
10.315142
10.7094529
11.2458939
12.090736
10.445978
11.0929883
10.0920133


PYCR1
8.19237492
8.25290876
9.25640319
9.79569033
8.82448729
9.07383443
7.68839803


ROR2
6.14361889
6.27470443
6.0394316
6.58708198
5.7583719
6.24186092
6.66175193


SIM2
7.80827202
7.82038456
8.7305589
7.80799199
8.57755541
10.1050121
6.84667269


SNAI2
11.2678841
8.66396438
7.11405551
7.9600653
11.0353323
7.02373053
11.1503284


SND1
10.6173702
10.4907323
10.9569735
11.1427315
11.0964041
11.4362721
10.329777


SYNGR2
11.4162047
11.6941476
12.1766787
10.9151129
8.31027438
11.9141037
10.4248928


TACSTD1
7.46799526
10.0009621
10.8087613
5.989988
7.01683131
11.4197111
6.26163501


TGFBR3
6.2685964
8.35927604
7.03093241
7.66104789
6.57384193
6.86915073
10.2204548


TP73L
11.8613758
8.79341279
7.81792381
6.93485108
10.3875912
7.60044072
6.69592993


ZNF278
7.26124898
7.80704616
8.19354373
7.56542099
8.01614261
9.16707999
8.21306791


ZNF85
7.38699349
7.39620968
7.60984772
7.40574541
7.54405141
7.73752119
7.50789492





Gene


symbol
18T
7N
7T
8T
1S
19T
9N





ABCC4
11.2271232
10.1963771
11.2746849
11.6944641
6.76257618
12.5994078
10.6776932


AMACR
12.1012097
7.98499564
8.38263711
11.2314667
8.07566602
12.2739694
8.10377527


APOCL
8.69754985
7.99587538
8.62266379
8.35568323
8.72869293
8.74838729
8.06936237


BIK
9.7003375
9.63258805
9.92888094
10.0777295
7.40838687
10.9448489
9.61936506


BNIP2
6.89861956
7.58226247
7.22528551
7.17294322
7.74667599
6.6702639
7.25782981


BPAG1
5.15851378
7.96957079
6.24850255
5.19641455
5.05811969
5.01998749
7.17262223


CAMKK2
11.5588663
9.87339804
11.0697585
11.1124115
9.8876309
11.5419852
10.6559128


CDK5
8.4242262
7.99945359
8.14608123
7.94815471
8.22560766
8.67233447
7.9192629


CLU
8.79369781
10.3280124
9.29143062
9.63955038
11.1643832
9.33597842
9.96149275


CSTA
5.77281025
7.95157917
7.25880051
6.09337536
6.34432573
7.24725807
8.49120307


CX3CL1
8.44186449
10.676204
8.63995295
9.08023699
9.15927735
8.44309163
9.86868202


DDR2
8.16745773
9.06621868
8.41740413
8.0789186
10.2486776
8.13530894
8.91851077


EIF3S2
10.9105817
10.3865965
10.6113503
10.5929438
10.3249773
11.0126954
10.2311564


EPHA2
7.17678801
7.89459938
7.15214688
7.33594481
7.44620259
7.14911012
7.56923343


ETS2
6.58405245
9.81248202
8.01769991
8.04258678
7.99424304
7.76639891
8.56305734


FOXA1
10.8011387
10.3592769
10.8530129
10.8797191
6.4191556
10.9823286
10.6645469


FOXF1
8.03302684
9.9046605
8.96329955
8.55239174
10.4449179
8.15703385
9.74120529


FOXO1A
8.54879356
8.68419304
8.59613866
8.26334093
8.95850628
7.81527162
8.51103171


GJA1
8.54551182
11.1134211
10.0046213
9.72701997
11.1550828
9.25171382
10.9683059


GJB1
9.46969306
7.66786411
8.71718679
9.61532346
7.36786826
10.2194731
8.4244208


GSTP1
8.36261156
9.86084926
9.53076751
8.5587529
10.4467656
8.54149475
9.79901801


HOXC6
11.339035
7.27473392
9.34627449
10.7670458
7.19492311
10.3374135
7.24618075


HPN
10.6561446
7.3060213
10.4190497
10.9288653
7.13195771
10.2213461
8.84860895


ICA1
8.25441438
7.63473075
8.15142808
8.29386454
7.399157
8.68291907
7.76154604


KRT5
7.65361029
10.996713
9.95219888
7.87476248
7.69837501
7.44972423
10.9700824


LAMB3
6.24370552
7.34218968
7.00141211
6.24640714
6.27950191
6.30696054
7.12132677


LASS2
11.2221976
10.6505211
10.8624308
11.0606051
10.4854995
10.9970835
10.6469129


MYO6
10.2624067
8.13317622
9.90336178
9.41819058
6.44314341
9.08177328
7.80745759


NIT2
9.34291532
8.67741234
9.15596053
9.43805857
8.52542787
9.82304069
8.6660776


NME1
10.0531402
9.5086149
10.5082895
10.3384312
9.64226277
10.6409689
9.64410232


NONO
12.1516229
11.9984819
12.1125889
12.2838717
12.1986165
12.2481884
11.9934848


PER2
7.47895612
9.99241721
9.09280918
8.02478892
8.93942007
7.96185138
8.78048182


PP3111
9.06774789
8.53884704
9.59167222
9.35656156
8.21257069
9.41421356
9.02282236


PRDX4
11.8467771
10.4503516
11.3533176
11.5130603
9.84736148
11.645151
10.7094521


PYCR1
9.65592092
8.12356225
9.01217629
9.04589705
8.4375687
9.56051736
8.50885394


ROR2
5.81120902
6.4418307
6.09783631
6.17662552
7.04921286
5.82903204
7.01585067


SIM2
9.59602723
7.26551652
8.99252974
9.50626016
7.51297683
9.84470392
8.06004373


SNAI2
7.62802062
9.63303817
7.95774174
7.83829665
11.3259327
7.50489613
8.50108358


SND11
1.4321273
10.5236746
11.1628238
11.3997514
10.6089806
11.1611431
10.9241905


SYNGR2
12.2782432
11.5294606
11.9930355
12.3009771
9.84340774
12.6146706
11.8216618


TACSTD1
11.4203004
10.1107088
10.885747
11.3747192
6.5805044
11.3564186
9.59057446


TGFBR3
7.55120446
8.02542909
7.53921394
7.24467127
8.0425394
7.28185016
7.91328107


TP73L
7.01981993
9.47834975
8.84440292
6.62033628
6.81020202
6.49145679
9.48596872


ZNF278
8.40430883
7.8764079
8.13858036
8.46504808
8.0258282
8.6522079
8.06108653


ZNF85
7.5170014
7.3222536
7.69830094
7.610817
7.55815322
7.58933786
7.3856278





Gene


symbol
9T
20T
10T
PC23
2S
12N
12T





ABCC4
11.9172056
11.8666953
11.8305509
7.00074367
7.73711574
10.7213306
11.8803074


AMACR
11.9137503
12.3148691
12.686271
8.19611317
7.80365344
7.9547764
12.6317357


APOC1
9.25696795
8.46908778
8.74221031
8.24086463
9.09325789
8.21965406
8.17480668


BIK
10.3816475
10.2355408
10.4208791
8.77225842
8.1790476
9.84535521
10.2826195


BNIP2
7.0962776
7.03616692
6.90589636
7.85213858
7.52775825
7.45985555
6.90690277


BPAG1
5.09828283
6.04496312
5.7789774
10.0520177
5.21978918
8.16139572
5.53123132


CAMKK2
11.0854414
10.716776
11.0647166
9.58164524
9.83063525
9.86036006
11.3006859


CDK5
8.18534228
8.30003247
8.33478769
8.26339132
8.1439549
7.74196732
8.21793045


CLU
7.91627628
9.14652294
8.65753757
6.65237257
11.8860275
10.854299
8.32138017


CSTA
6.49862846
6.90773272
5.89922147
11.0523625
6.24277999
8.33648741
6.27402215


CX3CL1
7.99163394
8.88640628
8.15991859
8.56298653
8.66255417
10.5781947
8.61855428


DDR2
8.06360361
8.03792758
8.47150703
7.48635132
10.5650926
9.28002014
8.10022071


ELF3S2
10.8210251
10.9882272
11.0577539
10.7062572
10.2233131
10.2738356
11.1034097


EPHA2
7.21012339
7.06902863
7.29855649
9.86704223
7.33431497
8.01389782
7.26052422


ETS2
7.39836695
7.67103368
7.45179102
8.92145676
8.42658776
9.77172875
7.64306615


FOXA1
11.8216618
10.7574154
10.778755
8.3575482
7.90333524
10.1801846
10.9857394


FOXF1
8.0014319
8.43369605
8.23295854
6.9259667
10.8964535
9.44419125
8.0822311


FOXO1A
8.43249743
7.97606323
8.24438446
8.29401042
8.60797096
8.64648558
8.2992001


GJA1
7.62007523
9.69771636
9.58136618
10.8074789
10.9017509
11.1092323
9.07115329


GJB1
8.1704462
9.79766466
10.1438614
7.53366868
7.91726826
7.40596851
10.2352758


GSTP1
8.27026785
9.22504244
8.87428302
12.1954488
10.3820169
10.3329038
8.58331838


IIOXC6
10.1430958
9.59059802
10.1496822
7.32780121
6.82549442
7.30894113
9.34228709


HPN
10.6062198
10.1904939
10.6235615
7.36632189
7.09007426
8.30883909
11.0701075


ICA1
7.64300295
8.48218665
8.77857582
7.61796929
7.55625552
7.74647255
8.7778574


KRT5
7.56688726
9.90191796
9.0722628
14.0633331
8.58576209
10.8761262
8.85417384


LAMB3
6.23981376
6.65204117
6.73403046
11.8603044
6.41607574
7.76250751
6.67757932


LASS2
11.0625167
11.0266773
10.9537084
10.1227796
10.5322732
10.8868142
11.1010842


MYO6
9.02253898
8.48102562
10.1881512
8.11527081
6.41650094
8.52772725
10.8986622


NIT2
9.91469278
9.23855062
9.37249237
9.34113599
8.35078053
8.33930438
9.50783613


NME1
10.5829942
10.8343772
11.006816
11.0178073
9.50191563
9.49069094
11.2762249


NONO
12.0228516
12.3080217
12.5132073
11.898023
12.1410995
11.9104189
12.4407576


PER2
7.23254734
8.64635518
8.58677335
7.80934006
8.97395062
9.47096163
8.65469168


PP3111
9.2413702
9.66725561
9.45465288
9.0435806
8.96214303
8.47818909
10.4966201


PRDX4
11.4998198
11.636186
11.4769312
10.6760176
9.64941822
10.0816307
11.2610282


PYCR1
9.30310099
9.61332877
9.42395928
8.65802373
8.50150214
7.81547857
10.0492023


ROR2
6.01020411
6.08957047
5.97318364
6.15034361
7.0043655
7.2274926
6.08277438


SIM2
9.22250128
9.75804661
9.56257978
8.18699921
7.41762197
7.6488079
10.0277817


SNAI2
6.38581045
7.37204318
7.17204849
11.1402032
11.620286
8.18863116
7.03459772


SND1
11.4811266
11.2599
11.2294704
10.577137
10.4735652
10.4302397
11.2681063


SYNGR2
12.3733994
12.3232535
12.3456541
11.3345889
9.71075107
11.479208
12.8233699


TACSTD1
10.7356762
11.7266979
11.8309216
7.59472761
6.2337136
10.1461824
11.6233819


TGFBR3
6.75824972
7.11710537
6.87298173
6.38526808
8.39942967
7.99807616
6.78688093


TP73L
6.68778861
8.0879124
7.91161675
11.352239
6.88907193
9.19017183
7.40700292


ZNF278
8.18649067
8.43322063
8.89554478
7.69469089
8.01165956
7.99509545
8.93017065


ZNF85
7.58805419
7.57040324
7.64225531
7.55798388
7.21574937
7.4252656
7.53454756





Gene


symbol
13N
13T
14N
14T
11T
15T
16T





ABCC4
9.87284943
10.7023005
10.4511337
11.8936811
12.0107951
11.8271787
11.819038


AMACR
7.23394577
10.9916346
7.10466664
11.6194729
10.2516113
12.2001907
12.3721041


APOC1
8.10633034
8.62785184
8.10948805
8.72052704
8.74113469
8.7104815
8.45999773


BIK
9.36912096
9.90651551
9.46838994
10.1726757
10.7442765
10.0776735
10.6479084


BNIP2
7.65293558
7.1300569
7.96998676
7.25981586
6.95776104
6.96775088
6.88542751


BPAG1
8.94788313
7.00538757
10.0929067
5.39972453
5.86105537
6.16669793
5.25432662


CAMKK2
10.2352501
10.4407544
10.0805183
10.8321742
10.6S37179
10.5318784
11.389258


CDK5
7.88525167
8.04634018
7.50770634
8.03387344
8.433982
8.33204895
8.80647145


CLU
10.0989244
8.73538286
10.9139483
10.5259445
8.40080486
8.6119414
7.58396723


CSTA
7.61595445
6.98755382
8.71605343
7.03633463
6.60909254
6.51969414
6.50639928


CX3CL1
9.92042383
8.89307138
10.7285592
8.89535509
8.43764378
9.2409657
8.15206923


DDR2
8.26986333
8.05580274
9.07930741
8.99650756
7.9552987
8.05775815
7.55224167


EIF3S2
10.4962482
10.940806
10.3916035
10.6831212
11.0231656
10.9835197
11.0540775


EPHA2
8.28777408
7.62973479
8.34062081
7.21294732
7.17000907
7.38265068
7.08239453


ETS2
10.137787
8.91562764
9.97872769
8.85796935
7.19962252
8.63750489
8.00813095


FOXA1
10.1193691
10.665893
9.9001488
10.8102308
10.9534422
10.6973363
11.5363974


FOXF1
10.4228639
8.78810962
9.52827881
10.2866597
8.06866022
8.64469714
7.49981135


FOXO1A
9.09845272
8.38652775
9.41526303
8.81821241
8.39295341
8.30528633
7.62815549


GJA1
10.9948259
10.3121276
11.5834059
10.4540912
9.4185747
9.70841654
8.91124804


GJB1
8.35342714
9.55748737
7.39028241
8.88234825
10.2712517
9.40653092
10.0348341


GSTP1
9.909S0043
9.04257025
10.3100795
10.0777898
9.00409974
8.98497999
8.60633725


HOXC6
7.23847465
9.99165798
6.97968714
9.0004868
10.5918901
7.36678855
10.8817903


HPN
8.32513868
9.7180687
7.35776409
10.0169966
10.5980528
10.2320083
11.0611131


ICA1
7.75237235
8.27457596
8.02554991
8.06257195
8.49418617
8.39530267
8.47294637


KRT5
10.9508705
9.76644376
12.1629672
7.44473053
9.64157957
9.46641915
8.45819028


LAMB3
7.38026052
6.S9S74533
9.06273995
6.39364642
6.59601678
6.55864806
6.63258024


LASS2
10.7164064
10.8330488
10.4867107
11.1811361
11.095375
10.9263813
10.7735895


MYO6
7.53960908
9.72015889
8.07733834
8.70022299
10.6315373
9.17690886
8.03484288


NIT2
8.31073282
8.88759289
8.19132807
8.87114075
9.89397532
9.18315645
9.74235571


NME1
9.94852791
10.5155577
10.5564837
10.2727076
10.5542469
10.3195047
11.3315375


NONO
12.055439
12.2627147
11.7064367
11.9732265
12.5242129
12.2909779
12.4518149


PER2
10.0941933
8.76292852
10.3200602
8.87004769
8.38657379
8.74722305
8.09258734


PP3111
8.63022636
9.14033715
8.72580833
9.29615413
9.40440095
9.70281354
9.83199096


PRDX4
10.4308487
11.3358816
10.0641533
10.6523311
11.3983874
11.1376659
11.8621885


PYCR1
8.39293341
9.19236757
7.96330584
9.2152436
9.12708041
9.32463429
9.55066892


ROR2
6.90060538
6.13096573
6.50870306
6.18072898
5.88782188
6.24507223
5.84650771


SIM2
8.27627143
8.64331076
7.92024044
8.62597651
9.90131372
9.47502981
10.8614305


SNAI2
9.08242588
7.8449708
8.76110432
7.82298177
7.15025613
6.89855193
6.07213975


SND1
10.7021338
10.6168193
10.850709
10.7493318
11.4544134
11.2395792
11.4547764


SYNGR2
11.6317612
11.9190577
11.2983082
12.1101888
12.043799
12.279038
12.7500744


TACSTD1
10.0066892
11.5231648
10.3110571
11.277493
11.8558022
11.2057988
11.5306351


TGFBR3
8.63782836
7.77760712
7.67851529
8.08520398
7.13647148
7.27343835
6.87345209


TP73L
9.01973146
7.89492281
9.72592518
6.60008695
7.81793676
7.9384222S
7.38255717


ZNF278
7.79027079
8.33786475
7.19809893
7.91250759
8.85698728
8.59516602
8.46917838


ZNF85
7.2808244
7.70416086
7.07571111
7.73406311
7.64220123
7.82936227
7.66607631





Gene


symbol
21N
21T
3T
4T
22T
5T
PoolN





ABCC4
10.7987406
11.3728689
11.5757011
12.0648794
11.2786267
11.9156492
10.3454557


AMACR
8.69577257
11.99929
11.0447135
10.1108062
12.8839261
11.5908069
7.98152182


APOCL
8.52264152
8.97659989
8.9263806
8.50670267
8.85075098
8.72836793
8.16064021


BIK
9.77191275
10.3833188
10.197079
11.1866636
9.99644752
10.5948164
9.52731013


BNIP2
7.23818697
7.13906841
7.23818697
7.01612877
7.00613392
6.98368469
7.37653792


BPAG1
7.2381955
5.8171571
6.45538392
5.23069383
5.92832715
5.90277552
8.01945315


CAMKK2
10.7747453
11.2779526
10.886789
11.08344
10.59382
11.305732
10.0967028


CDK5
7.95270947
8.46120132
8.25484113
8.4811559
8.12246741
8.54523175
7.81283032


CLU
9.35268604
8.51217761
8.37356534
7.48221854
8.57705227
8.9700039
10.5958875


CSTA
7.9531818
6.84973913
6.7888125
5.73748113
6.8515908
7.00949205
8.4014576


CX3CL1
9.02463229
8.55932974
9.20807982
7.76909535
8.57102024
9.12691829
10.0929465


DDR2
8.74803007
8.41740413
8.42036901
7.99142192
8.57332702
8.29318467
8.96671113


EIF3S2
10.5401121
11.1064603
10.7196193
10.8775701
10.4832358
10.9083898
10.3394479


EPHA2
7.68132884
7.28836732
7.43604758
7.25549484
7.35055576
7.33056725
7.75125282


ETS2
8.69471469
7.53562961
8.73896304
6.59356647
7.65836985
8.761367
9.00524251


FOXA1
10.7093697
10.7503471
10.7138659
11.2053323
10.7143607
10.7517395
10.3477084


FOXF1
9.21318089
8.32823723
8.61439456
6.80094862
8.20420908
8.04615239
9.96185792


FOXO1A
8.59994547
8.55377222
8.42533384
8.23321695
8.23364541
8.16604594
8.65463795


GJA1
10.9466858
9.89211284
10.0047312
6.31306283
9.82556202
9.93968212
11.1323778


GJB1
8.48612454
9.11576612
9.04990338
9.00338667
8.57846816
9.35583043
8.05627572


GSTP1
9.84513277
9.11867146
9.04897591
10.3268912
9.03178533
9.00149776
10.2176458


HOXC6
7.1017041
10.8908235
8.49521171
8.40720678
11.1104435
9.77780259
7.36191248


HPN
8.31609284
9.64575
9.81476603
10.2938254
9.30031929
9.4467024
8.09737091


ICAl
8.08266807
8.5863091
8.17044223
8.49784086
7.97177787
8.61104083
7.83970432


KRT5
10.9449876
9.08366066
9.75663217
8.14317321
9.57025743
9.49768678
11.236332


LAMB3
7.5548769
6.71572298
6.75502602
6.63929214
6.9458567
6.73719738
7.72972549


LASS2
10.7349071
11.0806776
10.7263753
10.8542195
10.8732259
10.9581809
10.4390986


MYO6
8.07722331
10.3375571
9.15013634
9.88691913
9.86050889
8.78526937
7.73393447


NIT2
8.30010875
9.19644646
8.57290557
9.58081927
8.62370167
9.02821281
8.47501495


NME1
10.017011
10.543603
10.2393309
11.1049441
10.4691113
10.7463599
9.90349677


NONO
12.1318017
12.5090225
12.2283024
12.3650511
12.2359642
12.1606526
11.9220344


PER2
9.42182334
8.76605844
8.95087367
8.35302696
8.38888943
9.02323837
9.34269673


PP3111
9.1081155
9.30457173
9.2177844
9.46601388
9.01106612
9.37842758
8.60821857


PRDX4
10.7020948
10.914888
11.1667704
11.2168946
10.9077281
11.465411
10.5237508


PYCR1
8.83464147
9.14088089
9.05983097
10.054324
9.24170896
9.529762
8.5357301


ROR2
6.31673771
6.09553418
6.18072898
6.03787627
6.19022785
5.81955849
6.50487963


SIM2
8.50815671
10.0231992
9.87971696
8.57728725
8.37394598
9.50571774
7.8807152


SNAI2
9.52076395
7.41224256
7.53950566
6.15634517
7.72468052
6.64439486
9.51616646


SND1
11.0489804
11.0917374
11.150879
11.7032092
10.9957737
11.0958036
10.7184297


SYNGR2
12.0176656
12.2420085
12.0624561
12.5872779
12.0864775
12.2906617
11.5631324


TACSTD1
10.7426884
11.3855844
11.315745
11.1602475
10.9368481
10.8953779
10.1764533


TGFBR3
7.11821992
6.8769741
6.72381038
6.67171728
6.82802005
7.32632724
8.88514945


TP73L
9.6250737
8.22036617
8.41237385
7.18759024
8.3260262
7.60799673
9.30445875


ZNF278
8.22884119
8.70860977
8.45462287
9.11172891
8.35058651
8.31310439
7.76028418


ZNF85
7.53391479
7.57548749
7.56171777
7.5835714
7.54405141
7.6510772
7.28360868





Gene


symbol
N100
T100
N101
T101
N102
T102
N103





CRYAB
304.03901
121.3244
234.84571
121.4657
184.40535
91.079765
267.03763


CYP27A1
65.723462
57.255932
118.45414
82.231491
86.129712
54.551266
161.1998


FGF2
72.28465
41.683247
110.79263
58.439855
58.822512
40.285734
96.177302


IKL
221.3674
134.85724
400.0126
243.9671
282.48142
173.30401
386.17055


PTGIS
84.010471
35.41209
92.180097
56.645933
63.568096
36.374147
96.028576


RARRES2
312.56608
118.36257
729.56327
171.82879
338.47124
122.62955
463.14646


PLP2
154.71311
98.26283
245.75441
179.81484
229.28739
160.83719
439.03806


TPM2
2191.002
630.14343
4733.4967
1665.5474
2238.5064
785.52533
1661.7838


S100A6
1102.7169
183.10162
936.06551
221.11275
917.98938
170.42856
761.10048


SCHIP1
56.385515
34.697276
85.153
51.091379
52.04548
34.915501
85.972245





Gene


symbol
T103
N104
T104
N105
T105
N106
T108





CRYAB
171.61672
374.1016
155.45401
497.87553
248.7899
254.07934
150.41964


CYP27A1
151.10053
115.91874
86.909619
248.54438
128.62298
197.9181
100.13125


FGF2
79.265714
84.667572
50.390227
85.61306
60.081732
78.62001
58.801731


IKL
285.44466
423.89278
240.43763
455.70408
330.25223
399.36471
300.49221


PTGIS
53.516357
81.416804
50.173377
115.50284
66.058047
107.81833
59.635915


RARRES2
300.75048
798.979
238.58758
587.93406
284.88557
548.42525
300.65621


PLP2
387.2087
217.1716
131.95625
332.56625
204.42778
279.81481
212.40802


TPM2
888.89642
2518.3831
931.75881
3691.8294
1537.8768
2022.4243
986.66895


S100A6
603.12132
1048.5913
241.93582
1074.0259
360.27407
1179.3059
493.35684


SCHIP1
76.562008
74.506549
47.691502
87.661258
56.415572
70.72289
45.462556





Gene


symbol
N109
T109
N110
T110
N111
T111
N112





CRYAB
728.10238
334.53896
462.08795
325.6897
509.11036
371.00667
173.81801


CYP27A1
239.02055
171.51142
201.60599
166.9975
242.08551
138.72297
127.96567


FGF2
167.55178
82.164604
105.73767
73.756409
121.12422
93.544057
60.320056


IKL
767.354
427.54557
767.98898
569.37903
601.85105
406.48678
229.16381


PTGIS
193.34741
83.867162
89.608846
64.023175
140.22699
80.861076
52.803617


RARRES2
1039.5743
422.29068
825.92271
637.63234
551.73661
452.50005
259.1351


PLP2
498.36522
395.31156
499.32137
476.76522
203.51874
144.79145
174.97119


TPM2
6456.0272
2111.102
5667.8325
3468.241
3276.7168
1659.5055
763.64856


S100A6
1634.893
524.18791
882.00173
593.89223
844.76849
696.22009
766.88887


SCHIP1
105.11061
57.558811
73.649872
63.114436
97.658762
63.515854
49.88602





Gene


symbol
T112
N113
T113
N114
T114
N115
T115





CRYAB
83.552528
338.66144
268.3403
170.68736
95.326169
284.52504
131.44346


CYP27A1
61.264804
206.98048
167.49587
121.3418
73.254299
47.575277
54.935472


FGF2
44.056521
65.584328
72.118586
70.591939
54.65119
69.653036
35.727261


IKL
153.96913
481.11944
405.45231
309.17561
280.90344
585.95996
304.44269


PTGIS
38.949907
63.680412
62.686521
74.588854
49.470357
78.316363
38.359732


RARRES2
111.87978
479.48903
471.57935
334.73814
244.31686
320.62506
165.23301


PLP2
111.66308
515.79167
416.21092
235.00366
199.09064
200.63167
134.02652


TPM2
316.99325
2595.0592
2091.2962
1324.363
867.44314
3486.8114
1054.9162


S100A6
157.90255
788.23454
717.07387
578.12705
469.57977
692.50989
178.17158


SCHIP1
36.655192
66.331927
57.880973
51.016331
38.812838
69.387065
35.437729





Gene


symbol
N116
T116
N117
T117
N118
T118
N119





CRYAB
404.0817
224.14001
324.66653
183.91509
311.92158
173.88265
509.32084


CYP27A1
166.82862
123.14593
163.47428
101.23399
119.90321
83.404239
206.19193


FGF2
83.608587
56.545679
50.133584
46.066545
102.23329
72.861872
144.89283


IKL
691.66921
433.76789
270.08383
250.19643
603.19588
430.88638
707.92432


PTGIS
102.36587
60.830401
56.524932
45.672466
102.9645
64.393988
181.16084


RARRES2
438.74004
232.52393
346.64738
308.90796
445.70099
339.66558
769.15839


PLP2
313.58729
233.63584
239.00368
178.31707
330.65921
289.6501
505.22801


TPM2
3810.2639
1896.3882
1290.6817
825.31944
3661.7346
1876.8149
4930.2728


S100A6
495.94026
257.83217
775.83832
488.69973
659.1014
536.45245
1271.7202


SCHIP1
67.144814
46.260516
45.5373
39.669492
72.731855
50.388148
95.127395





Gene


symbol
T119
N120
T120
N121
T121
N122
T122





CRYAB
241.30719
446.74579
300.32495
263.33073
171.00752
226.35029
139.75733


CYP27A1
113.54148
193.60243
192.93733
152.37572
101.91163
114.81597
68.309029


FGF2
69.485908
99.866694
75.195846
83.023188
87.083491
58.20828
53.240264


IKE
380.78091
705.61056
471.81067
265.87455
270.65707
312.68542
210.22655


PTGIS
70.422099
112.50089
68.878482
109.64089
82.782316
62.459911
48.049222


RARRES2
291.10814
863.59416
514.19645
708.29159
433.76044
297.55677
181.44196


PLP2
294.80086
430.17805
325.05215
228.68108
185.65522
53.52592
184.06683


TPM2
1667.1768
3937.2521
2037.4962
1793.6045
1280.9221
2831.2083
1327.8201


S100A6
368.15877
1077.4262
561.16506
1248.9698
519.31155
960.89695
314.64589


SCHIP1
59.308766
78.53731
54.949183
67.355968
50.068293
57.790933
47.563658





Gene


symbol
N123
T123
N124
T124
N125
T125
N126





CRYAB
453.42686
232.36527
154.08888
86.065023
374.12397
197.06421
199.74463


CYP27A1
171.35438
87.607702
99.263988
60.668192
128.592
125.32549
91.63633


FGF2
74.759799
57.036951
51.051763
40.438376
70.984813
51.011261
50.279591


IKL
676.1967
405.68007
167.84299
156.55068
442.20703
320.43114
253.02734


PTGIS
115.24549
56.431897
47.922418
42.698377
77.401207
50.167145
60.607752


RARRES2
843.92249
516.55354
358.35384
162.12343
566.59471
366.30283
198.17521


PLP2
331.18995
230.86984
161.20299
108.45643
342.31056
271.16785
163.7978


TPM2
3138.1606
1292.5339
932.31845
541.75693
2703.1059
1355.6778
1450.0833


S100A6
1122.4826
517.65282
775.07552
207.50308
972.50468
500.55534
632.23848


SCHIP1
67.183433
48.259538
44.262964
34.305785
58.242449
45.656086
47.310421





Gene


symbol
T126
N127
T127
POOL N
ESTROMA





CRYAB
96.129124
568.2609
288.78575
400.01287
201.15131


CYP27A1
69.303684
199.1946
135.93459
259.07487
139.26804


FGF2
49.630356
145.89982
71.376178
104.04685
77.537005


IKL
170.58396
922.70221
548.99246
538.43632
484.72469


PTGIS
41.592946
202.68193
76.223101
89.65319
58.084206


RARRES2
150.20819
1656.6418
737.39486
648.0575
478.11455


PLP2
111.89183
603.96989
349.01458
355.24807
269.24984


TPM2
549.3584
6958.1883
2091.051
3058.8217
1619.1178


S100A6
311.77816
2505.4578
817.71672
809.53278
522.70854


SCHIP1
37.328461
141.11389
72.216726
101.79522
63.256115





Gene


symbol
N100
T100
N101
T101
N102
T102
N103





GOLPH2
548.567
1758.116
428.472
696.334
306.976
429.274
1299.593


TRIM36
40.884
61.160
58.731
95.813
39.120
51.372
58.876


POLD2
242.189
342.861
316.706
398.170
267.462
376.412
374.818


CGREF1
43.909
52.016
61.246
78.168
43.138
62.443
39.320


HSD17B4
151.061
522.392
190.849
373.063
114.272
230.710
284.384





Gene


symbol
T103
N104
T104
N105
1105
N106
T108





GOLPH2
2295.108
359.972
884.085
509.311
1100.512
759.342
1447.196


TRIM36
85.077
56.296
66.954
67.421
78.446
47.790
50.417


POLD2
450.260
357.294
536.214
393.577
527.031
358.295
348.346


CGREF1
47.306
57.871
65.770
59.351
91.547
46.585
57.421


HSD17B4
455.514
186.581
229.884
246.901
329.792
374.754
484.743





Gene


symbol
N109
T109
N110
T110
N11
T11
N112





GOLPH2
1432.141
3919.071
649.470
1278.107
1244.365
1734.654
656.235


TRIM36
56.340
62.612
48.855
55.145
87.472
146.990
46.183


POLD2
786.995
848.817
736.788
920.814
606.222
837.522
292.656


CGREF1
66.247
112.783
51.450
77.906
60.020
76.655
45.815


HSD17B4
350.737
550.721
503.240
545.812
474.732
1302.274
275.292





Gene


symbol
T112
N113
T113
N114
T114
N115
T115





GOLPH2
1629.113
426.814
942.228
1069.926
992.021
461.324
336.536


TRIM36
69.817
52.169
61.860
44.424
49.223
40.542
47.187


POLD2
326.058
479.164
478.057
429.589
482.852
292.033
491.058


CGREF1
66.490
45.644
49.936
38.975
47.778
55.975
100.695


HSD17B4
786.224
283.142
509.154
189.126
292.380
86.714
384.498





Gene


symbol
N116
T116
N117
T117
N118
T118
N119





GOLPH2
532.229
725.970
562.227
840.270
1625.833
1814.714
1091.513


TRIM36
51.842
43.305
45.194
50.729
68.011
60.314
67.471


POLD2
405.684
499.330
335.409
402.676
570.865
717.750
620.046


CGREF1
46.016
73.956
46.021
50.005
56.845
67.400
57.579


HSD17B4
266.058
220.659
227.399
199.026
322.132
356.184
468.237





Gene


symbol
T119
N120
1120
N121
1121
N122
T122





GOLPH2
1389.390
729.486
2332.638
510.619
1687.893
317.782
921.068


TRIM36
155.958
49.435
70.731
50.641
63.975
38.919
49.653


POLD2
713.931
558.768
678.486
398.448
508.459
213.390
270.514


CGREF1
87.276
47.268
50.585
59.108
77.716
35.360
46.984


HSD17B4
1435.151
362.516
492.994
181.192
296.955
122.611
244.115





Gene


symbol
N123
T123
N124
T124
N125
T125
N126





GOLPH2
883.508
1148.884
312.777
1057.729
532.073
1256.653
329.977


TRIM36
41.809
48.110
35.446
42.212
43.216
60.862
45.204


POLD2
409.810
476.585
217.032
288.738
405.953
538.819
217.191


CGREF1
39.376
52.814
37.019
42.407
41.218
51.147
39.985


HSDI7B4
233.520
216.113
100.211
154.418
208.994
426.945
133.675





Gene


symbol
T126
N127
T127
POOL N
ESTROMA





GOLPH2
219.806
554.238
3034.527
2144.808
3374.781


TRIM36
64.658
47.924
116.552
94.617
125.958


POLD2
329.164
498.836
1026.307
659.505
772.858


CGREFI
41.370
44.067
62.324
73.421
110.461


HSD17B4
297.370
355.714
763.187
396.644
1007.737









REFERENCES



  • 1. Brawley O W, et al. (1998). The epidemiology of prostate cancer part I: descriptive epidemiology. Semin. Urol. Oncol. 16, 187-192.

  • 2. Simard J, et al. (2002). Perspective: Prostate cancer susceptibility genes. Endocrinology 143, 2029-2040.

  • 3. DeMarzo A M, Nelson W G, Isaacs W B, Epstein J I (2003). Pathological and molecular aspects of prostate cancer. Lancet 361, 955-964.

  • 4. Rubin M A, et al. (2004). Quantitative determination of expression of the prostate cancer protein α-methylacyl-CoA racemase using automated quantitative analysis (AQUA). Am. J. Pathol. 163, 831-840.

  • 5. van't Veer L J, et al. (2002). Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530-536.

  • 6. van de Vijver M J, et al. (2002). A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 347, 1999-2009.

  • 7. Lapointe J, et al. (2004). Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc. Natl. Acad. Sci. USA 101, 811-816.

  • 8. Rhodes D R, et al. (2002). Meta-analysis of microarrays: interstudy validation of gene expression profiles reveals pathway dysregulation in prostate cancer. Cancer Res. 62, 4427-4433.

  • 9. Sancho E, et al. (1998). Role of UEV-1, an inactive variant of the E2 ubiquitin-conjugating enzymes, in in vitro differentiation and cell cycle behavior of HT-29-M6 intestinal mucosecretory cells. Mol. Cell. Biol. 18, 576-589.

  • 10. Benedit P, et al. (2001). PTOV1, a novel protein overexpressed in prostate cancer containing a new class of protein homology blocks. Oncogene 20, 1455-1464.

  • 11. Santamaria A, et al. (2003). PTOV-1, a novel protein overexpressed in prostate cancer, shuttles between the cytoplasm and the nucleus and promotes entry into the S phase of the cell division cycle. Am. J. Pathol. 162, 897-905.

  • 12. Santamaria A, et al. (2005). PTOV1 enables the nuclear translocation and mitogenic activity of Flotillin-1, a major protein of lipid rafts. Mol. Cell. Biol. 25, 1900-1911.

  • 13. Lozano J J, et al. (2005). Dual activation of pathways regulated by steroid receptors and peptide growth factors in primary prostate cancer revealed by Factor Analysis of microarray data. BMC Genomics 6, 109.

  • 14. Irizarry, R. A., et al. (2003). Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics, 4, 249-264.

  • 15. Bolstad, B. M., et al. (2003). A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19, 185-193.

  • 16. Tukey, J. W. (1977). Exploratory data analysis. Addison-Wesley.

  • 17. Storey, J. D. and Tibshirani, R. (2003). Statistical significance for genomewide studies. Proc Natl Acad Sci USA 100, 9440-9445.

  • 18. Benjamin, Y., y Hochberg, Y. (1995). Controlling the False Discovery Rate—a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B-Methodological, 57, 289-300.

  • 19. Magee J A, et al. (2001).Expression profiling reveals hepsin overexpression in prostate cancer. Cancer Res. 61, 5692-5696.

  • 20. Welsh J B, et al. (2001). Analysis of gene expression identifies candidate markers and pharmacological targets in prostate cancer. Cancer Res. 61, 5974-5978.

  • 21. Dhanasekaran S M, et al. (2001). Delineation of prognostic biomarkers in prostate cancer. Nature 412, 822-826.

  • 22. Klezovitch O, et al. (2004). Hepsin promotes prostate cancer progression and metastasis. Cancer Cell 6, 185-195.

  • 23. Jiang Z, et al. (2001). P504S: a new molecular marker for the detection of prostate carcinoma. Am. J. Surg. Pathol. 25, 1397-1404.

  • 24. Rubin M A, et al. (2002). alpha-Methylacyl coenzyme A racemase as a tissue biomarker for prostate cancer. JAMA 287, 1662-1670.

  • 25. Luo J, et al. (2002). Alpha-methylacyl-CoA racemase: a new molecular marker for prostate cancer. Cancer Res. 62, 2220-2226.

  • 26. Beach R, et al. (2002). P504S immunohistochemical detection in 405 prostatic specimens including 376 18-gauge needle biopsies. Am. J. Surg. Pathol. 26, 1588-1596.

  • 27. Evans A J (2003). Alpha-methylacyl CoA racemase (P504S): overview and potential uses in diagnostic pathology as applied to prostate needle biopsies. J. Clin. Pathol. 56, 892-897.

  • 28. Jiang Z, Woda B A (2004). Diagnostic utility of alpha-methylacyl CoA racemase (P504S) on prostate needle biopsy. Adv. Anat. Pathol. 11, 316-321.

  • 29. DeYoung M P, et al.(2003). Identification of Down's syndrome critical locus gene SIM2-s as a drug therapy target for solid tumors. Proc. Natl. Acad. Sci. USA 100, 4760-4765.

  • 30. Ratan R R (2003). Mining genome databases for therapeutic gold: SIM2 is a novel target for treatment of solid tumors. Trends Pharmacol. Sci. 24, 508-510.

  • 31. Aleman M J, et al.(2005). Inhibition of Single Minded 2 gene expression mediates tumor-selective apoptosis and differentiation in human colon cancer cells. Proc. Natl. Acad. Sci. USA 102, 12765-12670.

  • 32. Bijl J, et al. (1996). Expression of HOXC4, HOXC5, and HOXC6 in human lymphoid cell lines, leukemias, and benign and malignant lymphoid tissue. Blood 87, 1737-1745.

  • 33. Alami Y, et al. (1999). HOXC5 and HOXC8 expression are selectively turned on in human cervical cancer cells compared to normal keratinocytes. Biochem. Biophys. Res. Commun. 257, 738-745.

  • 34. Waltregny D, et al. (2002). Overexpression of the homeobox gene HOXC8 in human prostate cancer correlates with loss of tumor differentiation. Prostate 50, 162-169.

  • 35. Miller G J, et al. (2003). Aberrant HOXC expression accompanies the malignant phenotype in human prostate. Cancer Res. 63, 5879-5888.

  • 36. Ramachandran S, et al. (2005). Loss of HOXC6 expression induces apoptosis in prostate cancer cells. Oncogene 24, 188-198.

  • 37. Lee W H, et al. (1994). Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc. Natl. Acad. Sci. USA 91, 11733-11737.

  • 38. Lin X, et al. (2001). GSTP1 CpG island hypermethylation is responsible for the absence of GSTP1 expression in human prostate cancer cells. Am. J. Pathol. 159, 1815-1826.

  • 39. Yang A, et al.(2002). On the shoulders of giants: p63, p73 and the rise of p53. Trends Genet. 18, 90-95.

  • 40. Signoretti S, et al. (2000). p63 is a prostate basal cell marker and is required for prostate development. Am. J. Pathol. 157, 1769-1775.

  • 41. Yoshida H, et al. (2004). Lessons from border cell migration in the Drosophila ovary: A role for myosin VI in dissemination of human ovarian cancer. Proc. Natl. Acad. Sci. USA 101, 8144-8149.

  • 42. Varambally S, et al. (2005). Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression. Cancer Cell 8, 393-406.

  • 43. Andres A C, et al. (1994). Expression of two novel eph-related receptor protein tyrosine kinases in mammary gland development and carcinogenesis. Oncogene 9, 1461-1467.

  • 44. Nemoto T, et al. (1997). Overexpression of protein tyrosine kinases in human esophageal cancer. Pathobiology 65, 195-203.

  • 45. Walker-Daniels J, et al. (1999)Overexpression of the EphA2 tyrosine kinase in prostate cancer. Prostate 41, 275-280.

  • 46. Ogawa K, et al. (2000). The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19, 6043-6052.

  • 47. Zelinski D P, et al. (2001). EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res. 61, 2301-2306.

  • 48. Straume O, Akslen L A (2002). Importance of vascular phenotype by basic fibroblast growth factor, and influence of the angiogenic factors basic fibroblast growth factor/fibroblast growth factor receptor-1 and ephrin-A1/EphA2 on melanoma progression. Am. J. Pathol. 160, 1009-1019.

  • 49. Miyazaki T, et al. (2003). EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. Int. J. Cancer 103, 657-663.

  • 50. Kinch M S, et al. (2003). Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin. Cancer Res. 9, 613-618.

  • 51. Zeng G, et al. (2003). High-level expression of EphA2 receptor tyrosine kinase in prostatic intraepithelial neoplasia. Am. J. Pathol. 163, 2271-2276.

  • 52. Koffman K T, et al. (2003). Differential EphA2 epitope display on normal versus malignant cells. Cancer Res. 63, 7907-7912.

  • 53. Saito T, et al. (2004). Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol. Rep. 11, 605-611.

  • 54. Duxbury M S, et al. (2004). EphA2: a determinant of malignant cellular behavior and a potential therapeutic target in pancreatic adenocarcinoma. Oncogene 23, 1448-1456.

  • 55. Fox B P, Kandpal R P (2004). Invasiveness of breast carcinoma cells and transcript profile: Eph receptors and ephrin ligands as molecular markers of potential diagnostic and prognostic application. Biochem. Biophys. Res. Commun. 318, 882-892.

  • 56. Wu D, et al. (2004). Prognostic value of EphA2 and EphrinA-1 in squamous cell cervical carcinoma. Gynecol. Oncol. 94, 312-319.

  • 57. Thaker P H, et al. (2004). EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin. Cancer Res. 10, 5145-5150.

  • 58. Nakamura R, et al. (2005). EPHA2/EFNA1 expression in human gastric cancer. Cancer Sci. 96, 42-47.

  • 59. Herrem C J, et al. (2005). Expression of EphA2 is prognostic of disease-free interval and overall survival in surgically treated patients with renal cell carcinoma. Clin. Cancer Res. 11, 226-231.

  • 60. Hajra K M, et al. (2002). The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res. 62, 1613-1618.

  • 61. Uchikado Y, et al. (2005). Slug expression in the E-cadherin preserved tumors is related to prognosis in patients with esophageal squamous cell carcinoma. Clin. Cancer Res. 11, 1174-1180.

  • 62. Elloul S, et al. (2005). Snail, Slug, and Smad-interacting protein 1 as novel parameters of disease aggressiveness in metastatic ovarian and breast carcinoma. Cancer 103, 1631-1643.

  • 63. Martin T A, et al. (2005).Expression of the transcription factors snail, slug, and twist and their clinical significance in human breast cancer. Ann. Surg. Oncol. 12, 488-496.

  • 64. Venables, W. N. & Ripley, B. D. (2002). Modern Applied Statistics with S. Fourth Edition. Springer, N.Y.

  • 65. Liu, P., et al. (2006). Sex-Determining Region Y Box 4 Is a Transforming Oncogene in Human Prostate Cancer Cells. Cancer Research 66, 4011-4019.

  • 66. R Development Core Team (2006). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. http://www.R-project.org.


Claims
  • 1. A method for the molecular diagnosis of prostate cancer, the method comprising analysis of expression levels of at least two genes selected from the group consisting of TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4, wherein the capacity to discriminate between carcinomatous and noncarcinomatous samples when the expression levels of said selected genes are determined together is greater than the discriminating capacity of the selected genes separately.
  • 2. The method as claimed in claim 1, wherein the at least two genes are selected from the group consisting of TACSTD1, HPN, AMACR, APOC1, GJB1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, MYO6, and ABCC4.
  • 3. The method as claimed in claim 1, wherein the at least two genes are selected from the group consisting of TACSTD1, HPN, AMACR, APOC1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, MYO6, and ABCC4.
  • 4. The method as claimed in claim 1, wherein the at least two genes are selected from the group consisting of TACSTD1, HPN, DST, CSTA, LAMB3, EPHA2, and MYO6.
  • 5. The method as claimed in claim 1, wherein at least one of the genes selected from the group is the gene MYO6.
  • 6. The method as claimed in claim 1, wherein at least one of the genes selected from the group is the gene ABCC4.
  • 7. The method as claimed in claim 5, wherein the analysis of the expression level of the gene MYO6 is combined with the analysis of the expression level of at least one gene from the group consisting of ABCC4, AMACR, BIK, BNIP2, CDK5, CSTA, DST, EIF3S2, EPHA2, ETS2, GJB1, HPN, NIT2, PYCR1, ROR2, TACSTD1, and TP73L.
  • 8. The method as claimed in claim 6, wherein the analysis of the expression level of the gene ABCC4 is combined with the analysis of the expression level of at least one gene from the group consisting of CSTA, GJB1, GSTP1, HOXC6, HPN, LAMB3, MYO6, PRDX4, and TP73L.
  • 9. The method as claimed in claim 1, wherein the analysis of the expression level of said genes is performed by determining the level of mRNA derived from their transcription.
  • 10. The method as claimed in claim 9, wherein the analysis comprises amplification by PCR, RT-PCR, RT-LCR, SDA, or any other method of nucleic acid amplification.
  • 11. The method as claimed in claim 9, wherein the analysis is performed by DNA chips produced with oligonucleotides deposited by any procedure.
  • 12. The method as claimed in claim 9, wherein the analysis is performed by DNA chips produced with oligonucleotides synthesized in situ by means of photolithography or by any other procedure.
  • 13. The method as claimed in claim 9, wherein the analysis is performed by in situ hybridization using specific probes labeled by any labeling method.
  • 14. The method as claimed in claim 9, wherein the analysis is performed by gel electrophoresis.
  • 15. The method as claimed in claim 14, wherein the analysis is performed by means of membrane transfer and hybridization with a specific probe.
  • 16. The method as claimed in claim 9, wherein the analysis is performed by means of NMR or any other diagnostic imaging technique.
  • 17. The method as claimed in claim 16, wherein the analysis is performed using paramagnetic nanoparticles or any other type of detectable nanoparticles functionalized with antibodies or by any other means.
  • 18. The method as claimed in claim 1, wherein the analysis of the expression level of said genes is performed by determining the level of protein encoded by the gene or fragments thereof.
  • 19. The method as claimed in claim 18, wherein the analysis is performed by means of incubation with a specific antibody.
  • 20. The method as claimed in claim 19, wherein the analysis is performed by means of a Western blot method.
  • 21. The method as claimed in claim 19, wherein the analysis is performed by means of immunohistochemistry.
  • 22. The method as claimed in claim 18, wherein the analysis is performed by means of gel electrophoresis.
  • 23. The method as claimed in claim 18, wherein the analysis is performed by means of protein chips.
  • 24. The method as claimed in claim 18, wherein the analysis is performed by means of ELISA or any other enzymatic method.
  • 25. The method as claimed in claim 18, wherein the analysis is performed by means of NMR or any other diagnostic imaging technique.
  • 26. The method as claimed in claim 25, wherein the analysis is performed using paramagnetic nanoparticles or any other type of detectable nanoparticles functionalized with antibodies or by any other means.
  • 27. A kit for the molecular diagnosis of prostate cancer, the kit comprising: means for determining an expression level of a first gene, said gene elected from the group consisting of TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4, andmeans for determining an expression level of a second gene, different from the first gene, said second gene independently selected from the group consisting of TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4,wherein the ability to diagnose prostate cancer, when the expression levels of the selected genes are determined together, is greater than the diagnostic ability of the selected genes separately.
  • 28. The method as claimed in claim 1, wherein overexpression of gene or genes MYO6, TACSTD1, or HPN, or underexpression of gene or genes DST, CSTA, LAMB3, or EPHA2 is used for diagnosing presence of prostate cancer or of a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.
  • 29. The method as claimed in claim 1, wherein overexpression of gene or genes MYO6, ABCC4, TACSTD1, HPN, AMACR, or APOC1, or underexpression of gene or genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, or EPHA2 is used for diagnosing presence of prostate cancer or of a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.
  • 30. The method as claimed in claim 1, wherein overexpression of gene or genes MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, or GJB1, or underexpression of gene or genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2 , GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, or ETS2 is used to diagnose prostate cancer or a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.
  • 31. The method as claimed in claim 1, wherein overexpression of gene or genes MYO6, ABCC4, TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, GOLPH2, TRIM36, POLD2, CGREF1, or HSD17B4, or underexpression of gene or genes CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, or SCHIP1 is used to diagnose prostate cancer or a premalignant condition thereof, or for the prognosis of the progression of the prostate cancer or of a premalignant condition thereof, or for the prognosis of the risk of recurrence of said disease.
  • 32. The method as claimed in claim 1, wherein the discriminating capacity between carcinomous and non-carcinomous samples, when the expression levels of two or more genes are determined together, is at least 1% greater than the discriminating capacity of any one of the genes when their expression levels are determined separately.
  • 33. The method according to claim 1, wherein the method is performed in vitro in a test sample.
  • 34. A method of diagnosing prostate cancer in a subject, the method comprising: determining the subject's expression level of a first gene selected from the group consisting of TACSTD1, HPN, AMACR, APOC1, GJB1, PP3111, CAMKK2, ZNF85, SND1, NONO, ICA1, PYCR1, ZNF278, BIK, HOXC6, CDK5, LASS2, NME1, PRDX4, SYNGR2, SIM2, EIF3S2, NIT2, FOXA1, CX3CL1, SNAI2, GSTP1, DST, KRT5, CSTA, LAMB3, EPHA2, GJA1, PER2, FOXO1A, TGFBR3, CLU, ROR2, ETS2, TP73L, DDR2, BNIP2, FOXF1, MYO6, ABCC4, CRYAB, CYP27A1, FGF2, IKL, PTGIS, RARRES2, PLP2, TPM2, S100A6, SCHIP1, GOLPH2, TRIM36, POLD2, CGREF1, and HSD17B4;determining the subject's expression level of a second gene, said second gene different from the first gene, but independently selected from said group;diagnosing, based upon the subject's thus determined selected gene expression levels, whether or not the subject has prostate cancer,wherein the ability to diagnose prostate cancer in the subject, when the expression levels of the selected genes are determined together, is greater than the ability to diagnose prostate cancer of the selected genes separately.
Priority Claims (1)
Number Date Country Kind
P200600348 Feb 2006 ES national
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/ES2007/000085 2/15/2007 WO 00 5/24/2010