Novel nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis of prostate cancer

- COMPUGEN LTD.

Novel markers for prostate cancer that are both sensitive and accurate. Furthermore, these markers are able to distinguish between prostate cancer and benign prostate hyperplasia (“BPH”). These markers are overexpressed in prostate cancer specifically, as opposed to normal prostate tissue and/or BPH. The measurement of these markers, alone or in combination, in patient samples provides information that the diagnostician can correlate with a probable diagnosis of prostate cancer. The markers of the present invention, alone or in combination, show a high degree of differential detection between prostate cancer and non-cancerous states.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATION(S)

This application is related to Novel Nucleotide and Amino Acid Sequences, and Assays and Methods of use thereof for Diagnosis of Prostate Cancer, and claims priority to the below U.S. provisional and non-provisional applications, each of which is incorporated herein by reference in its entirety:

Application No. 60/620,916 filed Oct. 22, 2004—Differential Expression of Markers in Colon Cancer

Application No. 60/628,123 filed Nov. 17, 2004—Differential Expression of Markers in Colon Cancer II

Application No. 60/621,131 filed Oct. 25, 2004—Diagnostic Markers for Colon Cancer, and Assays and Methods of use thereof.

Application No. 60/628,101 filed Nov. 17, 2004—Differential Expression of Markers in Breast Cancer II

Application No. 60/620,874 filed Oct. 22, 2004—Differential Expression of Markers in Ovarian Cancer

Application No. 60/628,134 filed Nov. 17, 2004—Differential Expression of Markers in Ovarian Cancer II

Application No. 60/620,853 filed Oct. 22, 2004-28814—Differential Expression of Markers in Lung Cancer

Application No. 60/628,112 filed Nov. 17, 2004—Differential Expression of Markers in Lung Cancer II

Application No. 60/628,145 filed Nov. 17, 2004—Differential Expression of Markers in Pancreatic Cancer II

Application No. 60/620,656 filed Oct. 22, 2004—Differential Expression of Markers in Prostate Cancer

Application No. 60/628,251 filed Nov. 17, 2004—Differential Expression of Markers in Prostate Cancer II

Application No. 60/628,178 filed Nov. 17, 2004—Differential Expression of Markers in Brain Cancer II

Application No. 60/628,231 filed Nov. 17, 2004—Novel Diagnostic Serum Markers, and Assays and Methods of use thereof.

Application No. 60/620,918 filed Oct. 22, 2004—Diagnostic Markers for Renal Cancer, and Assays and Methods of Use thereof.

Application No. 60/628,156 filed Nov. 17, 2004—Diagnostic Markers for Renal Cancer, and Assays and Methods of Use thereof II

Application No. 60/620,677 filed Oct. 22, 2004—Differential Expression of Markers in Bladder Cancer I

Application No. 60/628,167 filed Nov. 17, 2004—Differential Expression of Markers in Bladder Cancer II

Application No. - - - NA - - - filed Nov. 17, 2004—Novel Diagnostic Markers, and Assays and Methods of Use thereof.

Application No. 60/539,129 filed Jan. 27, 2004—Methods and Systems for Annotating Biomolecular Sequences

Application No. 60/539,128 filed Jan. 27, 2004—Evolutionary Conserved Spliced Sequences and Methods and Systems for Identifying thereof.

application Ser. No. 11/043,806 filed Jan. 27, 2005—NOVEL NUCLEOTIDE AND AMINO ACID SEQUENCES, AND ASSAYS AND METHODS OF USE THEREOF FOR DIAGNOSIS OF PROSTATE CANCER

FIELD OF THE INVENTION

The present invention is related to novel nucleotide and protein sequences that are diagnostic markers for prostate cancer, and assays and methods of use thereof.

BACKGROUND OF THE INVENTION

Prostate cancer is the most commonly diagnosed malignancy and the second most frequent cause of cancer-related deaths in the western male population. Prostate cancer therapies are most effective in the earlier stages of the disease, before metastasis has occurred. Treatment is expected to be even more effective before significant local growth of the cancerous tissue has taken place. Therefore, efforts to control the disease (i.e., to decrease prostate cancer mortality) have focused on increasing detection of the cancer while it is still locally confined and potentially curable, through diagnostic assays that are suitable for early detection of prostate cancer. Unfortunately, such detection also has significant drawbacks, because diagnostic assays that use currently available prostate cancer markers lead to high numbers of false positive diagnoses, and/or are not sufficiently sensitive (potentially leading to high numbers of false negative diagnoses).

Measurements of serum concentrations of prostatic marker enzymes have recognized value in the clinical detection, diagnosis and management of prostate cancer. The two most widely used prostatic marker enzymes are prostatic acid phosphatase (PAP) and prostate-specific antigen (PSA). Normally, both enzymes are secreted from the prostatic epithelial cells into the seminal fluid, but in patients with prostatic disease they leak into the circulation, where they can be detected by means of immunological assays (Armbruster, Clin. Che. 39:181-95 (1993)).

Prostatic acid phosphatase, one of the earliest serum markers for prostate, has an as yet undetermined function and is one of the most predominant protein components in human prostatic secretions. The use of PAP as a marker for prostatic tumors is complicated by the reported structural similarities between the prostate-specific acid phosphatase and the lysosomal acid phosphatase occurring in all tissues. Furthermore, there is a tendency towards lower PAP mRNA and protein levels in prostate cancer in comparison with benign prostatic hyperplasia (BPH). In recent years, PAP measurements were superseded by serum PSA measurements in the clinical management of prostate cancer.

Prostate-specific antigen (PSA) was identified by several groups as a prostate-specific protein from the seminal fluid, and was subsequently determined to be an antigen from prostate cancer tissue. PSA is produced exclusively by the columnar epithelial cells of the prostate and periuretural glands. Normal prostate epithelium and benign hyperplastic tissue actually produce more PSA mRNA and protein than does prostate cancer tissue. Furthermore, it was shown that loss of differentiation of prostatic carcinomas is associated with a decrease in the level of intraprostatic PSA.

Prostate-specific membrane antigen (PSM) was originally identified using an antibody developed by immunizing mice with the membrane fraction of LNCaP human prostatic adenocarcinoma cells. Like PAP and PSA, PSM can be detected in normal prostate, BPH and prostate cancer and is absent from most other tissues. However, the usefulness of PSM as marker for prostatic cancer has not been fully established.

Other markers have recently been considered. For example, PCA3 DD3 is a new marker from DiagnoCure, which has been described as being useful in a urine-based test (PCA3 itself is described in PCT Application Nos. WO 98/45420 and WO 2000/123550). This marker is apparently only expressed in prostate cancer, and therefore may be used to distinguish between BPH and prostate cancer. However, as described in greater detail below, the sensitivity and accuracy of this marker may be improved when used in combination with one or more additional markers.

Therefore, PSA is recognized as the best available marker for prostate cancer, being useful for screening selected populations of patients with symptoms indicative of prostate cancer and for monitoring patients after therapy, especially after surgical prostatectomy. However, PSA has significant drawbacks in terms of false positive measurements, since it cannot distinguish prostate cancer from BPH. It may also lead to false negative measurements, since de-differentiation of prostate cancerous tissue (which may occur with some types of prostate cancers) also leads to decreased expression of this marker. New markers are currently being developed to overcome this problem, but these markers have their own drawbacks. Clearly, new markers are required.

SUMMARY OF THE INVENTION

The background art does not teach or suggest markers for prostate cancer that are sufficiently sensitive and/or accurate, alone or in combination.

The present invention overcomes these deficiencies of the background art by providing novel markers for prostate cancer that are both sensitive and accurate. Furthermore, at least some of these markers are able to distinguish between prostate cancer and benign prostate hyperplasia (“BPH”). These markers are differentially expressed, and preferably overexpressed in prostate cancer specifically, as opposed to normal prostate tissue and/or BPH. The measurement of these markers, alone or in combination, in patient samples (biological samples) provides information that the diagnostician can correlate with a probable diagnosis of prostate cancer. The markers of the present invention, alone or in combination, show a high degree of differential detection between prostate cancer and non-cancerous states.

According to preferred embodiments of the present invention, examples of suitable biological samples include but are not limited to blood, serum, plasma, blood cells, urine, sputum, saliva, stool, spinal fluid or CSF, lymph fluid, the external secretions of the skin, respiratory, intestinal, and genitourinary tracts, tears, milk, neuronal tissue, prostate tissue or mucous and any human organ or tissue, or any sample obtained by lavage (for example of the bronchial system), and also samples of in vivo cell culture constituents. In a preferred embodiment, the biological sample comprises prostate tissue and/or other tissues of the male genitalia, or reproductive or urinary tracts, and/or a serum (and/or any blood) sample and/or a urine sample and/or a semen sample and/or any other tissue or liquid sample. The sample can optionally be diluted with a suitable eluant before contacting the sample to an antibody and/or performing any other diagnostic assay.

Information given in the text with regard to cellular localization was determined according to four different software programs: (i) tmhmm (from Center for Biological Sequence Analysis, Technical University of Denmark DTU, www “dot” cbs “dot” dtu “dot” dk/services/TMHMM/TMHMM2.0b.guide.php) or (ii) tmpred (from EMBnet, maintained by the ISREC Bionformatics group and the LICR Information Technology Office, Ludwig Institute for Cancer Research, Swiss Institute of Bioinformatics, www “dot” ch “dot” embnet “dot” org/software/TMPRED_form “dot” html) for transmembrane region prediction; (iii) signalp_hmm or (iv) signalp_nn (both from Center for Biological Sequence Analysis, Technical University of Denmark DTU, www “dot” cbs “dot” dtu “dot” dk/services/SignalP/background/prediction.php) for signal peptide prediction. The terms “signalp_hrm” and “signalp_nn” refer to two modes of operation for the program SignalP: hmm refers to Hidden Markov Model, while nn refers to neural networks. Localization was also determined through manual inspection of known protein localization and/or gene structure, and the use of heuristics by the individual inventor. In some cases for the manual inspection of cellular localization prediction inventors used the ProLoc computational platform [Einat Hazkani-Covo, Erez Levanon, Galit Rotman, Dan Graur and Amit Novik; (2004) “Evolution of multicellularity in metazoa: comparative analysis of the subcellular localization of proteins in Saccharomyces, Drosophila and Caenorhabditis.” Cell Biology International 2004; 28(3):171-8.], which predicts protein localization based on various parameters including, protein domains (e.g., prediction of trans-membranous regions and localization thereof within the protein), pI, protein length, amino acid composition, homology to pre-annotated proteins, recognition of sequence patterns which direct the protein to a certain organelle (such as, nuclear localization signal, NLS, mitochondria localization signal), signal peptide and anchor modeling and using unique domains from Pfam that are specific to a single compartment.

Information is given in the text with regard to SNPs (single nucleotide polymorphisms). A description of the abbreviations is as follows. “T->C”, for example, means that the SNP results in a change at the position given in the table from T to C. Similarly, “M->Q”, for example, means that the SNP has caused a change in the corresponding amino acid sequence, from methionine (M) to glutamine (Q). If, in place of a letter at the right hand side for the nucleotide sequence SNP, there is a space, it indicates that a frameshift has occurred. A frameshift may also be indicated with a hyphen (-). A stop codon is indicated with an asterisk at the right hand side (*). As part of the description of an SNP, a comment may be found in parentheses after the above description of the SNP itself. This comment may include an FTId, which is an identifier to a SwissProt entry that was created with the indicated SNP. An FTId is a unique and stable feature identifier, which allows construction of links directly from position-specific annotation in the feature table to specialized protein-related databases. The FTId is always the last component of a feature in the description field, as follows: FTId=XXX_number, in which XXX is the 3-letter code for the specific feature key, separated by an underscore from a 6-digit number. In the table of the amino acid mutations of the wild type proteins of the selected splice variants of the invention, the header of the first column is “SNP position(s) on amino acid sequence”, representing a position of a known mutation on amino acid sequence.

SNPs may optionally be used as diagnostic markers according to the present invention, alone or in combination with one or more other SNPs and/or any other diagnostic marker. Preferred embodiments of the present invention comprise such SNPs, including but not limited to novel SNPs on the known (WT or wild type) protein sequences given below, as well as novel nucleic acid and/or amino acid sequences formed through such SNPs, and/or any SNP on a variant amino acid and/or nucleic acid sequence described herein.

Information given in the text with regard to the Homology to the known proteins was determined by Smith-Waterman version 5.1.2 using special (non default) parameters as follows:

model=sw.model

GAPEXT=0

GAPOP=100.0

    • MATRIX=blosum 100

Information is given with regard to overexpression of a cluster in cancer based on ESTs. A key to the p values with regard to the analysis of such overexpression is as follows:

    • library-based statistics: P-value without including the level of expression in cell-lines (P1)
    • library based statistics: P-value including the level of expression in cell-lines (P2)
    • EST clone statistics: P-value without including the level of expression in cell-lines (SP1)
    • EST clone statistics: predicted overexpression ratio without including the level of expression in cell-lines (R3)
    • EST clone statistics: P-value including the level of expression in cell-lines (SP2)
    • EST clone statistics: predicted overexpression ratio including the level of expression in cell-lines (R4)

Library-based statistics refer to statistics over an entire library, while EST clone statistics refer to expression only for ESTs from a particular tissue or cancer.

Information is given with regard to overexpression of a cluster in cancer based on microarrays. As a microarray reference, in the specific segment paragraphs, the unabbreviated tissue name was used as the reference to the type of chip for which expression was measured.

There are two types of microarray results: those from microarrays prepared according to a design by the present inventors, for which the microarray fabrication procedure is described in detail in Materials and Experimental Procedures section herein; and those results from microarrays using Affymetrix technology. As a microarray reference, in the specific segment paragraphs, the unabbreviated tissue name was used as the reference to the type of chip for which expression was measured. For microarrays prepared according to a design by the present inventors, the probe name begins with the name of the cluster (gene), followed by an identifying number. Oligonucleotide microarray results taken from Affymetrix data were from chips available from Affymetrix Inc, Santa Clara, Calif., USA (see for example data regarding the Human Genome U133 (HG-U133) Set at www “dot” affymetrix “dot” com/products/arrays/specific/hgu133.affx; GeneChip Human Genome U133A 2.0 Array at www “dot” affymetrix “dot” com/products/arrays/specific/hgu133av2.affx; and Human Genome U133 Plus 2.0 Array at www “dot” affymetrix “dot” com/products/arrays/specific/hgu133plus.affx). The probe names follow the Affymetrix naming convention. The data is available from NCBI Gene Expression Omnibus (see www.ncbi.nlm.nih.gov/projects/geo/ and Edgar et al, Nucleic Acids Research, 2002, Vol. 30, No. 1 207-210). The dataset (including results) is available from www “dot” ncbi “dot” nlm “dot” nih “dot” gov/geo/query/acc.cgi?acc=GSE1133 for the Series GSE1133 database (published on March 2004); a reference to these results is as follows: Su et al (Proc Natl Acad Sci USA. 2004 Apr. 20; 101(16):6062-7. Epub 2004 Apr. 9). A list of probes designed according to the present inventors is given below.

>HSSTROL3_0_0_12518 ATGAGAGTAACCTCACCCGTGCACTAGTTTACAGAGCATTCACTGCCCCA >HSSTROL3_0_0_12517 CAGAGATGAGAGCCTGGAGCATTGCAGATGCCAGGGACTTCACAAATGAA

The following list of abbreviations for tissues was used in the TAA histograms. The term “TAA” stands for “Tumor Associated Antigen”, and the TAA histograms, given in the text, represent the cancerous tissue expression pattern as predicted by the biomarkers selection engine, as described in detail in examples 1-5 below:

“BONE” for “bone”;

    • “COL” for “colon”;
    • “EPI” for “epithelial”;
    • “GEN” for “general”;
    • “LIVER” for “liver”;
    • “LUN” for “lung”;
    • “LYMPH” for “lymph nodes”;
    • “MARROW” for “bone marrow”;
    • “OVA” for “ovary”;
    • “PANCREAS” for “pancreas”;
    • “PRO” for “prostate”;
    • “STOMACH” for “stomach”;
    • “TCELL” for “T cells”;
    • “THYROID” for “Thyroid”;
    • “MAM” for “breast”;
    • “BRAIN” for “brain”;
    • “UTERUS” for “uterus”;
    • “SKIN” for “skin”;
    • “KIDNEY” for “kidney”;
    • “MUSCLE” for “muscle”;
    • “ADREN” for “adrenal”;
    • “HEAD” for “head and neck”;
    • “BLADDER” for “bladder”;

It should be noted that the terms “segment”, “seg” and “node” are used interchangeably in reference to nucleic acid sequences of the present invention; they refer to portions of nucleic acid sequences that were shown to have one or more properties as described below. They are also the building blocks that were used to construct complete nucleic acid sequences as described in greater detail below. Optionally and preferably, they are examples of oligonucleotides which are embodiments of the present invention, for example as amplicons, hybridization units and/or from which primers and/or complementary oligonucleotides may optionally be derived, and/or for any other use.

As used herein the phrase “prostate cancer” refers to cancers of the prostate tissue and/or other tissues of the male genitalia, or reproductive or urinary tracts.

The term “marker” in the context of the present invention refers to a nucleic acid fragment, a peptide, or a polypeptide, which is differentially present in a sample taken from subjects (patients) having prostate cancer as compared to a comparable sample taken from subjects who do not have prostate cancer.

The phrase “differentially present” refers to differences in the quantity of a marker present in a sample taken from patients having prostate cancer as compared to a comparable sample taken from patients who do not have prostate cancer. For example, a nucleic acid fragment may optionally be differentially present between the two samples if the amount of the nucleic acid fragment in one sample is significantly different from the amount of the nucleic acid fragment in the other sample, for example as measured by hybridization and/or NAT-based assays. A polypeptide is differentially present between the two samples if the amount of the polypeptide in one sample is significantly different from the amount of the polypeptide in the other sample. It should be noted that if the marker is detectable in one sample and not detectable in the other, then such a marker can be considered to be differentially present.

As used herein the phrase “diagnostic” means identifying the presence or nature of a pathologic condition. Diagnostic methods differ in their sensitivity and specificity. The “sensitivity” of a diagnostic assay is the percentage of diseased individuals who test positive (percent of “true positives”). Diseased individuals not detected by the assay are “false negatives.” Subjects who are not diseased and who test negative in the assay are termed “true negatives.” The “specificity” of a diagnostic assay is 1 minus the false positive rate, where the “false positive” rate is defined as the proportion of those without the disease who test positive. While a particular diagnostic method may not provide a definitive diagnosis of a condition, it suffices if the method provides a positive indication that aids in diagnosis.

As used herein the phrase “diagnosing” refers to classifying a disease or a symptom, determining a severity of the disease, monitoring disease progression, forecasting an outcome of a disease and/or prospects of recovery. The term “detecting” may also optionally encompass any of the above.

Diagnosis of a disease according to the present invention can be affected by determining a level of a polynucleotide or a polypeptide of the present invention in a biological sample obtained from the subject, wherein the level determined can be correlated with predisposition to, or presence or absence of the disease. It should be noted that a “biological sample obtained from the subject” may also optionally comprise a sample that has not been physically removed from the subject, as described in greater detail below.

As used herein, the term “level” refers to expression levels of RNA and/or protein or to DNA copy number of a marker of the present invention.

Typically the level of the marker in a biological sample obtained from the subject is different (i.e., increased or decreased) from the level of the same variant in a similar sample obtained from a healthy individual (examples of biological samples are described herein).

Numerous well known tissue or fluid collection methods can be utilized to collect the biological sample from the subject in order to determine the level of DNA, RNA and/or polypeptide of the variant of interest in the subject.

Examples include, but are not limited to, fine needle biopsy, needle biopsy, core needle biopsy and surgical biopsy (e.g., brain biopsy), and lavage. Regardless of the procedure employed, once a biopsy/sample is obtained the level of the variant can be determined and a diagnosis can thus be made.

Determining the level of the same variant in normal tissues of the same origin is preferably effected along-side to detect an elevated expression and/or amplification and/or a decreased expression, of the variant as opposed to the normal tissues.

A “test amount” of a marker refers to an amount of a marker in a subject's sample that is consistent with a diagnosis of prostate cancer. A test amount can be either in absolute amount (e.g., microgram/ml) or a relative amount (e.g., relative intensity of signals).

A “control amount” of a marker can be any amount or a range of amounts to be compared against a test amount of a marker. For example, a control amount of a marker can be the amount of a marker in a patient with prostate cancer or a person without prostate cancer. A control amount can be either in absolute amount (e.g., microgram/ml) or a relative amount (e.g., relative intensity of signals).

“Detect” refers to identifying the presence, absence or amount of the object to be detected.

A “label” includes any moiety or item detectable by spectroscopic, photo chemical, biochemical, immunochemical, or chemical means. For example, useful labels include 32P, 35S, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin-streptavadin, dioxigenin, haptens and proteins for which antisera or monoclonal antibodies are available, or nucleic acid molecules with a sequence complementary to a target. The label often generates a measurable signal, such as a radioactive, chromogenic, or fluorescent signal, that can be used to quantify the amount of bound label in a sample. The label can be incorporated in or attached to a primer or probe either covalently, or through ionic, van der Waals or hydrogen bonds, e.g., incorporation of radioactive nucleotides, or biotinylated nucleotides that are recognized by streptavadin. The label may be directly or indirectly detectable. Indirect detection can involve the binding of a second label to the first label, directly or indirectly. For example, the label can be the ligand of a binding partner, such as biotin, which is a binding partner for streptavadin, or a nucleotide sequence, which is the binding partner for a complementary sequence, to which it can specifically hybridize. The binding partner may itself be directly detectable, for example, an antibody may be itself labeled with a fluorescent molecule. The binding partner also may be indirectly detectable, for example, a nucleic acid having a complementary nucleotide sequence can be a part of a branched DNA molecule that is in turn detectable through hybridization with other labeled nucleic acid molecules (see, e.g., P. D. Fahrlander and A. Klausner, Bio/Technology 6:1165 (1988)). Quantitation of the signal is achieved by, e.g., scintillation counting, densitometry, or flow cytometry.

Exemplary detectable labels, optionally and preferably for use with immunoassays, include but are not limited to magnetic beads, fluorescent dyes, radiolabels, enzymes (e.g., horse radish peroxide, alkaline phosphatase and others commonly used in an ELISA), and calorimetric labels such as colloidal gold or colored glass or plastic beads. Alternatively, the marker in the sample can be detected using an indirect assay, wherein, for example, a second, labeled antibody is used to detect bound marker-specific antibody, and/or in a competition or inhibition assay wherein, for example, a monoclonal antibody which binds to a distinct epitope of the marker are incubated simultaneously with the mixture.

“Immunoassay” is an assay that uses an antibody to specifically bind an antigen. The immunoassay is characterized by the use of specific binding properties of a particular antibody to isolate, target, and/or quantify the antigen.

The phrase “specifically (or selectively) binds” to an antibody or “specifically (or selectively) immunoreactive with,” when referring to a protein or peptide (or other epitope), refers to a binding reaction that is determinative of the presence of the protein in a heterogeneous population of proteins and other biologics. Thus, under designated immunoassay conditions, the specified antibodies bind to a particular protein at least two times greater than the background (non-specific signal) and do not substantially bind in a significant amount to other proteins present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein. For example, polyclonal antibodies raised to seminal basic protein from specific species such as rat, mouse, or human can be selected to obtain only those polyclonal antibodies that are specifically immunoreactive with seminal basic protein and not with other proteins, except for polymorphic variants and alleles of seminal basic protein. This selection may be achieved by subtracting out antibodies that cross-react with seminal basic protein molecules from other species. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select antibodies specifically immunoreactive with a protein (see, e.g., Harlow & Lane, Antibodies, A Laboratory Manual (1988), for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity). Typically a specific or selective reaction will be at least twice background signal or noise and more typically more than 10 to 100 times background.

According to preferred embodiments of the present invention, there is provided an isolated polynucleotide comprising the sequence selected from the group consisting of SEQ ID NOs: 5, 6, 7, 8, 9 and 10.

According to preferred embodiments of the present invention, there is provided an isolated polynucleotide comprising the segment selected from the group consisting of SEQ ID NOs: 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114 and 115.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide comprising SEQ ID NOs: 331, 332, 333, 334 and 335.

According to preferred embodiments of the present invention, there is provided an isolated polynucleotide comprising the sequence selected from the group consisting of SEQ ID NOs: 34, 35, 36, 37, 38 and 39.

According to preferred embodiments of the present invention, there is provided an isolated polynucleotide comprising a segment SEQ ID NOs: 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236 and 237.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide comprising SEQ ID NOs: 359, 360, 361, 362 and 363.

According to preferred embodiments of the present invention, there is provided an isolated polynucleotide comprising the sequence selected from the group consisting of SEQ ID NOs: 13 and 14.

According to preferred embodiments of the present invention, there is provided an isolated polynucleotide comprising the node selected from the group consisting of SEQ ID NOs: 136, 137, 138, 139, 140, 141 and 142.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide comprising SEQ ID NOs: 338 and 339.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 359, comprising a first amino acid sequence being at least 90% homologous to corresponding to amino acids 1-163 of SEQ ID NOs. 391, which also corresponds to amino acids 1-163 of SEQ ID NOs. 359, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NOs. 359, a second amino acid sequence being at least 90% homologous to corresponding to amino acids 165-445 of SEQ ID NOs. 391, which also corresponds to amino acids 165-445 of SEQ ID NO. 359, and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence corresponding to amino acids 446-496 of SEQ ID NO. 359, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 359, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 446-496 in SEQ ID NO. 359.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NOs.360, comprising a first amino acid sequence being at least 90% homologous to corresponding to amino acids 1-163 of SEQ ID NOs. 391, which also corresponds to amino acids 1-163 of SEQ ID NO:360, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO. 360, a second amino acid sequence being at least 90% homologous to corresponding to amino acids 165-358 of SEQ ID NOs. 391, which also corresponds to amino acids 165-358 of SEQ ID NO. 360, and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence corresponding to amino acids 359-382 of SEQ ID NO. 360, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 360, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 359-382 in SEQ ID NO. 360.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 361, comprising a first amino acid sequence being at least 90% homologous to corresponding to amino acids 1-163 of SEQ ID NOs. 391, which also corresponds to amino acids 1-163 of SEQ ID NO. 361, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO. 361, a second amino acid sequence being at least 90% homologous to corresponding to amino acids 165-359 of SEQ ID NOs. 391, which also corresponds to amino acids 165-359 of SEQ ID NO. 361, and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence corresponding to amino acids 360-370 of SEQ ID NO. 361, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 361, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 360-370 in SEQ ID NO. 361.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 362, comprising a first amino acid sequence being at least 90% homologous to corresponding to amino acids 1-163 of SEQ ID NOs. SEQ ID NOs. 391, which also corresponds to amino acids 1-163 of SEQ ID NO:362, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO:362, a second amino acid sequence being at least 90% homologous to corresponding to amino acids 165-286 of SEQ ID NOs. 391, which also corresponds to amino acids 165-286 of SEQ ID NO:362, and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence corresponding to amino acids 287-301 of SEQ ID NO:362, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO:362, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 287-301 in SEQ ID NO. 362.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 363, comprising a first amino acid sequence being at least 90% homologous to corresponding to amino acids 1-96 of SEQ ID NOs. 391, which also corresponds to amino acids 1-96 of SEQ ID NO. 363, a second amino acid sequence being at least 90% homologous to corresponding to amino acids 113-163 of SEQ ID NOs. 391, which also corresponds to amino acids 97-147 of SEQ ID NO:363, a bridging amino acid H corresponding to amino acid 148 of SEQ ID NO. 363, a third amino acid sequence being at least 90% homologous to corresponding to amino acids 165-359 of SEQ ID NOs. 391, which also corresponds to amino acids 149-343 of SEQ ID NO:363, and a fourth amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 344-354 of SEQ ID NO. 363, wherein said first amino acid sequence, second amino acid sequence, bridging amino acid, third amino acid sequence and fourth amino acid sequence are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of an edge portion of SEQ ID NO:363, comprising a polypeptide having a length “n”, wherein n is at least about 10 amino acids in length, optionally at least about 20 amino acids in length, preferably at least about 30 amino acids in length, more preferably at least about 40 amino acids in length and most preferably at least about 50 amino acids in length, wherein at least two amino acids comprise KR, having a structure as follows: a sequence starting from any of amino acid numbers 96-x to 96; and ending at any of amino acid numbers 97+((n-2)−x), in which x varies from 0 to n-2.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 363, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence in SEQ ID NO. 363.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 339, comprising a first amino acid sequence being at least 90% homologous to corresponding to amino acids 1-27 of SEQ ID NO. 387, which also corresponds to amino acids 1-27 of SEQ ID NO. 339, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence corresponding to amino acids 28-41 of SEQ ID NO. 339, wherein said first amino acid sequence and second amino acid sequence are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 339, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 28-41 in SEQ ID NO. 339.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 332, comprising a first amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 1-110 of SEQ ID NO. 332, and a second amino acid sequence being at least 90% homologous to TQ corresponding to amino acids 1-112 of Q8IXM0, which also corresponds to amino acids 111-222 of SEQ ID NO. 332, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a head of SEQ ID NO. 332, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 1-110 of SEQ ID NO. 332.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 332, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-83 of Q96AC2, which also corresponds to amino acids 1-83 of SEQ ID NO. 332, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 84-222 of SEQ ID NO. 332, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 332, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 84-222 in SEQ ID NO. 332.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide of SEQ ID NO. 332, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-83 of Q8N2G4, which also corresponds to amino acids 1-83 of SEQ ID NO. 332, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 84-222 of SEQ ID NO. 332, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 332, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 84-222 in SEQ ID NO. 332.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 332, comprising a first amino acid sequence being at least 90% homologous to amino acids 24-106 of BAC85518, which also corresponds to amino acids 1-83 of SEQ ID NO. 332, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 84-222 of SEQ ID NO. 332, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 332, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 84-222 in SEQ ID NO. 332.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 333, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-64 of Q96AC2, which also corresponds to amino acids 1-64 of SEQ ID NO. 333, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 65-93 of SEQ ID NO. 333, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 333, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 65-93 in SEQ ID NO. 333.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 333, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-64 of Q8N2G4, which also corresponds to amino acids 1-64 of SEQ ID NO. 333, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 65-93 of SEQ ID NO. 333, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 333, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 65-93 in SEQ ID NO. 333.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 333, comprising a first amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 1-5 of SEQ ID NO. 333, second amino acid sequence being at least 90% homologous to amino acids 22-80 of BAC85273, which also corresponds to amino acids 6-64 of SEQ ID NO. 333, and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 65-93 of SEQ ID NO. 333, wherein said first, second and third amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a head of SEQ ID NO. 333, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 1-5 of SEQ ID NO. 333.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 333, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 65-93 in SEQ ID NO. 333.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 333, comprising a first amino acid sequence being at least 90% homologous to amino acids 24-87 of BAC85518, which also corresponds to amino acids 1-64 of SEQ ID NO. 333, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 65-93 of SEQ ID NO. 333, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 333, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 65-93 in SEQ ID NO. 333.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 334, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-63 of Q96AC2, which also corresponds to amino acids 1-63 of SEQ ID NO. 334, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 64-84 of SEQ ID NO. 334, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 334, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 64-84 in SEQ ID NO. 334.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 335, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-63 of SEQ ID NOs. Q96AC2, which also corresponds to amino acids 1-63 of SEQ ID NO. 335, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 64-90 of SEQ ID NO. 335, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 335, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 64-90 in SEQ ID NO. 335.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 335, comprising a first amino acid sequence being at least 90% homologous to amino acids 1-63 of Q8N2G4, which also corresponds to amino acids 1-63 of SEQ ID NO. 335, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 64-90 of SEQ ID NO. 335, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 335, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 64-90 in SEQ ID NO. 335.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 335, comprising a first amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 1-5 of SEQ ID NO. 335, second amino acid sequence being at least 90% homologous to amino acids 22-79 of BAC85273, which also corresponds to amino acids 6-63 of SEQ ID NO. 335, and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 64-90 of SEQ ID NO. 335, wherein said first, second and third amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a head of SEQ ID NO. 335, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 1-5 of SEQ ID NO. 335.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 335, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the amino acids 64-90 in SEQ ID NO. 335.

According to preferred embodiments of the present invention, there is provided an isolated chimeric polypeptide encoding for SEQ ID NO. 335, comprising a first amino acid sequence being at least 90% homologous to amino acids 24-86 of BAC85518, which also corresponds to amino acids 1-63 of SEQ ID NO. 335, and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide sequence corresponding to amino acids 64-90 of SEQ ID NO. 335, wherein said first and second amino acid sequences are contiguous and in a sequential order.

According to preferred embodiments of the present invention, there is provided an isolated polypeptide encoding for a tail of SEQ ID NO. 335, comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to amino acids 64-90 in SEQ ID NO. 335.

According to preferred embodiments of the present invention, there is provided an antibody from cluster R11723, HUTMTREFAC, HSSTROL3, capable of specifically binding to an epitope of an amino acid sequence.

Optionally the amino acid sequence corresponds to a bridge, edge portion, tail, head or insertion.

Optionally the antibody is capable of differentiating between a splice variant having said epitope and a corresponding known protein.

According to preferred embodiments of the present invention, there is provided a kit for detecting prostate cancer, comprising a kit from cluster R11723, HUMTREFAC, HSSTROL3 for detecting overexpression of a splice variant.

Optionally the kit comprises a NAT-based technology.

Optionally the kit further comprises at least one primer pair capable of selectively hybridizing to a nucleic acid sequence.

Optionally the kit further comprises at least one oligonucleotide capable of selectively hybridizing to a nucleic acid sequence.

Optionally the kit comprises an antibody.

Optionally the kit further comprises at least one reagent for performing an ELISA or a Western blot.

According to preferred embodiments of the present invention, there is provided a method for detecting prostate cancer, comprising detecting overexpression of a splice variant from cluster R11723, HUMTREFAC, and HSSTROL3.

Optionally detecting overexpression is performed with a NAT-based technology.

Optionally detecting overexpression is performed with an immunoassay.

Optionally the immunoassay comprises an antibody.

According to preferred embodiments of the present invention, there is provided a biomarker capable of detecting prostate cancer, comprising nucleic acid sequences or a fragment thereof, or amino acid sequences or a fragment thereof from cluster R11723, HUMTREFAC and HSSTROL3.

According to preferred embodiments of the present invention, there is provided a method for screening for prostate cancer, comprising detecting prostate cancer cells with a biomarker or an antibody or a method or assay from cluster R11723, HUMTREFAC and HSSTROL3.

According to preferred embodiments of the present invention, there is provided a method for diagnosing prostate cancer, comprising detecting prostate cancer cells with a biomarker or an antibody or a method or assay from cluster R11723, HUMTREFAC and HSSTROL3.

According to preferred embodiments of the present invention, there is provided a method for monitoring disease progression, treatment efficacy, relapse of prostate cancer, comprising detecting prostate cancer cells with a biomarker or an antibody or a method or assay from cluster R11723, HUMTREFAC and HSSTROL3.

According to preferred embodiments of the present invention, there is provided a method of selecting a therapy for prostate cancer, comprising detecting prostate cancer cells with a biomarker or an antibody or a method or assay from cluster R11723, HUMTREFAC and HSSTROL3.

According to preferred embodiments of the present invention, preferably any of the above nucleic acid and/or amino acid sequences further comprises any sequence having at least about 70%, preferably at least about 80%, more preferably at least about 90%, most preferably at least about 95% homology thereto.

All nucleic acid sequences and/or amino acid sequences shown herein as embodiments of the present invention relate to their isolated form, as isolated polynucleotides (including for all transcripts), oligonucleotides (including for all segments, amplicons and primers), peptides (including for all tails, bridges, insertions or heads, optionally including other antibody epitopes as described herein) and/or polypeptides (including for all proteins). It should be noted that oligonucleotide and polynucleotide, or peptide and polypeptide, may optionally be used interchangeably.

Unless otherwise noted, all experimental data relates to variants of the present invention, named according to the segment being tested (as expression was tested through RT-PCR as described).

Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). All of these are hereby incorporated by reference as if fully set forth herein. As used herein, the following terms have the meanings ascribed to them unless specified otherwise.

BRIEF DESCRIPTION OF DRAWINGS

FIG. 1 is a schematic description of the cancer biomarker selection engine.

FIG. 2 is a schematic illustration, depicting grouping of transcripts of a given cluster based on presence or absence of unique sequence regions.

FIG. 3 is a schematic summary of quantitative real-time PCR analysis.

FIG. 4 is a schematic presentation of the oligonucleotide based microarray fabrication.

FIG. 5 is a schematic summary of the oligonucleotide based microarray experimental flow.

FIG. 6 is a histogram showing Cancer and cell-line vs. normal tissue expression for Cluster R11723, demonstrating overexpression in epithelial malignant tumors, a mixture of malignant tumors from different tissues and kidney malignant tumors.

FIG. 7 is a histogram showing over expression of the R11723 transcripts which are detectable by amplicon as depicted in sequence name R11723 seg13 (SEQ ID NO:492) in cancerous prostate samples relative to the normal samples.

FIG. 8 is a histogram showing expression of R11723 transcripts, which are detectable by amplicon as depicted in sequence name R11723seg13 (SEQ ID NO:492), in different normal tissues.

FIG. 9A are histograms showing over expression of the R11723 transcripts, which are detectable by amplicon as depicted in sequence name R11723 junc11-18 (SEQ ID NO:495) in cancerous prostate samples relative to the normal samples (FIG. 9A) or expression in normal tissues (FIG. 9B).

FIG. 10 is a histogram showing Cancer and cell-line vs. normal tissue expression for Cluster HUMTREFAC, demonstrating overexpression in a mixture of malignant tumors from different tissues, breast malignant tumors, pancreas carcinoma and prostate cancer.

FIG. 11 is a histogram showing Cancer and cell-line vs. normal tissue expression for Cluster HSSTROL3, demonstrating overexpression in transitional cell carcinoma, epithelial malignant tumors, a mixture of malignant tumors from different tissues and pancreas carcinoma.

FIG. 12 is a histogram showing the over expression of the Stromelysin-3 precursor (SEQ ID NO:391) transcripts, which are detectable by amplicon as depicted in sequence name HSSTROL3 seg24 (SEQ ID NO:499), in cancerous Prostate samples relative to the normal samples.

FIG. 13 is a histogram demonstrating the expression of Stromelysin-3 transcripts which are detectable by amplicon as depicted in sequence name HSSTROL3 seg24 (SEQ ID NO:499) in different normal tissues.

FIGS. 14 and 15 are histograms showing over expression of the Thrombospondin 1 (THBS1) transcripts in cancerous and benign (BPH) prostate samples relative to the normal samples.

FIG. 16 is a histogram showing the relative expression of Thrombospondin 1 (THBS1) variants in normal, benign and tumor derived prostate samples as determined by oligonucleotide-based micro-array experiments with SEQ ID NOs: 477, 478, 479, 480, 481, 482.

DESCRIPTION OF PREFERRED EMBODIMENTS

The present invention is of novel markers for prostate cancer that are both sensitive and accurate. Biomolecular sequences (amino acid and/or nucleic acid sequences) uncovered using the methodology of the present invention and described herein can be efficiently utilized as tissue or pathological markers and/or as drugs or drug targets for treating or preventing a disease.

These markers are specifically released to the bloodstream under conditions of prostate cancer and/or other prostate pathology, and/or are otherwise expressed at a much higher level and/or specifically expressed in prostate cancer tissue or cells. The measurement of these markers, alone or in combination, in patient samples provides information that the diagnostician can correlate with a probable diagnosis of prostate cancer and/or pathology.

The present invention therefore also relates to diagnostic assays for prostate cancer and/or prostate pathology, and methods of use of such markers for detection of prostate cancer and/or prostate pathology, optionally and preferably in a sample taken from a subject (patient), which is more preferably some type of blood sample.

The markers of the present invention, alone or in combination, can be used for prognosis, prediction, screening, early diagnosis, staging, therapy selection and treatment monitoring of prostate cancer. For example, optionally and preferably, these markers may be used for staging prostate cancer and/or monitoring the progression of the disease. Furthermore, the markers of the present invention, alone or in combination, can be used for detection of the source of metastasis found in anatomical places other then prostate. Also, one or more of the markers may optionally be used in combination with one or more other prostate cancer markers (other than those described herein).

Biomolecular sequences (amino acid and/or nucleic acid sequences) uncovered using the methodology of the present invention and described herein can be efficiently utilized as tissue or pathological markers and/or as drugs or drug targets for treating or preventing a disease.

These markers are specifically released to the bloodstream under conditions of prostate cancer (or one of the above indicative conditions), and/or are otherwise expressed at a much higher level and/or specifically expressed in prostate cancer tissue or cells, and/or tissue or cells under one of the above indicative conditions. The measurement of these markers, alone or in combination, in patient samples provides information that the diagnostician can correlate with a probable diagnosis of prostate cancer and/or a condition that it is indicative of a higher risk for prostate cancer.

The present invention therefore also relates to diagnostic assays for prostate cancer and/or an indicative condition, and methods of use of such markers for detection of prostate cancer and/or an indicative condition, optionally and preferably in a sample taken from a subject (patient), which is more preferably some type of blood sample.

According to a preferred embodiment of the present invention, use of the marker optionally and preferably permits a non-cancerous prostate disease state to be distinguished from prostate cancer and/or an indicative condition. A non limiting example of a non-cancerous prostate disease state includes BPH. According to another preferred embodiment of the present invention, use of the marker optionally and preferably permits an indicative condition to be distinguished from prostate cancer.

In another embodiment, the present invention relates to bridges, tails, heads and/or insertions, and/or analogs, homologs and derivatives of such peptides. Such bridges, tails, heads and/or insertions are described in greater detail below with regard to the Examples.

As used herein a “tail” refers to a peptide sequence at the end of an amino acid sequence that is unique to a splice variant according to the present invention. Therefore, a splice variant having such a tail may optionally be considered as a chimera, in that at least a first portion of the splice variant is typically highly homologous (often 100% identical) to a portion of the corresponding known protein, while at least a second portion of the variant comprises the tail.

As used herein a “head” refers to a peptide sequence at the beginning of an amino acid sequence that is unique to a splice variant according to the present invention. Therefore, a splice variant having such a head may optionally be considered as a chimera, in that at least a first portion of the splice variant comprises the head, while at least a second portion is typically highly homologous (often 100% identical) to a portion of the corresponding known protein.

As used herein “an edge portion” refers to a connection between two portions of a splice variant according to the present invention that were not joined in the wild type or known protein. An edge may optionally arise due to a join between the above “known protein” portion of a variant and the tail, for example, and/or may occur if an internal portion of the wild type sequence is no longer present, such that two portions of the sequence are now contiguous in the splice variant that were not contiguous in the known protein. A “bridge” may optionally be an edge portion as described above, but may also include a join between a head and a “known protein” portion of a variant, or a join between a tail and a “known protein” portion of a variant, or a join between an insertion and a “known protein” portion of a variant.

Optionally and preferably, a bridge between a tail or a head or a unique insertion, and a “known protein” portion of a variant, comprises at least about 10 amino acids, more preferably at least about 20 amino acids, most preferably at least about 30 amino acids, and even more preferably at least about 40 amino acids, in which at least one amino acid is from the tail/head/insertion and at least one amino acid is from the “known protein” portion of a variant. Also optionally, the bridge may comprise any number of amino acids from about 10 to about 40 amino acids (for example, 10, 11, 12, 13 . . . 37, 38, 39, 40 amino acids in length, or any number in between).

It should be noted that a bridge cannot be extended beyond the length of the sequence in either direction, and it should be assumed that every bridge description is to be read in such manner that the bridge length does not extend beyond the sequence itself.

Furthermore, bridges are described with regard to a sliding window in certain contexts below. For example, certain descriptions of the bridges feature the following format: a bridge between two edges (in which a portion of the known protein is not present in the variant) may optionally be described as follows: a bridge portion of CONTIG-NAME_P1 (representing the name of the protein), comprising a polypeptide having a length “n”, wherein n is at least about 10 amino acids in length, optionally at least about 20 amino acids in length, preferably at least about 30 amino acids in length, more preferably at least about 40 amino acids in length and most preferably at least about 50 amino acids in length, wherein at least two amino acids comprise XX (2 amino acids in the center of the bridge, one from each end of the edge), having a structure as follows (numbering according to the sequence of CONTIG-NAME_P1): a sequence starting from any of amino acid numbers 49-x to 49 (for example); and ending at any of amino acid numbers 50+((n-2)−x) (for example), in which x varies from 0 to n-2. In this example, it should also be read as including bridges in which n is any number of amino acids between 10-50 amino acids in length. Furthermore, the bridge polypeptide cannot extend beyond the sequence, so it should be read such that 49-x (for example) is not less than 1, nor 50+((n-2)−x) (for example) greater than the total sequence length.

In another embodiment, this invention provides antibodies specifically recognizing the splice variants and polypeptide fragments thereof of this invention. Preferably such antibodies differentially recognize splice variants of the present invention but do not recognize a corresponding known protein (such known proteins are discussed with regard to their splice variants in the Examples below).

In another embodiment, this invention provides an isolated nucleic acid molecule encoding for a splice variant according to the present invention, having a nucleotide sequence as set forth in any one of the sequences listed herein, or a sequence complementary thereto. In another embodiment, this invention provides an isolated nucleic acid molecule, having a nucleotide sequence as set forth in any one of the sequences listed herein, or a sequence complementary thereto. In another embodiment, this invention provides an oligonucleotide of at least about 12 nucleotides, specifically hybridizable with the nucleic acid molecules of this invention. In another embodiment, this invention provides vectors, cells, liposomes and compositions comprising the isolated nucleic acids of this invention.

In another embodiment, this invention provides a method for detecting a splice variant according to the present invention in a biological sample, comprising: contacting a biological sample with an antibody specifically recognizing a splice variant according to the present invention under conditions whereby the antibody specifically interacts with the splice variant in the biological sample but do not recognize known corresponding proteins (wherein the known protein is discussed with regard to its splice variant(s) in the Examples below), and detecting said interaction; wherein the presence of an interaction correlates with the presence of a splice variant in the biological sample.

In another embodiment, this invention provides a method for detecting a splice variant nucleic acid sequences in a biological sample, comprising: hybridizing the isolated nucleic acid molecules or oligonucleotide fragments of at least about a minimum length to a nucleic acid material of a biological sample and detecting a hybridization complex; wherein the presence of a hybridization complex correlates with the presence of a splice variant nucleic acid sequence in the biological sample.

According to the present invention, the splice variants described herein are non-limiting examples of markers for diagnosing prostate cancer and/or prostate pathology. Each splice variant marker of the present invention can be used alone or in combination, for various uses, including but not limited to, prognosis, prediction, screening, early diagnosis, determination of progression, therapy selection and treatment monitoring of prostate cancer and/or prostate pathology.

According to optional but preferred embodiments of the present invention, any marker according to the present invention may optionally be used alone or combination. Such a combination may optionally comprise a plurality of markers described herein, optionally including any subcombination of markers, and/or a combination featuring at least one other marker, for example a known marker. Furthermore, such a combination may optionally and preferably be used as described above with regard to determining a ratio between a quantitative or semi-quantitative measurement of any marker described herein to any other marker described herein, and/or any other known marker, and/or any other marker. With regard to such a ratio between any marker described herein (or a combination thereof) and a known marker, more preferably the known marker comprises the “known protein” as described in greater detail below with regard to each cluster or gene.

According to other preferred embodiments of the present invention, a splice variant protein or a fragment thereof, or a splice variant nucleic acid sequence or a fragment thereof, may be featured as a biomarker for detecting prostate cancer and/or prostate pathology, such that a biomarker may optionally comprise any of the above.

According to still other preferred embodiments, the present invention optionally and preferably encompasses any amino acid sequence or fragment thereof encoded by a nucleic acid sequence corresponding to a splice variant protein as described herein. Any oligopeptide or peptide relating to such an amino acid sequence or fragment thereof may optionally also (additionally or alternatively) be used as a biomarker, including but not limited to the unique amino acid sequences of these proteins that are depicted as tails, heads, insertions, edges or bridges. The present invention also optionally encompasses antibodies capable of recognizing, and/or being elicited by, such oligopeptides or peptides.

The present invention also optionally and preferably encompasses any nucleic acid sequence or fragment thereof, or amino acid sequence or fragment thereof, corresponding to a splice variant of the present invention as described above, optionally for any application.

Non-limiting examples of methods or assays are described below.

The present invention also relates to kits based upon such diagnostic methods or assays.

Nucleic Acid Sequences and Oligonucleotides

Various embodiments of the present invention encompass nucleic acid sequences described hereinabove; fragments thereof, sequences hybridizable therewith, sequences homologous thereto, sequences encoding similar polypeptides with different codon usage, altered sequences characterized by mutations, such as deletion, insertion or substitution of one or more nucleotides, either naturally occurring or artificially induced, either randomly or in a targeted fashion.

The present invention encompasses nucleic acid sequences described herein; fragments thereof, sequences hybridizable therewith, sequences homologous thereto [e.g., at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 95% or more say 100% identical to the nucleic acid sequences set forth below], sequences encoding similar polypeptides with different codon usage, altered sequences characterized by mutations, such as deletion, insertion or substitution of one or more nucleotides, either naturally occurring or man induced, either randomly or in a targeted fashion. The present invention also encompasses homologous nucleic acid sequences (i.e., which form a part of a polynucleotide sequence of the present invention) which include sequence regions unique to the polynucleotides of the present invention.

In cases where the polynucleotide sequences of the present invention encode previously unidentified polypeptides, the present invention also encompasses novel polypeptides or portions thereof, which are encoded by the isolated polynucleotide and respective nucleic acid fragments thereof described hereinabove.

A “nucleic acid fragment” or an “oligonucleotide” or a “polynucleotide” are used herein interchangeably to refer to a polymer of nucleic acids. A polynucleotide sequence of the present invention refers to a single or double stranded nucleic acid sequences which is isolated and provided in the form of an RNA sequence, a complementary polynucleotide sequence (cDNA), a genomic polynucleotide sequence and/or a composite polynucleotide sequences (e.g., a combination of the above).

As used herein the phrase “complementary polynucleotide sequence” refers to a sequence, which results from reverse transcription of messenger RNA using a reverse transcriptase or any other RNA dependent DNA polymerase. Such a sequence can be subsequently amplified in vivo or in vitro using a DNA dependent DNA polymerase.

As used herein the phrase “genomic polynucleotide sequence” refers to a sequence derived (isolated) from a chromosome and thus it represents a contiguous portion of a chromosome.

As used herein the phrase “composite polynucleotide sequence” refers to a sequence, which is composed of genomic and cDNA sequences. A composite sequence can include some exonal sequences required to encode the polypeptide of the present invention, as well as some intronic sequences interposing therebetween. The intronic sequences can be of any source, including of other genes, and typically will include conserved splicing signal sequences. Such intronic sequences may further include cis acting expression regulatory elements.

Preferred embodiments of the present invention encompass oligonucleotide probes.

An example of an oligonucleotide probe which can be utilized by the present invention is a single stranded polynucleotide which includes a sequence complementary to the unique sequence region of any variant according to the present invention, including but not limited to a nucleotide sequence coding for an amino sequence of a bridge, tail, head and/or insertion according to the present invention, and/or the equivalent portions of any nucleotide sequence given herein (including but not limited to a nucleotide sequence of a node, segment or amplicon described herein).

Alternatively, an oligonucleotide probe of the present invention can be designed to hybridize with a nucleic acid sequence encompassed by any of the above nucleic acid sequences, particularly the portions specified above, including but not limited to a nucleotide sequence coding for an amino sequence of a bridge, tail, head and/or insertion according to the present invention, and/or the equivalent portions of any nucleotide sequence given herein (including but not limited to a nucleotide sequence of a node, segment or amplicon described herein).

Oligonucleotides designed according to the teachings of the present invention can be generated according to any oligonucleotide synthesis method known in the art such as enzymatic synthesis or solid phase synthesis. Equipment and reagents for executing solid-phase synthesis are commercially available from, for example, Applied Biosystems. Any other means for such synthesis may also be employed; the actual synthesis of the oligonucleotides is well within the capabilities of one skilled in the art and can be accomplished via established methodologies as detailed in, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., “Current Protocols in Molecular Biology”, John Wiley and Sons, Baltimore, Md. (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988) and “Oligonucleotide Synthesis” Gait, M. J., ed. (1984) utilizing solid phase chemistry, e.g. cyanoethyl phosphoramidite followed by deprotection, desalting and purification by for example, an automated trityl-on method or HPLC.

Oligonucleotides used according to this aspect of the present invention are those having a length selected from a range of about 10 to about 200 bases preferably about 15 to about 150 bases, more preferably about 20 to about 100 bases, most preferably about 20 to about 50 bases. Preferably, the oligonucleotide of the present invention features at least 17, at least 18, at least 19, at least 20, at least 22, at least 25, at least 30 or at least 40, bases specifically hybridizable with the biomarkers of the present invention.

The oligonucleotides of the present invention may comprise heterocylic nucleosides consisting of purines and the pyrimidines bases, bonded in a 3′ to 5′ phosphodiester linkage.

Preferably used oligonucleotides are those modified at one or more of the backbone, internucleoside linkages or bases, as is broadly described hereinunder.

Specific examples of preferred oligonucleotides useful according to this aspect of the present invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages. Oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone, as disclosed in U.S. Pat. Nos. 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; and 5,625,050.

Preferred modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkyl phosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3′-5′ to 5′-3′ or 2′-5′ to 5′-2′. Various salts, mixed salts and free acid forms can also be used.

Alternatively, modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts, as disclosed in U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439.

Other oligonucleotides which can be used according to the present invention, are those modified in both sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for complementation with the appropriate polynucleotide target. An example for such an oligonucleotide mimetic, includes peptide nucleic acid (PNA). United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Other backbone modifications, which can be used in the present invention are disclosed in U.S. Pat. No. 6,303,374.

Oligonucleotides of the present invention may also include base modifications or substitutions. As used herein, “unmodified” or “natural” bases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified bases include but are not limited to other synthetic and natural bases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further bases particularly useful for increasing the binding affinity of the oligomeric compounds of the invention include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. and are presently preferred base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.

Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates, which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-S-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety, as disclosed in U.S. Pat. No. 6,303,374.

It is not necessary for all positions in a given oligonucleotide molecule to be uniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an oligonucleotide.

It will be appreciated that oligonucleotides of the present invention may include further modifications for more efficient use as diagnostic agents and/or to increase bioavailability, therapeutic efficacy and reduce cytotoxicity.

To enable cellular expression of the polynucleotides of the present invention, a nucleic acid construct according to the present invention may be used, which includes at least a coding region of one of the above nucleic acid sequences, and further includes at least one cis acting regulatory element. As used herein, the phrase “cis acting regulatory element” refers to a polynucleotide sequence, preferably a promoter, which binds a trans acting regulator and regulates the transcription of a coding sequence located downstream thereto.

Any suitable promoter sequence can be used by the nucleic acid construct of the present invention.

Preferably, the promoter utilized by the nucleic acid construct of the present invention is active in the specific cell population transformed. Examples of cell type-specific and/or tissue-specific promoters include promoters such as albumin that is liver specific, lymphoid specific promoters [Calame et al., (1988) Adv. Immunol. 43:235-275]; in particular promoters of T-cell receptors [Winoto et al., (1989) EMBO J. 8:729-733] and immunoglobulins; [Banerji et al. (1983) Cell 33729-740], neuron-specific promoters such as the neurofilament promoter [Byrne et al. (1989) Proc. Natl. Acad. Sci. USA 86:5473-5477], pancreas-specific promoters [Edlunch et al. (1985) Science 230:912-916] or mammary gland-specific promoters such as the milk whey promoter (U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). The nucleic acid construct of the present invention can further include an enhancer, which can be adjacent or distant to the promoter sequence and can function in up regulating the transcription therefrom.

The nucleic acid construct of the present invention preferably further includes an appropriate selectable marker and/or an origin of replication. Preferably, the nucleic acid construct utilized is a shuttle vector, which can propagate both in E. coli (wherein the construct comprises an appropriate selectable marker and origin of replication) and be compatible for propagation in cells, or integration in a gene and a tissue of choice. The construct according to the present invention can be, for example, a plasmid, a bacmid, a phagemid, a cosmid, a phage, a virus or an artificial chromosome.

Examples of suitable constructs include, but are not limited to, pcDNA3, pcDNA3.1 (+/−), pGL3, PzeoSV2 (+/−), pDisplay, pEF/myc/cyto, pCMV/myc/cyto each of which is commercially available from Invitrogen Co. (www “dot” invitrogen “dot” com). Examples of retroviral vector and packaging systems are those sold by Clontech, San Diego, Calif., including Retro-X vectors pLNCX and pLXSN, which permit cloning into multiple cloning sites and the transgene is transcribed from CMV promoter. Vectors derived from Mo-MuLV are also included such as pBabe, where the transgene will be transcribed from the 5′LTR promoter.

Currently preferred in vivo nucleic acid transfer techniques include transfection with viral or non-viral constructs, such as adenovirus, lentivirus, Herpes simplex I virus, or adeno-associated virus (AAV) and lipid-based systems. Useful lipids for lipid-mediated transfer of the gene are, for example, DOTMA, DOPE, and DC-Chol [Tonkinson et al., Cancer Investigation, 14(1): 54-65 (1996)]. The most preferred constructs for use in gene therapy are viruses, most preferably adenoviruses, AAV, lentiviruses, or retroviruses. A viral construct such as a retroviral construct includes at least one transcriptional promoter/enhancer or locus-defining element(s), or other elements that control gene expression by other means such as alternate splicing, nuclear RNA export, or post-translational modification of messenger. Such vector constructs also include a packaging signal, long terminal repeats (LTRs) or portions thereof, and positive and negative strand primer binding sites appropriate to the virus used, unless it is already present in the viral construct. In addition, such a construct typically includes a signal sequence for secretion of the peptide from a host cell in which it is placed. Preferably the signal sequence for this purpose is a mammalian signal sequence or the signal sequence of the polypeptide variants of the present invention. Optionally, the construct may also include a signal that directs polyadenylation, as well as one or more restriction sites and a translation termination sequence. By way of example, such constructs will typically include a 5′ LTR, a tRNA binding site, a packaging signal, an origin of second-strand DNA synthesis, and a 3′ LTR or a portion thereof. Other vectors can be used that are non-viral, such as cationic lipids, polylysine, and dendrimers.

Hybridization Assays

Detection of a nucleic acid of interest in a biological sample may optionally be effected by hybridization-based assays using an oligonucleotide probe (non-limiting examples of probes according to the present invention were previously described).

Traditional hybridization assays include PCR, RT-PCR, Real-time PCR, RNase protection, in-situ hybridization, primer extension, Southern blots (DNA detection), dot or slot blots (DNA, RNA), and Northern blots (RNA detection) (NAT type assays are described in greater detail below). More recently, PNAs have been described (Nielsen et al. 1999, Current Opin. Biotechnol. 10:71-75). Other detection methods include kits containing probes on a dipstick setup and the like.

Hybridization based assays which allow the detection of a variant of interest (i.e., DNA or RNA) in a biological sample rely on the use of oligonucleotides which can be 10, 15, 20, or to 100 nucleotides long preferably from 10 to 50, more preferably from 40 to 50 nucleotides long.

Thus, the isolated polynucleotides (oligonucleotides) of the present invention are preferably hybridizable with any of the herein described nucleic acid sequences under moderate to stringent hybridization conditions.

Moderate to stringent hybridization conditions are characterized by a hybridization solution such as containing 10% dextrane sulfate, 1 M NaCl, 1% SDS and 5×106 cpm 32P labeled probe, at 65° C., with a final wash solution of 0.2×SSC and 0.1% SDS and final wash at 65° C. and whereas moderate hybridization is effected using a hybridization solution containing 10% dextrane sulfate, 1 M NaCl, 1% SDS and 5×106 cpm 32P labeled probe, at 65° C., with a final wash solution of 1×SSC and 0.1% SDS and final wash at 50° C.

More generally, hybridization of short nucleic acids (below 200 bp in length, e.g. 17-40 bp in length) can be effected using the following exemplary hybridization protocols which can be modified according to the desired stringency; (i) hybridization solution of 6×SSC and 1% SDS or 3 M TMACI, 0.01 M sodium phosphate (pH 6.8), 1 mM EDTA (pH 7.6), 0.5% SDS, 100 μg/ml denatured salmon sperm DNA and 0.1% nonfat dried milk, hybridization temperature of 1-1.5° C. below the Tm, final wash solution of 3 M TMACI, 0.01 M sodium phosphate (pH 6.8), 1 mM EDTA (pH 7.6), 0.5% SDS at 1-1.5° C. below the Tm; (ii) hybridization solution of 6×SSC and 0.1% SDS or 3 M TMACI, 0.01 M sodium phosphate (pH 6.8), 1 mM EDTA (pH 7.6), 0.5% SDS, 100 μg/ml denatured salmon sperm DNA and 0.1% nonfat dried milk, hybridization temperature of 2-2.5° C. below the Tm, final wash solution of 3 M TMACI, 0.01 M sodium phosphate (pH 6.8), 1 mM EDTA (pH 7.6), 0.5% SDS at 1-1.5° C. below the Tm, final wash solution of 6×SSC, and final wash at 22° C.; (iii) hybridization solution of 6×SSC and 1% SDS or 3 M TMACI, 0.01 M sodium phosphate (pH 6.8), 1 mM EDTA (pH 7.6), 0.5% SDS, 100 μg/ml denatured salmon sperm DNA and 0.1% nonfat dried milk, hybridization temperature.

The detection of hybrid duplexes can be carried out by a number of methods. Typically, hybridization duplexes are separated from unhybridized nucleic acids and the labels bound to the duplexes are then detected. Such labels refer to radioactive, fluorescent, biological or enzymatic tags or labels of standard use in the art. A label can be conjugated to either the oligonucleotide probes or the nucleic acids derived from the biological sample.

Probes can be labeled according to numerous well known methods. Non-limiting examples of radioactive labels include 3H, 14C, 32P, and 35S, Non-limiting examples of detectable markers include ligands, fluorophores, chemiluminescent agents, enzymes, and antibodies. Other detectable markers for use with probes, which can enable an increase in sensitivity of the method of the invention, include biotin and radio-nucleotides. It will become evident to the person of ordinary skill that the choice of a particular label dictates the manner in which it is bound to the probe.

For example, oligonucleotides of the present invention can be labeled subsequent to synthesis, by incorporating biotinylated dNTPs or rNTP, or some similar means (e.g., photo-cross-linking a psoralen derivative of biotin to RNAs), followed by addition of labeled streptavidin (e.g., phycoerythrin-conjugated streptavidin) or the equivalent. Alternatively, when fluorescently-labeled oligonucleotide probes are used, fluorescein, lissamine, phycoerythrin, rhodamine (Perkin Elmer Cetus), Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7, FluorX (Amersham) and others [e.g., Kricka et al. (1992), Academic Press San Diego, Calif] can be attached to the oligonucleotides.

Those skilled in the art will appreciate that wash steps may be employed to wash away excess target DNA or probe as well as unbound conjugate. Further, standard heterogeneous assay formats are suitable for detecting the hybrids using the labels present on the oligonucleotide primers and probes.

It will be appreciated that a variety of controls may be usefully employed to improve accuracy of hybridization assays. For instance, samples may be hybridized to an irrelevant probe and treated with RNAse A prior to hybridization, to assess false hybridization.

Although the present invention is not specifically dependent on the use of a label for the detection of a particular nucleic acid sequence, such a label might be beneficial, by increasing the sensitivity of the detection. Furthermore, it enables automation. Probes can be labeled according to numerous well known methods.

As commonly known, radioactive nucleotides can be incorporated into probes of the invention by several methods. Non-limiting examples of radioactive labels include 3H, 14C, 32P, and 35S Those skilled in the art will appreciate that wash steps may be employed to wash away excess target DNA or probe as well as unbound conjugate. Further, standard heterogeneous assay formats are suitable for detecting the hybrids using the labels present on the oligonucleotide primers and probes.

It will be appreciated that a variety of controls may be usefully employed to improve accuracy of hybridization assays.

Probes of the invention can be utilized with naturally occurring sugar-phosphate backbones as well as modified backbones including phosphorothioates, dithionates, alkyl phosphonates and a-nucleotides and the like. Probes of the invention can be constructed of either ribonucleic acid (RNA) or deoxyribonucleic acid (DNA), and preferably of DNA.

NAT Assays

Detection of a nucleic acid of interest in a biological sample may also optionally be effected by NAT-based assays, which involve nucleic acid amplification technology, such as PCR for example (or variations thereof such as real-time PCR for example).

As used herein, a “primer” defines an oligonucleotide which is capable of annealing to (hybridizing with) a target sequence, thereby creating a double stranded region which can serve as an initiation point for DNA synthesis under suitable conditions.

Amplification of a selected, or target, nucleic acid sequence may be carried out by a number of suitable methods. See generally Kwoh et al., 1990, Am. Biotechnol. Lab. 8:14 Numerous amplification techniques have been described and can be readily adapted to suit particular needs of a person of ordinary skill. Non-limiting examples of amplification techniques include polymerase chain reaction (PCR), ligase chain reaction (LCR), strand displacement amplification (SDA), transcription-based amplification, the q3 replicase system and NASBA (Kwoh et al., 1989, Proc. Natl. Acad. Sci. USA 86, 1173-1177; Lizardi et al., 1988, BioTechnology 6:1197-1202; Malek et al., 1994, Methods Mol. Biol., 28:253-260; and Sambrook et al., 1989, supra).

The terminology “amplification pair” (or “primer pair”) refers herein to a pair of oligonucleotides (oligos) of the present invention, which are selected to be used together in amplifying a selected nucleic acid sequence by one of a number of types of amplification processes, preferably a polymerase chain reaction. Other types of amplification processes include ligase chain reaction, strand displacement amplification, or nucleic acid sequence-based amplification, as explained in greater detail below. As commonly known in the art, the oligos are designed to bind to a complementary sequence under selected conditions.

In one particular embodiment, amplification of a nucleic acid sample from a patient is amplified under conditions which favor the amplification of the most abundant differentially expressed nucleic acid. In one preferred embodiment, RT-PCR is carried out on an mRNA sample from a patient under conditions which favor the amplification of the most abundant mRNA. In another preferred embodiment, the amplification of the differentially expressed nucleic acids is carried out simultaneously. It will be realized by a person skilled in the art that such methods could be adapted for the detection of differentially expressed proteins instead of differentially expressed nucleic acid sequences.

The nucleic acid (i.e. DNA or RNA) for practicing the present invention may be obtained according to well known methods.

Oligonucleotide primers of the present invention may be of any suitable length, depending on the particular assay format and the particular needs and targeted genomes employed. Optionally, the oligonucleotide primers are at least 12 nucleotides in length, preferably between 15 and 24 molecules, and they may be adapted to be especially suited to a chosen nucleic acid amplification system. As commonly known in the art, the oligonucleotide primers can be designed by taking into consideration the melting point of hybridization thereof with its targeted sequence (Sambrook et al., 1989, Molecular Cloning—A Laboratory Manual, 2nd Edition, CSH Laboratories; Ausubel et al., 1989, in Current Protocols in Molecular Biology, John Wiley & Sons Inc., N.Y.).

It will be appreciated that antisense oligonucleotides may be employed to quantify expression of a splice isoform of interest. Such detection is effected at the pre-mRNA level. Essentially the ability to quantitate transcription from a splice site of interest can be effected based on splice site accessibility. Oligonucleotides may compete with splicing factors for the splice site sequences. Thus, low activity of the antisense oligonucleotide is indicative of splicing activity.

The polymerase chain reaction and other nucleic acid amplification reactions are well known in the art (various non-limiting examples of these reactions are described in greater detail below). The pair of oligonucleotides according to this aspect of the present invention are preferably selected to have compatible melting temperatures (Tm), e.g., melting temperatures which differ by less than that 7° C., preferably less than 5° C., more preferably less than 4° C., most preferably less than 3° C., ideally between 3° C. and 0° C.

Polymerase Chain Reaction (PCR): The polymerase chain reaction (PCR), as described in U.S. Pat. Nos. 4,683,195 and 4,683,202 to Mullis and Mullis et al., is a method of increasing the concentration of a segment of target sequence in a mixture of genomic DNA without cloning or purification. This technology provides one approach to the problems of low target sequence concentration. PCR can be used to directly increase the concentration of the target to an easily detectable level. This process for amplifying the target sequence involves the introduction of a molar excess of two oligonucleotide primers which are complementary to their respective strands of the double-stranded target sequence to the DNA mixture containing the desired target sequence. The mixture is denatured and then allowed to hybridize. Following hybridization, the primers are extended with polymerase so as to form complementary strands. The steps of denaturation, hybridization (annealing), and polymerase extension (elongation) can be repeated as often as needed, in order to obtain relatively high concentrations of a segment of the desired target sequence.

The length of the segment of the desired target sequence is determined by the relative positions of the primers with respect to each other, and, therefore, this length is a controllable parameter. Because the desired segments of the target sequence become the dominant sequences (in terms of concentration) in the mixture, they are said to be “PCR-amplified.”

Ligase Chain Reaction (LCR or LAR): The ligase chain reaction [LCR; sometimes referred to as “Ligase Amplification Reaction” (LAR)] has developed into a well-recognized alternative method of amplifying nucleic acids. In LCR, four oligonucleotides, two adjacent oligonucleotides which uniquely hybridize to one strand of target DNA, and a complementary set of adjacent oligonucleotides, which hybridize to the opposite strand are mixed and DNA ligase is added to the mixture. Provided that there is complete complementarity at the junction, ligase will covalently link each set of hybridized molecules. Importantly, in LCR, two probes are ligated together only when they base-pair with sequences in the target sample, without gaps or mismatches. Repeated cycles of denaturation, and ligation amplify a short segment of DNA. LCR has also been used in combination with PCR to achieve enhanced detection of single-base changes: see for example Segev, PCT Publication No. W09001069 A1 (1990). However, because the four oligonucleotides used in this assay can pair to form two short ligatable fragments, there is the potential for the generation of target-independent background signal. The use of LCR for mutant screening is limited to the examination of specific nucleic acid positions.

Self-Sustained Synthetic Reaction (3SR/NASBA): The self-sustained sequence replication reaction (3SR) is a transcription-based in vitro amplification system that can exponentially amplify RNA sequences at a uniform temperature. The amplified RNA can then be utilized for mutation detection. In this method, an oligonucleotide primer is used to add a phage RNA polymerase promoter to the 5′ end of the sequence of interest. In a cocktail of enzymes and substrates that includes a second primer, reverse transcriptase, RNase H, RNA polymerase and ribo- and deoxyribonucleoside triphosphates, the target sequence undergoes repeated rounds of transcription, cDNA synthesis and second-strand synthesis to amplify the area of interest. The use of 3SR to detect mutations is kinetically limited to screening small segments of DNA (e.g., 200-300 base pairs).

Q-Beta (Qβ) Replicase: In this method, a probe which recognizes the sequence of interest is attached to the replicatable RNA template for Qβ replicase. A previously identified major problem with false positives resulting from the replication of unhybridized probes has been addressed through use of a sequence-specific ligation step. However, available thermostable DNA ligases are not effective on this RNA substrate, so the ligation must be performed by T4 DNA ligase at low temperatures (37 degrees C.). This prevents the use of high temperature as a means of achieving specificity as in the LCR, the ligation event can be used to detect a mutation at the junction site, but not elsewhere.

A successful diagnostic method must be very specific. A straight-forward method of controlling the specificity of nucleic acid hybridization is by controlling the temperature of the reaction. While the 3SR/NASBA, and Qβ systems are all able to generate a large quantity of signal, one or more of the enzymes involved in each cannot be used at high temperature (i.e., >55 degrees C.). Therefore the reaction temperatures cannot be raised to prevent non-specific hybridization of the probes. If probes are shortened in order to make them melt more easily at low temperatures, the likelihood of having more than one perfect match in a complex genome increases. For these reasons, PCR and LCR currently dominate the research field in detection technologies.

The basis of the amplification procedure in the PCR and LCR is the fact that the products of one cycle become usable templates in all subsequent cycles, consequently doubling the population with each cycle. The final yield of any such doubling system can be expressed as: (1+X)n=y, where “X” is the mean efficiency (percent copied in each cycle), “n” is the number of cycles, and “y” is the overall efficiency, or yield of the reaction. If every copy of a target DNA is utilized as a template in every cycle of a polymerase chain reaction, then the mean efficiency is 100%. If 20 cycles of PCR are performed, then the yield will be 220, or 1,048,576 copies of the starting material. If the reaction conditions reduce the mean efficiency to 85%, then the yield in those 20 cycles will be only 1.8520, or 220,513 copies of the starting material. In other words, a PCR running at 85% efficiency will yield only 21% as much final product, compared to a reaction running at 100% efficiency. A reaction that is reduced to 50% mean efficiency will yield less than 1% of the possible product.

In practice, routine polymerase chain reactions rarely achieve the theoretical maximum yield, and PCRs are usually run for more than 20 cycles to compensate for the lower yield. At 50% mean efficiency, it would take 34 cycles to achieve the million-fold amplification theoretically possible in 20, and at lower efficiencies, the number of cycles required becomes prohibitive. In addition, any background products that amplify with a better mean efficiency than the intended target will become the dominant products.

Also, many variables can influence the mean efficiency of PCR, including target DNA length and secondary structure, primer length and design, primer and dNTP concentrations, and buffer composition, to name but a few. Contamination of the reaction with exogenous DNA (e.g., DNA spilled onto lab surfaces) or cross-contamination is also a major consideration. Reaction conditions must be carefully optimized for each different primer pair and target sequence, and the process can take days, even for an experienced investigator. The laboriousness of this process, including numerous technical considerations and other factors, presents a significant drawback to using PCR in the clinical setting. Indeed, PCR has yet to penetrate the clinical market in a significant way. The same concerns arise with LCR, as LCR must also be optimized to use different oligonucleotide sequences for each target sequence. In addition, both methods require expensive equipment, capable of precise temperature cycling.

Many applications of nucleic acid detection technologies, such as in studies of allelic variation, involve not only detection of a specific sequence in a complex background, but also the discrimination between sequences with few, or single, nucleotide differences. One method of the detection of allele-specific variants by PCR is based upon the fact that it is difficult for Taq polymerase to synthesize a DNA strand when there is a mismatch between the template strand and the 3′ end of the primer. An allele-specific variant may be detected by the use of a primer that is perfectly matched with only one of the possible alleles; the mismatch to the other allele acts to prevent the extension of the primer, thereby preventing the amplification of that sequence. This method has a substantial limitation in that the base composition of the mismatch influences the ability to prevent extension across the mismatch, and certain mismatches do not prevent extension or have only a minimal effect.

A similar 3′-mismatch strategy is used with greater effect to prevent ligation in the LCR. Any mismatch effectively blocks the action of the thermostable ligase, but LCR still has the drawback of target-independent background ligation products initiating the amplification. Moreover, the combination of PCR with subsequent LCR to identify the nucleotides at individual positions is also a clearly cumbersome proposition for the clinical laboratory.

The direct detection method according to various preferred embodiments of the present invention may be, for example a cycling probe reaction (CPR) or a branched DNA analysis.

When a sufficient amount of a nucleic acid to be detected is available, there are advantages to detecting that sequence directly, instead of making more copies of that target, (e.g., as in PCR and LCR). Most notably, a method that does not amplify the signal exponentially is more amenable to quantitative analysis. Even if the signal is enhanced by attaching multiple dyes to a single oligonucleotide, the correlation between the final signal intensity and amount of target is direct. Such a system has an additional advantage that the products of the reaction will not themselves promote further reaction, so contamination of lab surfaces by the products is not as much of a concern. Recently devised techniques have sought to eliminate the use of radioactivity and/or improve the sensitivity in automatable formats. Two examples are the “Cycling Probe Reaction” (CPR), and “Branched DNA” (bDNA).

Cycling probe reaction (CPR): The cycling probe reaction (CPR), uses a long chimeric oligonucleotide in which a central portion is made of RNA while the two termini are made of DNA. Hybridization of the probe to a target DNA and exposure to a thermostable RNase H causes the RNA portion to be digested. This destabilizes the remaining DNA portions of the duplex, releasing the remainder of the probe from the target DNA and allowing another probe molecule to repeat the process. The signal, in the form of cleaved probe molecules, accumulates at a linear rate. While the repeating process increases the signal, the RNA portion of the oligonucleotide is vulnerable to RNases that may carried through sample preparation.

Branched DNA: Branched DNA (bDNA), involves oligonucleotides with branched structures that allow each individual oligonucleotide to carry 35 to 40 labels (e.g., alkaline phosphatase enzymes). While this enhances the signal from a hybridization event, signal from non-specific binding is similarly increased.

The detection of at least one sequence change according to various preferred embodiments of the present invention may be accomplished by, for example restriction fragment length polymorphism (RFLP analysis), allele specific oligonucleotide (ASO) analysis, Denaturing/Temperature Gradient Gel Electrophoresis (DGGE/TGGE), Single-Strand Conformation Polymorphism (SSCP) analysis or Dideoxy fingerprinting (ddF).

The demand for tests which allow the detection of specific nucleic acid sequences and sequence changes is growing rapidly in clinical diagnostics. As nucleic acid sequence data for genes from humans and pathogenic organisms accumulates, the demand for fast, cost-effective, and easy-to-use tests for as yet mutations within specific sequences is rapidly increasing.

A handful of methods have been devised to scan nucleic acid segments for mutations. One option is to determine the entire gene sequence of each test sample (e.g., a bacterial isolate). For sequences under approximately 600 nucleotides, this may be accomplished using amplified material (e.g., PCR reaction products). This avoids the time and expense associated with cloning the segment of interest. However, specialized equipment and highly trained personnel are required, and the method is too labor-intense and expensive to be practical and effective in the clinical setting.

In view of the difficulties associated with sequencing, a given segment of nucleic acid may be characterized on several other levels. At the lowest resolution, the size of the molecule can be determined by electrophoresis by comparison to a known standard run on the same gel. A more detailed picture of the molecule may be achieved by cleavage with combinations of restriction enzymes prior to electrophoresis, to allow construction of an ordered map. The presence of specific sequences within the fragment can be detected by hybridization of a labeled probe, or the precise nucleotide sequence can be determined by partial chemical degradation or by primer extension in the presence of chain-terminating nucleotide analogs.

Restriction fragment length polymorphism (RFLP): For detection of single-base differences between like sequences, the requirements of the analysis are often at the highest level of resolution. For cases in which the position of the nucleotide in question is known in advance, several methods have been developed for examining single base changes without direct sequencing. For example, if a mutation of interest happens to fall within a restriction recognition sequence, a change in the pattern of digestion can be used as a diagnostic tool (e.g., restriction fragment length polymorphism [RFLP] analysis).

Single point mutations have been also detected by the creation or destruction of RFLPs. Mutations are detected and localized by the presence and size of the RNA fragments generated by cleavage at the mismatches. Single nucleotide mismatches in DNA heteroduplexes are also recognized and cleaved by some chemicals, providing an alternative strategy to detect single base substitutions, generically named the “Mismatch Chemical Cleavage” (MCC). However, this method requires the use of osmium tetroxide and piperidine, two highly noxious chemicals which are not suited for use in a clinical laboratory.

RFLP analysis suffers from low sensitivity and requires a large amount of sample. When RFLP analysis is used for the detection of point mutations, it is, by its nature, limited to the detection of only those single base changes which fall within a restriction sequence of a known restriction endonuclease. Moreover, the majority of the available enzymes have 4 to 6 base-pair recognition sequences, and cleave too frequently for many large-scale DNA manipulations. Thus, it is applicable only in a small fraction of cases, as most mutations do not fall within such sites.

A handful of rare-cutting restriction enzymes with 8 base-pair specificities have been isolated and these are widely used in genetic mapping, but these enzymes are few in number, are limited to the recognition of G+C-rich sequences, and cleave at sites that tend to be highly clustered. Recently, endonucleases encoded by group I introns have been discovered that might have greater than 12 base-pair specificity, but again, these are few in number.

Allele specific oligonucleotide (ASO): If the change is not in a recognition sequence, then allele-specific oligonucleotides (ASOs), can be designed to hybridize in proximity to the mutated nucleotide, such that a primer extension or ligation event can bused as the indicator of a match or a mis-match. Hybridization with radioactively labeled allelic specific oligonucleotides (ASO) also has been applied to the detection of specific point mutations. The method is based on the differences in the melting temperature of short DNA fragments differing by a single nucleotide. Stringent hybridization and washing conditions can differentiate between mutant and wild-type alleles. The ASO approach applied to PCR products also has been extensively utilized by various researchers to detect and characterize point mutations in ras genes and gsp/gip oncogenes. Because of the presence of various nucleotide changes in multiple positions, the ASO method requires the use of many oligonucleotides to cover all possible oncogenic mutations.

With either of the techniques described above (i.e., RFLP and ASO), the precise location of the suspected mutation must be known in advance of the test. That is to say, they are inapplicable when one needs to detect the presence of a mutation within a gene or sequence of interest.

Denaturing/Temperature Gradient Gel Electrophoresis (DGGE/TGGE): Two other methods rely on detecting changes in electrophoretic mobility in response to minor sequence changes. One of these methods, termed “Denaturing Gradient Gel Electrophoresis” (DGGE) is based on the observation that slightly different sequences will display different patterns of local melting when electrophoretically resolved on a gradient gel. In this manner, variants can be distinguished, as differences in melting properties of homoduplexes versus heteroduplexes differing in a single nucleotide can detect the presence of mutations in the target sequences because of the corresponding changes in their electrophoretic mobilities. The fragments to be analyzed, usually PCR products, are “clamped” at one end by a long stretch of G-C base pairs (30-80) to allow complete denaturation of the sequence of interest without complete dissociation of the strands. The attachment of a GC “clamp” to the DNA fragments increases the fraction of mutations that can be recognized by DGGE. Attaching a GC clamp to one primer is critical to ensure that the amplified sequence has a low dissociation temperature. Modifications of the technique have been developed, using temperature gradients, and the method can be also applied to RNA:RNA duplexes.

Limitations on the utility of DGGE include the requirement that the denaturing conditions must be optimized for each type of DNA to be tested. Furthermore, the method requires specialized equipment to prepare the gels and maintain the needed high temperatures during electrophoresis. The expense associated with the synthesis of the clamping tail on one oligonucleotide for each sequence to be tested is also a major consideration. In addition, long running times are required for DGGE. The long running time of DGGE was shortened in a modification of DGGE called constant denaturant gel electrophoresis (CDGE). CDGE requires that gels be performed under different denaturant conditions in order to reach high efficiency for the detection of mutations.

A technique analogous to DGGE, termed temperature gradient gel electrophoresis (TGGE), uses a thermal gradient rather than a chemical denaturant gradient. TGGE requires the use of specialized equipment which can generate a temperature gradient perpendicularly oriented relative to the electrical field. TGGE can detect mutations in relatively small fragments of DNA therefore scanning of large gene segments requires the use of multiple PCR products prior to running the gel.

Single-Strand Conformation Polymorphism (SSCP): Another common method, called “Single-Strand Conformation Polymorphism” (SSCP) was developed by Hayashi, Sekya and colleagues and is based on the observation that single strands of nucleic acid can take on characteristic conformations in non-denaturing conditions, and these conformations influence electrophoretic mobility. The complementary strands assume sufficiently different structures that one strand may be resolved from the other. Changes in sequences within the fragment will also change the conformation, consequently altering the mobility and allowing this to be used as an assay for sequence variations.

The SSCP process involves denaturing a DNA segment (e.g., a PCR product) that is labeled on both strands, followed by slow electrophoretic separation on a non-denaturing polyacrylamide gel, so that intra-molecular interactions can form and not be disturbed during the run. This technique is extremely sensitive to variations in gel composition and temperature. A serious limitation of this method is the relative difficulty encountered in comparing data generated in different laboratories, under apparently similar conditions.

Dideoxy fingerprinting (ddF): The dideoxy fingerprinting (ddF) is another technique developed to scan genes for the presence of mutations. The ddF technique combines components of Sanger dideoxy sequencing with SSCP. A dideoxy sequencing reaction is performed using one dideoxy terminator and then the reaction products are electrophoresed on nondenaturing polyacrylamide gels to detect alterations in mobility of the termination segments as in SSCP analysis. While ddF is an improvement over SSCP in terms of increased sensitivity, ddF requires the use of expensive dideoxynucleotides and this technique is still limited to the analysis of fragments of the size suitable for SSCP (i.e., fragments of 200-300 bases for optimal detection of mutations).

In addition to the above limitations, all of these methods are limited as to the size of the nucleic acid fragment that can be analyzed. For the direct sequencing approach, sequences of greater than 600 base pairs require cloning, with the consequent delays and expense of either deletion sub-cloning or primer walking, in order to cover the entire fragment. SSCP and DGGE have even more severe size limitations. Because of reduced sensitivity to sequence changes, these methods are not considered suitable for larger fragments. Although SSCP is reportedly able to detect 90% of single-base substitutions within a 200 base-pair fragment, the detection drops to less than 50% for 400 base pair fragments. Similarly, the sensitivity of DGGE decreases as the length of the fragment reaches 500 base-pairs. The ddF technique, as a combination of direct sequencing and SSCP, is also limited by the relatively small size of the DNA that can be screened.

According to a presently preferred embodiment of the present invention the step of searching for any of the nucleic acid sequences described here, in tumor cells or in cells derived from a cancer patient is effected by any suitable technique, including, but not limited to, nucleic acid sequencing, polymerase chain reaction, ligase chain reaction, self-sustained synthetic reaction, Qβ-Replicase, cycling probe reaction, branched DNA, restriction fragment length polymorphism analysis, mismatch chemical cleavage, heteroduplex analysis, allele-specific oligonucleotides, denaturing gradient gel electrophoresis, constant denaturant gel electrophoresis, temperature gradient gel electrophoresis and dideoxy fingerprinting.

Detection may also optionally be performed with a chip or other such device. The nucleic acid sample which includes the candidate region to be analyzed is preferably isolated, amplified and labeled with a reporter group. This reporter group can be a fluorescent group such as phycoerythrin. The labeled nucleic acid is then incubated with the probes immobilized on the chip using a fluidics station. describe the fabrication of fluidics devices and particularly microcapillary devices, in silicon and glass substrates.

Once the reaction is completed, the chip is inserted into a scanner and patterns of hybridization are detected. The hybridization data is collected, as a signal emitted from the reporter groups already incorporated into the nucleic acid, which is now bound to the probes attached to the chip. Since the sequence and position of each probe immobilized on the chip is known, the identity of the nucleic acid hybridized to a given probe can be determined.

It will be appreciated that when utilized along with automated equipment, the above described detection methods can be used to screen multiple samples for a disease and/or pathological condition both rapidly and easily.

Amino Acid Sequences and Peptides

The terms “polypeptide,” “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an analog or mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers. Polypeptides can be modified, e.g., by the addition of carbohydrate residues to form glycoproteins. The terms “polypeptide,” “peptide” and “protein” include glycoproteins, as well as non-glycoproteins.

Polypeptide products can be biochemically synthesized such as by employing standard solid phase techniques. Such methods include but are not limited to exclusive solid phase synthesis, partial solid phase synthesis methods, fragment condensation, classical solution synthesis. These methods are preferably used when the peptide is relatively short (i.e., 10 kDa) and/or when it cannot be produced by recombinant techniques (i.e., not encoded by a nucleic acid sequence) and therefore involves different chemistry.

Solid phase polypeptide synthesis procedures are well known in the art and further described by John Morrow Stewart and Janis Dillaha Young, Solid Phase Peptide Syntheses (2nd Ed., Pierce Chemical Company, 1984).

Synthetic polypeptides can optionally be purified by preparative high performance liquid chromatography [Creighton T. (1983) Proteins, structures and molecular principles. WH Freeman and Co. N.Y.], after which their composition can be confirmed via amino acid sequencing.

In cases where large amounts of a polypeptide are desired, it can be generated using recombinant techniques such as described by Bitter et al., (1987) Methods in Enzymol. 153:516-544, Studier et al. (1990) Methods in Enzymol. 185:60-89, Brisson et al. (1984) Nature 310:511-514, Takamatsu et al. (1987) EMBO J. 6:307-311, Coruzzi et al. (1984) EMBO J. 3:1671-1680 and Brogli et al., (1984) Science 224:838-843, Gurley et al. (1986) Mol. Cell. Biol. 6:559-565 and Weissbach & Weissbach, 1988, Methods for Plant Molecular Biology, Academic Press, NY, Section VIII, pp 421-463.

The present invention also encompasses polypeptides encoded by the polynucleotide sequences of the present invention, as well as polypeptides according to the amino acid sequences described herein. The present invention also encompasses homologues of these polypeptides, such homologues can be at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 95% or more say 100% homologous to the amino acid sequences set forth below, as can be determined using BlastP software of the National Center of Biotechnology Information (NCBI) using default parameters, optionally and preferably including the following: filtering on (this option filters repetitive or low-complexity sequences from the query using the Seg (protein) program), scoring matrix is BLOSUM62 for proteins, word size is 3, E value is 10, gap costs are 11, 1 (initialization and extension), and number of alignments shown is 50. Optionally and preferably, nucleic acid sequence identity/homology is determined with BlastN software of the National Center of Biotechnology Information (NCBI) using default parameters, which preferably include using the DUST filter program, and also preferably include having an E value of 10, filtering low complexity sequences and a word size of 11. Finally, the present invention also encompasses fragments of the above described polypeptides and polypeptides having mutations, such as deletions, insertions or substitutions of one or more amino acids, either naturally occurring or artificially induced, either randomly or in a targeted fashion.

It will be appreciated that peptides identified according the present invention may be degradation products, synthetic peptides or recombinant peptides as well as peptidomimetics, typically, synthetic peptides and peptoids and semipeptoids which are peptide analogs, which may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells. Such modifications include, but are not limited to N terminus modification, C terminus modification, peptide bond modification, including, but not limited to, CH2-NH, CH2-S, CH2-S═O, O═C—NH, CH2-O, CH2-CH2, S═C—NH, CH═CH or CF═CH, backbone modifications, and residue modification. Methods for preparing peptidomimetic compounds are well known in the art and are specified. Further details in this respect are provided hereinunder.

Peptide bonds (—CO—NH—) within the peptide may be substituted, for example, by N-methylated bonds (—N(CH3)-CO—), ester bonds (—C(R)H—C—O—O—C(R)—N—), ketomethylen bonds (—CO—CH2-), α-aza bonds (—NH—N(R)—CO—), wherein R is any alkyl, e.g., methyl, carba bonds (—CH2-NH—), hydroxyethylene bonds (—CH(OH)—CH2—), thioamide bonds (—CS—NH—), olefinic double bonds (—CH═CH—), retro amide bonds (—NH—CO—), peptide derivatives (—N(R)—CH2—CO—), wherein R is the “normal” side chain, naturally presented on the carbon atom.

These modifications can occur at any of the bonds along the peptide chain and even at several (2-3) at the same time.

Natural aromatic amino acids, Trp, Tyr and Phe, may be substituted for synthetic non-natural acid such as Phenylglycine, TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.

In addition to the above, the peptides of the present invention may also include one or more modified amino acids or one or more non-amino acid monomers (e.g. fatty acids, complex carbohydrates etc).

As used herein in the specification and in the claims section below the term “amino acid” or “amino acids” is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phosphothreonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine. Furthermore, the term “amino acid” includes both D- and L-amino acids.

Table 1 non-conventional or modified amino acids which can be used with the present invention.

TABLE 1 Non-conventional amino acid Code Non-conventional amino acid Code α-aminobutyric acid Abu L-N-methylalanine Nmala α-amino-α-methylbutyrate Mgabu L-N-methylarginine Nmarg aminocyclopropane- Cpro L-N-methylasparagine Nmasn Carboxylate L-N-methylaspartic acid Nmasp aminoisobutyric acid Aib L-N-methylcysteine Nmcys aminonorbornyl- Norb L-N-methylglutamine Nmgin Carboxylate L-N-methylglutamic acid Nmglu Cyclohexylalanine Chexa L-N-methylhistidine Nmhis Cyclopentylalanine Cpen L-N-methylisolleucine Nmile D-alanine Dal L-N-methylleucine Nmleu D-arginine Darg L-N-methyllysine Nmlys D-aspartic acid Dasp L-N-methylmethionine Nmmet D-cysteine Dcys L-N-methylnorleucine Nmnle D-glutamine Dgln L-N-methylnorvaline Nmnva D-glutamic acid Dglu L-N-methylornithine Nmorn D-histidine Dhis L-N-methylphenylalanine Nmphe D-isoleucine Dile L-N-methylproline Nmpro D-leucine Dleu L-N-methylserine Nmser D-lysine Dlys L-N-methylthreonine Nmthr D-methionine Dmet L-N-methyltryptophan Nmtrp D-ornithine Dorn L-N-methyltyrosine Nmtyr D-phenylalanine Dphe L-N-methylvaline Nmval D-proline Dpro L-N-methylethylglycine Nmetg D-serine Dser L-N-methyl-t-butylglycine Nmtbug D-threonine Dthr L-norleucine Nle D-tryptophan Dtrp L-norvaline Nva D-tyrosine Dtyr α-methyl-aminoisobutyrate Maib D-valine Dval α-methyl-γ-aminobutyrate Mgabu D-α-methylalanine Dmala α-methylcyclohexylalanine Mchexa D-α-methylarginine Dmarg α-methylcyclopentylalanine Mcpen D-α-methylasparagine Dmasn α-methyl-α-napthylalanine Manap D-α-methylaspartate Dmasp α-methylpenicillamine Mpen D-α-methylcysteine Dmcys N-(4-aminobutyl)glycine Nglu D-α-methylglutamine Dmgln N-(2-aminoethyl)glycine Naeg D-α-methylhistidine Dmhis N-(3-aminopropyl)glycine Norn D-α-methylisoleucine Dmile N-amino-α-methylbutyrate Nmaabu D-α-methylleucine Dmleu α-napthylalanine Anap D-α-methyllysine Dmlys N-benzylglycine Nphe D-α-methylmethionine Dmmet N-(2-carbamylethyl)glycine Ngln D-α-methylornithine Dmorn N-(carbamylmethyl)glycine Nasn D-α-methylphenylalanine Dmphe N-(2-carboxyethyl)glycine Nglu D-α-methylproline Dmpro N-(carboxymethyl)glycine Nasp D-α-methylserine Dmser N-cyclobutylglycine Ncbut D-α-methylthreonine Dmthr N-cycloheptylglycine Nchep D-α-methyltryptophan Dmtrp N-cyclohexylglycine Nchex D-α-methyltyrosine Dmty N-cyclodecylglycine Ncdec D-α-methylvaline Dmval N-cyclododeclglycine Ncdod D-α-methylalnine Dnmala N-cyclooctylglycine Ncoct D-α-methylarginine Dnmarg N-cyclopropylglycine Ncpro D-α-methylasparagine Dnmasn N-cycloundecylglycine Ncund D-α-methylasparatate Dnmasp N-(2,2-diphenylethyl)glycine Nbhm D-α-methylcysteine Dnmcys N-(3,3-diphenylpropyl)glycine Nbhe D-N-methylleucine Dnmleu N-(3-indolylyethyl) glycine Nhtrp D-N-methyllysine Dnmlys N-methyl-γ-aminobutyrate Nmgabu N- Nmchexa D-N-methylmethionine Dnmmet methylcyclohexylalanine D-N-methylornithine Dnmom N-methylcyclopentylalanine Nmcpen N-methylglycine Nala D-N-methylphenylalanine Dnmphe N-methylaminoisobutyrate Nmaib D-N-methylproline Dnmpro N-(1-methylpropyl)glycine Nile D-N-methylserine Dnmser N-(2-methylpropyl)glycine Nile D-N-methylserine Dnmser N-(2-methylpropyl)glycine Nleu D-N-methylthreonine Dnmthr D-N-methyltryptophan Dnmtrp N-(1-methylethyl)glycine Nva D-N-methyltyrosine Dnmtyr N-methyla-napthylalanine Nmanap D-N-methylvaline Dnmval N-methylpenicillamine Nmpen γ-aminobutyric acid Gabu N-(p-hydroxyphenyl)glycine Nhtyr L-t-butylglycine Tbug N-(thiomethyl)glycine Ncys L-ethylglycine Etg Penicillamine Pen L-homophenylalanine Hphe L-α-methylalanine Mala L-α-methylarginine Marg L-α-methylasparagine Masn L-α-methylaspartate Masp L-α-methyl-t-butylglycine Mtbug L-α-methylcysteine Mcys L-methylethylglycine Metg L-α-methylglutamine Mgln L-α-methylglutamate Mglu L-α-methylhistidine Mhis L-α-methylhomo Mhphe phenylalanine L-α-methylisoleucine Mile N-(2-methylthioethyl)glycine Nmet D-N-methylglutamine Dnmgln N-(3-guanidinopropyl)glycine Narg D-N-methylglutamate Dnmglu N-(1-hydroxyethyl)glycine Nthr D-N-methylhistidine Dnmhis N-(hydroxyethyl)glycine Nser D-N-methylisoleucine Dnmile N-(imidazolylethyl)glycine Nhis D-N-methylleucine Dnmleu N-(3-indolylyethyl)glycine Nhtrp D-N-methyllysine Dnmlys N-methyl-γ-aminobutyrate Nmgabu N- Nmchexa D-N-methylmethionine Dnmmet methylcyclohexylalanine D-N-methylornithine Dnmorn N-methylcyclopentylalanine Nmcpen N-methylglycine Nala D-N-methylphenylalanine Dnmphe N-methylaminoisobutyrate Nmaib D-N-methylproline Dnmpro N-(1-methylpropyl)glycine Nile D-N-methylserine Dnmser N-(2-methylpropyl)glycine Nleu D-N-methylthreonine Dnmthr D-N-methyltryptophan Dnmtrp N-(1-methylethyl)glycine Nval D-N-methyltyrosine Dnmtyr N-methyla-napthylalanine Nmanap D-N-methylvaline Dnmval N-methylpenicillamine Nmpen γ-aminobutyric acid Gabu N-(p-hydroxyphenyl)glycine Nhtyr L-t-butylglycine Tbug N-(thiomethyl)glycine Ncys L-ethylglycine Etg Penicillamine Pen L-homophenylalanine Hphe L-α-methylalanine Mala L-α-methylarginine Marg L-α-methylasparagine Masn L-α-methylaspartate Masp L-α-methyl-t-butylglycine Mtbug L-α-methylcysteine Mcys L-methylethylglycine Metg L-α-methylglutamine Mgln L-α-methylglutamate Mglu L-α-methylhistidine Mhis L-α-methylhomophenylalanine Mhphe L-α-methylisoleucine Mile N-(2-methylthioethyl)glycine Nmet L-α-methylleucine Mleu L-α-methyllysine Mlys L-α-methylmethionine Mmet L-α-methylnorleucine Mnle L-α-methylnorvaline Mnva L-α-methylornithine Morn L-α-methylphenylalanine Mphe L-α-methylproline Mpro L-α-methylserine Mser L-α-methylthreonine Mthr L-α-methylvaline Mtrp L-α-methyltyrosine Mtyr L-α-methylleucine Mval L-N- Nmhphe Nnbbm methylhomophenylalanine N-(N-(2,2-diphenylethyl) N-(N-(3,3-diphenylpropyl) carbamylmethyl-glycine Nnbhm Carbamylmethyl(1)glycine Nnbhe 1-carboxy-1-(2,2-diphenyl Nmbc ethylamino)cyclopropane

Since the peptides of the present invention are preferably utilized in diagnostics which require the peptides to be in soluble form, the peptides of the present invention preferably include one or more non-natural or natural polar amino acids, including but not limited to serine and threonine which are capable of increasing peptide solubility due to their hydroxyl-containing side chain.

The peptides of the present invention are preferably utilized in a linear form, although it will be appreciated that in cases where cyclicization does not severely interfere with peptide characteristics, cyclic forms of the peptide can also be utilized.

The peptides of present invention can be biochemically synthesized such as by using standard solid phase techniques. These methods include exclusive solid phase synthesis well known in the art, partial solid phase synthesis methods, fragment condensation, classical solution synthesis. These methods are preferably used when the peptide is relatively short (i.e., 10 kDa) and/or when it cannot be produced by recombinant techniques (i.e., not encoded by a nucleic acid sequence) and therefore involves different chemistry.

Synthetic peptides can be purified by preparative high performance liquid chromatography and the composition of which can be confirmed via amino acid sequencing.

In cases where large amounts of the peptides of the present invention are desired, the peptides of the present invention can be generated using recombinant techniques such as described by Bitter et al., (1987) Methods in Enzymol. 153:516-544, Studier et al. (1990)

Methods in Enzymol. 185:60-89, Brisson et al. (1984) Nature 310:511-514, Takamatsu et al. (1987) EMBO J. 6:307-311, Coruzzi et al. (1984) EMBO J. 3:1671-1680 and Brogli et al., (1984) Science 224:838-843, Gurley et al. (1986) Mol. Cell. Biol. 6:559-565 and Weissbach & Weissbach, 1988, Methods for Plant Molecular Biology, Academic Press, NY, Section VIII, pp 421-463 and also as described above.

Antibodies

“Antibody” refers to a polypeptide ligand that is preferably substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof, which specifically binds and recognizes an epitope (e.g., an antigen). The recognized immunoglobulin genes include the kappa and lambda light chain constant region genes, the alpha, gamma, delta, epsilon and mu heavy chain constant region genes, and the myriad-immunoglobulin variable region genes. Antibodies exist, e.g., as intact immunoglobulins or as a number of well characterized fragments produced by digestion with various peptidases. This includes, e.g., Fab′ and F(ab)′2 fragments. The term “antibody,” as used herein, also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies. It also includes polyclonal antibodies, monoclonal antibodies, chimeric antibodies, humanized antibodies, or single chain antibodies. “Fc” portion of an antibody refers to that portion of an immunoglobulin heavy chain that comprises one or more heavy chain constant region domains, CH1, CH2 and CH3, but does not include the heavy chain variable region.

The functional fragments of antibodies, such as Fab, F(ab′)2, and Fv that are capable of binding to macrophages, are described as follows: (1) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule, can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain; (2) Fab′, the fragment of an antibody molecule that can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab′ fragments are obtained per antibody molecule; (3) (Fab′)2, the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab′)2 is a dimer of two Fab′ fragments held together by two disulfide bonds; (4) Fv, defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; and (5) Single chain antibody (“SCA”), a genetically engineered molecule containing the variable region of the light chain and the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.

Methods of producing polyclonal and monoclonal antibodies as well as fragments thereof are well known in the art (See for example, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1988, incorporated herein by reference).

Antibody fragments according to the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli or mammalian cells (e.g. Chinese hamster ovary cell culture or other protein expression systems) of DNA encoding the fragment. Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods. For example, antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab′)2. This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab′ monovalent fragments. Alternatively, an enzymatic cleavage using pepsin produces two monovalent Fab′ fragments and an Fc fragment directly. These methods are described, for example, by Goldenberg, U.S. Pat. Nos. 4,036,945 and 4,331,647, and references contained therein, which patents are hereby incorporated by reference in their entirety. See also Porter, R. R. [Biochem. J. 73: 119-126 (1959)]. Other methods of cleaving antibodies, such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody.

Fv fragments comprise an association of VH and VL chains. This association may be noncovalent, as described in Inbar et al. [Proc. Nat'l Acad. Sci. USA 69:2659-62 (19720]. Alternatively, the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde. Preferably, the Fv fragments comprise VH and VL chains connected by a peptide linker. These single-chain antigen binding proteins (sFv) are prepared by constructing a structural gene comprising DNA sequences encoding the VH and VL domains connected by an oligonucleotide. The structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli. The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains. Methods for producing sFvs are described, for example, by [Whitlow and Filpula, Methods 2: 97-105 (1991); Bird et al., Science 242:423-426 (1988); Pack et al., Bio/Technology 11:1271-77 (1993); and U.S. Pat. No. 4,946,778, which is hereby incorporated by reference in its entirety.

Another form of an antibody fragment is a peptide coding for a single complementarity-determining region (CDR). CDR peptides (“minimal recognition units”) can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells. See, for example, Larrick and Fry [Methods, 2: 106-10 (1991)].

Humanized forms of non-human (e.g., murine) antibodies are chimeric molecules of immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab′, F(ab′) or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)].

Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as import residues, which are typically taken from an import variable domain. Humanization can be essentially performed following the method of Winter and co-workers [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)], by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such humanized antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.

Human antibodies can also be produced using various techniques known in the art, including phage display libraries [Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581 (1991)]. The techniques of Cole et al. and Boerner et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985) and Boerner et al., J. Immunol., 147(1):86-95 (1991)]. Similarly, human antibodies can be made by introduction of human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in the following scientific publications: Marks et al., Bio/Technology 10: 779-783 (1992); Lonberg et al., Nature 368: 856-859 (1994); Morrison, Nature 368 812-13 (1994); Fishwild et al., Nature Biotechnology 14, 845-51 (1996); Neuberger, Nature Biotechnology 14: 826 (1996); and Lonberg and Huszar, Intern. Rev. Immunol. 13, 65-93 (1995).

Preferably, the antibody of this aspect of the present invention specifically binds at least one epitope of the polypeptide variants of the present invention. As used herein, the term “epitope” refers to any antigenic determinant on an antigen to which the paratope of an antibody binds.

Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or carbohydrate side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.

Optionally, a unique epitope may be created in a variant due to a change in one or more post-translational modifications, including but not limited to glycosylation and/or phosphorylation, as described below. Such a change may also cause a new epitope to be created, for example through removal of glycosylation at a particular site.

An epitope according to the present invention may also optionally comprise part or all of a unique sequence portion of a variant according to the present invention in combination with at least one other portion of the variant which is not contiguous to the unique sequence portion in the linear polypeptide itself, yet which are able to form an epitope in combination. One or more unique sequence portions may optionally combine with one or more other non-contiguous portions of the variant (including a portion which may have high homology to a portion of the known protein) to form an epitope.

Immunoassays

In another embodiment of the present invention, an immunoassay can be used to qualitatively or quantitatively detect and analyze markers in a sample. This method comprises: providing an antibody that specifically binds to a marker; contacting a sample with the antibody; and detecting the presence of a complex of the antibody bound to the marker in the sample.

To prepare an antibody that specifically binds to a marker, purified protein markers can be used. Antibodies that specifically bind to a protein marker can be prepared using any suitable methods known in the art.

After the antibody is provided, a marker can be detected and/or quantified using any of a number of well recognized immunological binding assays. Useful assays include, for example, an enzyme immune assay (EIA) such as enzyme-linked immunosorbent assay (ELISA), a radioimmune assay (RIA), a Western blot assay, or a slot blot assay see, e.g., U.S. Pat. Nos. 4,366,241; 4,376,110; 4,517,288; and 4,837,168). Generally, a sample obtained from a subject can be contacted with the antibody that specifically binds the marker.

Optionally, the antibody can be fixed to a solid support to facilitate washing and subsequent isolation of the complex, prior to contacting the antibody with a sample. Examples of solid supports include but are not limited to glass or plastic in the form of, e.g., a microtiter plate, a stick, a bead, or a microbead. Antibodies can also be attached to a solid support.

After incubating the sample with antibodies, the mixture is washed and the antibody-marker complex formed can be detected. This can be accomplished by incubating the washed mixture with a detection reagent. Alternatively, the marker in the sample can be detected using an indirect assay, wherein, for example, a second, labeled antibody is used to detect bound marker-specific antibody, and/or in a competition or inhibition assay wherein, for example, a monoclonal antibody which binds to a distinct epitope of the marker are incubated simultaneously with the mixture.

Throughout the assays, incubation and/or washing steps may be required after each combination of reagents. Incubation steps can vary from about 5 seconds to several hours, preferably from about 5 minutes to about 24 hours. However, the incubation time will depend upon the assay format, marker, volume of solution, concentrations and the like. Usually the assays will be carried out at ambient temperature, although they can be conducted over a range of temperatures, such as 10° C. to 40° C.

The immunoassay can be used to determine a test amount of a marker in a sample from a subject. First, a test amount of a marker in a sample can be detected using the immunoassay methods described above. If a marker is present in the sample, it will form an antibody-marker complex with an antibody that specifically binds the marker under suitable incubation conditions described above. The amount of an antibody-marker complex can optionally be determined by comparing to a standard. As noted above, the test amount of marker need not be measured in absolute units, as long as the unit of measurement can be compared to a control amount and/or signal.

Preferably used are antibodies which specifically interact with the polypeptides of the present invention and not with wild type proteins or other isoforms thereof, for example. Such antibodies are directed, for example, to the unique sequence portions of the polypeptide variants of the present invention, including but not limited to bridges, heads, tails and insertions described in greater detail below. Preferred embodiments of antibodies according to the present invention are described in greater detail with regard to the section entitled “Antibodies”.

Radio-immunoassay (RJA): In one version, this method involves precipitation of the desired substrate and in the methods detailed hereinbelow, with a specific antibody and radiolabelled antibody binding protein (e.g., protein A labeled with I125) immobilized on a precipitable carrier such as agarose beads. The number of counts in the precipitated pellet is proportional to the amount of substrate.

In an alternate version of the RIA, a labeled substrate and an unlabelled antibody binding protein are employed. A sample containing an unknown amount of substrate is added in varying amounts. The decrease in precipitated counts from the labeled substrate is proportional to the amount of substrate in the added sample.

Enzyme linked immunosorbent assay (ELISA): This method involves fixation of a sample (e.g., fixed cells or a proteinaceous solution) containing a protein substrate to a surface such as a well of a microtiter plate. A substrate specific antibody coupled to an enzyme is applied and allowed to bind to the substrate. Presence of the antibody is then detected and quantitated by a colorimetric reaction employing the enzyme coupled to the antibody. Enzymes commonly employed in this method include horseradish peroxidase and alkaline phosphatase. If well calibrated and within the linear range of response, the amount of substrate present in the sample is proportional to the amount of color produced. A substrate standard is generally employed to improve quantitative accuracy.

Western blot: This method involves separation of a substrate from other protein by means of an acrylamide gel followed by transfer of the substrate to a membrane (e.g., nylon or PVDF). Presence of the substrate is then detected by antibodies specific to the substrate, which are in turn detected by antibody binding reagents. Antibody binding reagents may be, for example, protein A, or other antibodies. Antibody binding reagents may be radiolabelled or enzyme linked as described hereinabove. Detection may be by autoradiography, colorimetric reaction or chemiluminescence. This method allows both quantitation of an amount of substrate and determination of its identity by a relative position on the membrane which is indicative of a migration distance in the acrylamide gel during electrophoresis.

Immunohistochemical analysis: This method involves detection of a substrate in situ in fixed cells by substrate specific antibodies. The substrate specific antibodies may be enzyme linked or linked to fluorophores. Detection is by microscopy and subjective evaluation. If enzyme linked antibodies are employed, a colorimetric reaction may be required.

Fluorescence activated cell sorting (FACS): This method involves detection of a substrate in situ in cells by substrate specific antibodies. The substrate specific antibodies are linked to fluorophores. Detection is by means of a cell sorting machine which reads the wavelength of light emitted from each cell as it passes through a light beam. This method may employ two or more antibodies simultaneously.

Radio-Imaging Methods

These methods include but are not limited to, positron emission tomography (PET) single photon emission computed tomography (SPECT). Both of these techniques are non-invasive, and can be used to detect and/or measure a wide variety of tissue events and/or functions, such as detecting cancerous cells for example. Unlike PET, SPECT can optionally be used with two labels simultaneously. SPECT has some other advantages as well, for example with regard to cost and the types of labels that can be used. For example, U.S. Pat. No. 6,696,686 describes the use of SPECT for detection of breast cancer, and is hereby incorporated by reference as if fully set forth herein.

Display Libraries

According to still another aspect of the present invention there is provided a display library comprising a plurality of display vehicles (such as phages, viruses or bacteria) each displaying at least 6, at least 7, at least 8, at least 9, at least 10, 10-15, 12-17, 15-20, 15-30 or 20-50 consecutive amino acids derived from the polypeptide sequences of the present invention.

Methods of constructing such display libraries are well known in the art. Such methods are described in, for example, Young A C, et al., “The three-dimensional structures of a polysaccharide binding antibody to Cryptococcus neoformans and its complex with a peptide from a phage display library: implications for the identification of peptide mimotopes” J Mol Biol 1997 Dec. 12; 274(4):622-34; Giebel L B et al. “Screening of cyclic peptide phage libraries identifies ligands that bind streptavidin with high affinities” Biochemistry 1995 Nov. 28; 34(47):15430-5; Davies E L et al., “Selection of specific phage-display antibodies using libraries derived from chicken immunoglobulin genes” J Immunol Methods 1995 Oct. 12; 186(1):125-35; Jones C R T al. “Current trends in molecular recognition and bioseparation” J Chromatogr A 1995 Jul. 14; 707(1):3-22; Deng S J et al. “Basis for selection of improved carbohydrate-binding single-chain antibodies from synthetic gene libraries” Proc Natl Acad Sci USA 1995 May 23; 92(11):4992-6; and Deng S J et al. “Selection of antibody single-chain variable fragments with improved carbohydrate binding by phage display” J Biol Chem 1994 Apr. 1; 269(13):9533-8, which are incorporated herein by reference.

The following sections relate to Candidate Marker Examples.

Candidate Marker Examples Section

This Section relates to Examples of sequences according to the present invention, including illustrative methods of selection thereof.

Description of the Methodology Undertaken to Uncover the Biomolecular Sequences of the Present Invention

Human ESTs and cDNAs were obtained from GenBank versions 136 (Jun. 15, 2003 ftp.ncbi.nih.gov/genbank/release.notes/gb136.release.notes); NCBI genome assembly of April 2003; RefSeq sequences from June 2003; Genbank version 139 (December 2003); Human Genome from NCBI (Build 34) (from October 2003); and RefSeq sequences from December 2003; and the LifeSeq library from Incyte Corporation (Wilmington, Del., USA; ESTs only). With regard to GenBank sequences, the human EST sequences from the EST (GBEST) section and the human mRNA sequences from the primate (GBPRI) section were used; also the human nucleotide RefSeq mRNA sequences were used (see for example www “dot” ncbi “dot” nlm “dot” nih “dot” gov/Genbank/GenbankOverview “dot” html and for a reference to the EST section, see www “dot” ncbi “dot” nlm “dot” nih “dot” gov/dbEST/; a general reference to dbEST, the EST database in GenBank, may be found in Boguski et al, Nat. Genet. 1993 August; 4(4):332-3; all of which are hereby incorporated by reference as if fully set forth herein).

Novel splice variants were predicted using the LEADS clustering and assembly system as described in Sorek, R., Ast, G. & Graur, D. Alu-containing exons are alternatively spliced. Genome Res 12, 1060-7 (2002); U.S. Pat. No. 6,625,545; and U.S. patent application Ser. No. 10/426,002, published as US20040101876 on May 27, 2004; all of which are hereby incorporated by reference as if fully set forth herein. Briefly, the software cleans the expressed sequences from repeats, vectors and immunoglobulins. It then aligns the expressed sequences to the genome taking alternatively splicing into account and clusters overlapping expressed sequences into “clusters” that represent genes or partial genes.

These were annotated using the GeneCarta (Compugen, Tel-Aviv, Israel) platform. The GeneCarta platform includes a rich pool of annotations, sequence information (particularly of spliced sequences), chromosomal information, alignments, and additional information such as SNPs, gene ontology terms, expression profiles, functional analyses, detailed domain structures, known and predicted proteins and detailed homology reports.

A brief explanation is provided with regard to the method of selecting the candidates. However, it should noted that this explanation is provided for descriptive purposes only, and is not intended to be limiting in any way. The potential markers were identified by a computational process that was designed to find genes and/or their splice variants that are over-expressed in tumor tissues, by using databases of expressed sequences. Various parameters related to the information in the EST libraries, determined according to a manual classification process, were used to assist in locating genes and/or splice variants thereof that are over-expressed in cancerous tissues. The detailed description of the selection method is presented in Example 1 below. The cancer biomarkers selection engine and the following wet validation stages are schematically summarized in FIG. 1.

Example 1 Identification of Differentially Expressed Gene Products Algorithm

In order to distinguish between differentially expressed gene products and constitutively expressed genes (i.e., house keeping genes) an algorithm based on an analysis of frequencies was configured. A specific algorithm for identification of transcripts over expressed in cancer is described hereinbelow.

Dry Analysis

Library Annotation—Est Libraries are Manually Classified According to:

(i) Tissue origin

(ii) Biological source—Examples of frequently used biological sources for construction of EST libraries include cancer cell-lines; normal tissues; cancer tissues; fetal tissues; and others such as normal cell lines and pools of normal cell-lines, cancer cell-lines and combinations thereof. A specific description of abbreviations used below with regard to these tissues/cell lines etc is given above.

(iii) Protocol of library construction—various methods are known in the art for library construction including normalized library construction; non-normalized library construction; subtracted libraries; ORESTES and others. It will be appreciated that at times the protocol of library construction is not indicated in GenBank and/or other library annotaion.

The following rules are followed:

EST libraries originating from identical biological samples are considered as a single library.

EST libraries which included above-average levels of contamination, such as DNA contamination for example, were eliminated. The presence of such contamination was determined as follows. For each library, the number of unspliced ESTs that are not fully contained within other spliced sequences was counted. If the percentage of such sequences (as compared to all other sequences) was at least 4 standard deviations above the average for all libraries being analyzed, this library was tagged as being contaminated and was eliminated from further consideration in the below analysis (see also Sorek, R. & Safer, H. M. A novel algorithm for computational identification of contaminated EST libraries. Nucleic Acids Res 31, 1067-74 (2003) for further details).

Clusters (genes) having at least five sequences including at least two sequences from the tissue of interest were analyzed. Splice variants were identified by using the LEADS software package as described above.

Example 2 Identification of Genes Over Expressed in Cancer

Two different scoring algorithms were developed.

Libraries score—candidate sequences which are supported by a number of cancer libraries, are more likely to serve as specific and effective diagnostic markers.

The basic algorithm—for each cluster the number of cancer and normal libraries contributing sequences to the cluster was counted. Fisher exact test was used to check if cancer libraries are significantly over-represented in the cluster as compared to the total number of cancer and normal libraries.

Library counting: Small libraries (e.g., less than 1000 sequences) were excluded from consideration unless they participate in the cluster. For this reason, the total number of libraries is actually adjusted for each cluster.

Clones no. score—Generally, when the number of ESTs is much higher in the cancer libraries relative to the normal libraries it might indicate actual over-expression.

The algorithm—

Clone counting: For counting EST clones each library protocol class was given a weight based on our belief of how much the protocol reflects actual expression levels:

(i) non-normalized: 1

(ii) normalized: 0.2

(iii) all other classes: 0.1

Clones number score—The total weighted number of EST clones from cancer libraries was compared to the EST clones from normal libraries. To avoid cases where one library contributes to the majority of the score, the contribution of the library that gives most clones for a given cluster was limited to 2 clones.

The score was computed as c + 1 C / n + 1 N

Where:

c—weighted number of “cancer” clones in the cluster.

C—weighted number of clones in all “cancer” libraries.

n—weighted number of “normal” clones in the cluster.

N—weighted number of clones in all “normal” libraries.

Clones number score significance—Fisher exact test was used to check if EST clones from cancer libraries are significantly over-represented in the cluster as compared to the total number of EST clones from cancer and normal libraries.

Two search approaches were used to find either general cancer-specific candidates or tumor specific candidates.

    • Libraries/sequences originating from tumor tissues are counted as well as libraries originating from cancer cell-lines (“normal” cell-lines were ignored).
    • Only libraries/sequences originating from tumor tissues are counted

Example 3 Identification of Tissue Specific Genes

For detection of tissue specific clusters, tissue libraries/sequences were compared to the total number of libraries/sequences in cluster. Similar statistical tools to those described in above were employed to identify tissue specific genes. Tissue abbreviations are the same as for cancerous tissues, but are indicated with the header “normal tissue”.

The algorithm—for each tested tissue T and for each tested cluster the following were examined:

1. Each cluster includes at least 2 libraries from the tissue T. At least 3 clones (weighed—as described above) from tissue T in the cluster; and

2. Clones from the tissue T are at least 40% from all the clones participating in the tested cluster

Fisher exact test P-values were computed both for library and weighted clone counts to check that the counts are statistically significant.

Example 4 Identification of Splice Variants Over Expressed in Cancer of Clusters which are not Over Expressed in Cancer

Cancer-Specific Splice Variants Containing a Unique Region were Identified.

Identification of unique sequence regions in splice variants

A Region is defined as a group of adjacent exons that always appear or do not appear together in each splice variant.

A “segment” (sometimes referred also as “seg” or “node”) is defined as the shortest contiguous transcribed region without known splicing inside.

Only reliable ESTs were considered for region and segment analysis. An EST was defined as unreliable if:

(i) Unspliced;

(ii) Not covered by RNA;

(iii) Not covered by spliced ESTs; and

(iv) Alignment to the genome ends in proximity of long poly-A stretch or starts in proximity of long poly-T stretch.

Only reliable regions were selected for further scoring. Unique sequence regions were considered reliable if:

(i) Aligned to the genome; and

(ii) Regions supported by more than 2 ESTs.

The algorithm

Each unique sequence region divides the set of transcripts into 2 groups:

(i) Transcripts containing this region (group TA).

(ii) Transcripts not containing this region (group TB).

The set of EST clones of every cluster is divided into 3 groups:

(i) Supporting (originating from) transcripts of group TA (S1).

(ii) Supporting transcripts of group TB (S2).

(iii) Supporting transcripts from both groups (S3).

Library and clones number scores described above were given to S1 group.

Fisher Exact Test P-values were used to check if:

S1 is significantly enriched by cancer EST clones compared to S2; and

S1 is significantly enriched by cancer EST clones compared to cluster background (S1+S2+S3).

Identification of unique sequence regions and division of the group of transcripts accordingly is illustrated in FIG. 2. Each of these unique sequence regions corresponds to a segment, also termed herein a “node”.

Region 1: common to all transcripts, thus it is not considered; Region 2: specific to Transcript 1: T1 unique regions (2+6) against T2+3 unique regions (3+4); Region 3: specific to Transcripts 2+3: T2+3 unique regions (3+4) against Ti unique regions (2+6); Region 4: specific to Transcript 3: T3 unique regions (4) against Tl+2 unique regions (2+5+6); Region 5: specific to Transcript 1+2: T1+2 unique regions (2+5+6) against T3 unique regions (4); Region 6: specific to Transcript 1: same as region 2.

Example 5

Identification of cancer specific splice variants of genes over expressed in cancer

A search for EST supported (no mRNA) regions for genes of:

(i) known cancer markers

(ii) Genes shown to be over-expressed in cancer in published micro-array experiments.

Reliable EST supported-regions were defined as supported by minimum of one of the following:

(i) 3 spliced ESTs; or

(ii) 2 spliced ESTs from 2 libraries;

(iii) 10 unspliced ESTs from 2 libraries, or

(iv) 3 libraries.

Actual Marker Examples

The following examples relate to specific actual marker examples.

EXPERIMENTAL EXAMPLES SECTION

This Section relates to Examples describing experiments involving these sequences, and illustrative, non-limiting examples of methods, assays and uses thereof. The materials and experimental procedures are explained first, as all experiments used them as a basis for the work that was performed.

The markers of the present invention were tested with regard to their expression in various cancerous and non-cancerous tissue samples. A description of the samples used in the panel is provided in Table 2 below. A description of the samples used in the normal tissue panel is provided in Table 3 below. Tests were then performed as described in the “Materials and Experimental Procedures” section below.

TABLE 2 Tissue samples in testing panel Lot No. Pathology Sex/Age Source 66-A-Adeno G1 GS-4 160202 Adenocarcinoma Gleason score 4 M/64 ABS 73-A-Adeno G1 GS-4 16026T2 Acinar Adenocarcinoma Gleason score 4(2 + 2) M/77 ABS 68-A-Adeno G1 GS-5 160172 Adenocarcinoma Gleason score 5 M/66 ABS 56-Am-Adeno G1 GS-5 36467 Adenocarcinoma, Gleason score 5(3 + 2); stage 2 M/72 Ambion 58-Am-Adeno G1 GS-5 37192 Adenocarcinoma, Gleason score 5; stage 2 M/52 Ambion 65-A-Adeno G2 GS-5 160022 Adenocarcinoma Gleason score 5 M/66 ABS 69-A-Adeno GS-5 160182 Acinar Adenocarcinoma Gleason score 5 M/58 ABS 55-Am-Adeno GS-5 36464 Adenocarcinoma, Gleason score 5; stage 1 M/53 Ambion 64-A-Adeno G2 GS-6 160092 Acinar Adenocarcinoma Gleason score 6 M/71 ABS 70-A-Adeno G2 GS-6 160192 Adenocarcinoma Gleason score 6 M/53 ABS 18-A-Adeno GS-6 5610020069T Adenocarcinoma, Gleason score 6 (3 + 3) M ABS 67-A-Adeno GS-6 160142 Acinar Adenocarcinoma Gleason score 6 M/62 ABS 25-A-Adeno GS-7 5605020052T Adenocarcinoma, Gleason score 7 (4 + 3) M ABS 26-A-Adeno GS-7 5609020067T Adenocarcinoma, Gleason score 7 (4 + 3) M ABS 72-A-Adeno GS-7 160122 Acinar Adenocarcinoma Gleason score 7 M/66 ABS 71-A-Adeno GS-7 160242 Acinar Adenocarcinoma Gleason score 7 M/70 ABS 57-Am-Adeno GS-7 26442 Adenocarcinoma, Gleason score 7 M/62 Ambion 32-A-Adeno GS-9 5604020042T Adenocarcinoma, Gleason score 9 (5 + 4) M ABS 54-B-Adeno G3 A610031 Adenocarcinoma Biochair 33-A-BPH 5607020058 BPH M ABS 34-A-BPH 5607020059 BPH M ABS 35-A-BPH 5607020060 BPH M ABS 43-B-PBH A609267 BPH M/66 Biochair 44-B-PBH A609268 BPH M/72 Biochair 45-B-PBH A609269 BPH M/69 Biochair 46-B-PBH A609270 BPH M/65 Biochair 47-B-PBH A609271 BPH M/71 Biochair 40-A-N M26 5609020067N Normal Matched M ABS 41-A-N M32 5604020042N Normal Matched M ABS 48-B-N A609257 Normal PM M/24 Biochair 49-B-N A609256 Normal PM M/36 Biochair 50-B-N A609255 Normal PM M/26 Biochair 51-B-N A609258 Normal PM M/27 Biochair 52-B-N A609254 Normal PM M/29 Biochair 53-Cl-N 1070317 Normal - Pool of 47 M&F Clontech 42-Am-N 061P04A Normal (IC BLEED) M/47 ambion 59-Am-N 25955 Normal PM (Head trauma) M/62 Ambion 60-Am-N 33605 Normal PM (Myocardial infraction) M/69 Ambion 61-Am-N 34077 Normal PM (Alzheimer's) M/71 Ambion 62-Am-N 31316 Normal (Renal failure) M/79 Ambion 63-Am-N 30991 Normal (Gall Bladder cancer) M/78 Ambion

TABLE 3 Tissue samples in normal panel: Lot no. Source Tissue Pathology Sex/Age 1-Am-Colon (C71) 071P10B Ambion Colon PM F/43 2-B-Colon (C69) A411078 Biochain Colon PM-Pool of 10 M&F 3-Cl-Colon (C70) 1110101 Clontech Colon PM-Pool of 3 M&F 4-Am-Small Intestine 091P0201A Ambion Small Intestine PM M/75 5-B-Small Intestine A501158 Biochain Small Intestine PM M/63 6-B-Rectum A605138 Biochain Rectum PM M/25 7-B-Rectum A610297 Biochain Rectum PM M/24 8-B-Rectum A610298 Biochain Rectum PM M/27 9-Am-Stomach 110P04A Ambion Stomach PM M/16 10-B-Stomach A501159 Biochain Stomach PM M/24 11-B-Esophagus A603814 Biochain Esophagus PM M/26 12-B-Esophagus A603813 Biochain Esophagus PM M/41 13-Am-Pancreas 071P25C Ambion Pancreas PM M/25 14-CG-Pancreas CG-255-2 Ichilov Pancreas PM M/75 15-B-Lung A409363 Biochain Lung PM F/26 16-Am-Lung (L93) 111P0103A Ambion Lung PM F/61 17-B-Lung (L92) A503204 Biochain Lung PM M/28 18-Am-Ovary (O47) 061P43A Ambion Ovary PM F/16 19-B-Ovary (O48) A504087 Biochain Ovary PM F/51 20-B-Ovary (O46) A504086 Biochain Ovary PM F/41 21-Am-Cervix 101P0101A Ambion Cervix PM F/40 22-B-Cervix A408211 Biochain Cervix PM F/36 23-B-Cervix A504089 Biochain Cervix PM-Pool of 5 M&F 24-B-Uterus A411074 Biochain Uterus PM-Pool of 10 M&F 25-B-Uterus A409248 Biochain Uterus PM F/43 26-B-Uterus A504090 Biochain Uterus PM-Pool of 5 M&F 27-B-Bladder A501157 Biochain Bladder PM M/29 28-Am-Bladder 071P02C Ambion Bladder PM M/20 29-B-Bladder A504088 Biochain Bladder PM-Pool of 5 M&F 30-Am-Placenta 021P33A Ambion Placenta PB F/33 31-B-Placenta A410165 Biochain Placenta PB F/26 32-B-Placenta A411073 Biochain Placenta PB-Pool of 5 M&F 33-B-Breast (B59) A607155 Biochain Breast PM F/36 34-Am-Breast (B63) 26486 Ambion Breast PM F/43 35-Am-Breast (B64) 23036 Ambion Breast PM F/57 36-Cl-Prostate (P53) 1070317 Clontech Prostate PB-Pool of 47 M&F 37-Am-Prostate (P42) 061P04A Ambion Prostate PM M/47 38-Am-Prostate (P59) 25955 Ambion Prostate PM M/62 39-Am-Testis 111P0104A Ambion Testis PM M/25 40-B-Testis A411147 Biochain Testis PM M/74 41-Cl-Testis 1110320 Clontech Testis PB-Pool of 45 M&F 42-CG-Adrenal CG-184-10 Ichilov Adrenal PM F/81 43-B-Adrenal A610374 Biochain Adrenal PM F/83 44-B-Heart A411077 Biochain Heart PB-Pool of 5 M&F 45-CG-Heart CG-255-9 Ichilov Heart PM M/75 46-CG-Heart CG-227-1 Ichilov Heart PM F/36 47-Am-Liver 081P0101A Ambion Liver PM M/64 48-CG-Liver CG-93-3 Ichilov Liver PM F/19 49-CG-Liver CG-124-4 Ichilov Liver PM F/34 50-Cl-BM 1110932 Clontech Bone Marrow PM-Pool of 8 M&F 51-CGEN-Blood WBC#5 CGEN Blood M 52-CGEN-Blood WBC#4 CGEN Blood M 53-CGEN-Blood WBC#3 CGEN Blood M 54-CG-Spleen CG-267 Ichilov Spleen PM F/25 55-CG-Spleen 111P0106B Ambion Spleen PM M/25 56-CG-Spleen A409246 Biochain Spleen PM F/12 56-CG-Thymus CG-98-7 Ichilov Thymus PM F/28 58-Am-Thymus 101P0101A Ambion Thymus PM M/14 59-B-Thymus A409278 Biochain Thymus PM M/28 60-B-Thyroid A610287 Biochain Thyroid PM M/27 61-B-Thyroid A610286 Biochain Thyroid PM M/24 62-CG-Thyroid CG-119-2 Ichilov Thyroid PM F/66 63-Cl-Salivary Gland 1070319 Clontech Salivary Gland PM-Pool of 24 M&F 64-Am-Kidney 111P0101B Ambion Kidney PM-Pool of 14 M&F 65-Cl-Kidney 1110970 Clontech Kidney PM-Pool of 14 M&F 66-B-Kidney A411080 Biochain Kidney PM-Pool of 5 M&F 67-CG-Cerebellum CG-183-5 Ichilov Cerebellum PM M/74 68-CG-Cerebellum CG-212-5 Ichilov Cerebellum PM M/54 69-B-Brain A411322 Biochain Brain PM M/28 70-Cl-Brain 1120022 Clontech Brain PM-Pool of 2 M&F 71-B-Brain A411079 Biochain Brain PM-Pool of 2 M&F 72-CG-Brain CG-151-1 Ichilov Brain PM F/86 73-Am-Skeletal Muscle 101P013A Ambion Skeletal Muscle PM F/28 74-Cl-Skeletal Muscle 1061038 Clontech Skeletal Muscle PM-Pool of 2 M&F

Materials and Experimental Procedures

RNA preparation —RNA was obtained from Clontech (Franklin Lakes, N.J. USA 07417, www“dot” clontech“dot” com), BioChain Inst. Inc. (Hayward, Calif. 94545 USA www.biochain.com), ABS (Wilmington, Del. 19801, USA, www“dot” absbioreagents“dot” com) or Ambion (Austin, Tex. 78744 USA, www“dot” ambion“dot” com). Alternatively, RNA was generated from tissue samples using TR1—Reagent (Molecular Research Center), according to Manufacturer's instructions. Tissue and RNA samples were obtained from patients or from postmortem. Total RNA samples were treated with DNaseI (Ambion) and purified using RNeasy columns (Qiagen).

RT PCR—Purified RNA (1 μg) was mixed with 150 ng Random Hexamer primers (Invitrogen) and 500 μM dNTP in a total volume of 15.6 μl. The mixture was incubated for 5 min at 65° C. and then quickly chilled on ice. Thereafter, 5 μl of 5× SuperscriptII first strand buffer (Invitrogen), 2.4 μl 0.1M DTT and 40 units RNasin (Promega) were added, and the mixture was incubated for 10 min at 25° C., followed by further incubation at 42° C. for 2 min. Then, 1 μl (200 units) of SuperscriptII (Invitrogen) was added and the reaction (final volume of 25 μl) was incubated for 50 min at 42° C. and then inactivated at 70° C. for 15 min. The resulting cDNA was diluted 1:20 in TE buffer (10 mM Tris pH=8, 1 mM EDTA pH=8).

Real-Time RT-PCR analysis—cDNA (5 μl), prepared as described above, was used as a template in Real-Time PCR reactions using the SYBR Green I assay (PE Applied Biosystem) with specific primers and UNG Enzyme (Eurogentech or ABI or Roche). The amplification was

effected as follows: 50° C. for 2 min, 95° C. for 10 min, and then 40 cycles of 95° C. for 15 sec, followed by 60° C. for 1 min. Detection was performed by using the PE Applied Biosystem SDS 7000. The cycle in which the reactions achieved a threshold level (Ct) of fluorescence was registered and was used to calculate the relative transcript quantity in the RT reactions. The relative quantity was calculated using the equation Q=efficiencyˆ−Ct. The efficiency of the PCR reaction was calculated from a standard curve, created by using serial dilutions of several reverse transcription (RT) reactions. To minimize inherent differences in the RT reaction, the resulting relative quantities were normalized to the geometric mean of the relative quantities of several housekeeping (HSKP) genes. Schematic summary of quantitative real-time PCR analysis is presented in FIG. 3. As shown, the x-axis shows the cycle number. The CT=Threshold Cycle point, which is the cycle that the amplification curve crosses the fluorescence threshold that was set in the experiment. This point is a calculated cycle number in which PCR product signal is above the background level (passive dye ROX) and still in the Geometric/Exponential phase (as shown, once the level of fluorescence crosses the measurement threshold, it has a geometrically increasing phase, during which measurements are most accurate, followed by a linear phase and a plateau phase; for quantitative measurements, the latter two phases do not provide accurate measurements). The y-axis shows the normalized reporter fluorescence. It should be noted that this type of analysis provides relative quantification.

The sequences of the housekeeping genes measured in all the examples below on prostate panel were as follows:

SDHA (GenBank Accession No. NM_004168 (SEQ ID NO: 508)) SDHA Forward primer (SEQ ID NO: 405): TGGGAACAAGAGGGCATCTG SDHA Reverse primer (SEQ ID NO: 406): CCACCACTGCATCAAATTCATG SDHA-amplicon (SEQ ID NO: 407): TGGGAACAAGAGGGCATCTGCTAAAGTTTCAGATTCCATTTCTGCTCAGT ATCCAGTAGTGGATCATGAATTTGATGCAGTGGTGG PBGD (GenBank Accession No. BC019323 (SEQ ID NO: 509)), PBGD Forward primer (SEQ ID NO: 402): TGAGAGTGATTCGCGTGGG PBGD Reverse primer (SEQ ID NO: 403): CCAGGGTACGAGGCTTTCAAT PBGD-amplicon (SEQ ID NO: 404): TGAGAGTGATTCGCGTGGGTACCCGCAAGAGCCAGCTTGCTCGCATACAG ACGGACAGTGTGGTGGCAACATTGAAAGCCTCGTACCCTGG HPRT1 (GenBank Accession No. NM_000194 (SEQ ID NO: 510)), HPRT1 Forward primer (SEQ ID NO: 399): TGACACTGGCAAAACAATGCA HPRT1 Reverse primer (SEQ ID NO: 400): GGTCCTTTTCACCAGCAAGCT HPRT1-amplicon (SEQ ID NO: 401): TGACACTGGCAAAACAATGCAGACTTTGCTTTCCTTGGTCAGGCAGTATA ATCCAAAGATGGTCAAGGTCGCAAGCTTGCTGGTGAAAAGGACC RPL19 (GenBank Accession No. NM_000981 (SEQ ID NO: 511) RPL19Forward primer (SEQ ID NO: 408): TGGCAAGAAGAAGGTCTGGTTAG RPL19Reverse primer (SEQ ID NO: 409): TGATCAGCCCATCTTTGATGAG RPL19-amplicon (SEQ ID NO: 410): TGGCAAGAAGAAGGTCTGGTTAGACCCCAATGAGACCAATGAAATCGCCA ATGCCAACTCCCGTCAGCAGATCCGGAAGCTCATCAAAGATGGGCTGATC A

The sequences of the housekeeping genes measured in all the examples on normal tissue samples panel were as follows:

RPL19 (GenBank Accession No. NM_000981 (SEQ ID NO: 511)), RPL19 Forward primer (SEQ ID NO: 408): TGGCAAGAAGAAGGTCTGGTTAG RPL19 Reverse primer (SEQ ID NO: 409): TGATCAGCCCATCTTTGATGAG RPL19 -amplicon (SEQ ID NO: 410): TGGCAAGAAGAAGGTCTGGTTAGACCCCAATGAGACCAATGAAATCGCCA ATGCCAACTCCCGTCAGCAGATCCGGAAGCTCATCAAAGATGGGCTGATC A TATA box (GenBank Accession No. NM_003194 (SEQ ID NO: 512)), TATA box Forward primer (SEQ ID NO: 513): CGGTTTGCTGCGGTAATCAT TATA box Reverse primer (SEQ ID NO: 514): TTTCTTGCTGCCAGTCTGGAC TATA box -amplicon (SEQ ID NO: 515): CGGTTTGCTGCGGTAATCATGAGGATAAGAGAGCCACGAACCACGGCACT GATTTTCAGTTCTGGGAAAATGGTGTGCACAGGAGCCAAGAGTGAAGAAC AGTCCAGACTGGCAGCAAGAAA UBC (GenBank Accession No. BC000449 (SEQ ID NO: 516)) UBC Forward primer (SEQ ID NO: 517): ATTTGGGTCGCGGTTCTTG UBC Reverse primer (SEQ ID NO: 518): TGCCTTGACATTCTCGATGGT UBC -amplicon (SEQ ID NO: 519): ATTTGGGTCGCGGTTCTTGTTTGTGGATCGCTGTGATCGTCACTTGACAA TGCAGATCTTCGTGAAGACTCTGACTGGTAAGACCATCACCCTCGAGG TTGAGCCCAGTGACACCATCGAGAATGTCAAGGCA SDHA (GenBank Accession No. NM_004168 (SEQ ID NO: 508)) SDHA Forward primer (SEQ ID NO: 405): TGGGAACAAGAGGGCATCTG SDHA Reverse primer (SEQ ID NO: 406): CCACCACTGCATCAAATTCATG SDHA-amplicon (SEQ ID NO: 407): TGGGAACAAGAGGGCATCTGCTAAAGTTTCAGATTCCATTTCTGCTCAGT ATCCAGTAGTGGATCATGAATTTGATGCAGTGGTGG

Oligonucleotide-Based Micro-Array Experiment Protocol—

Microarray Fabrication

Microarrays (chips) were printed by pin deposition using the MicroGrid II MGII 600 robot from BioRobtics Limited (Cambridge, UK). 50-mer oligonucleotides target sequences were designed by Compugen Ltd (Tel-Aviv, Ill.) as described by A. Shoshan et al, “Optical technologies and informatics”, Proceedings of SPIE. Vol 4266, pp. 86-95 (2001). The designed oligonucleotides were synthesized and purified by desalting with the Sigma-Genosys system (The Woodlands, Tex., US) and all of the oligonucleotides were joined to a C6 amino-modified linker at the 5′ end, or being attached directly to CodeLink slides (Cat #25-6700-01. Amersham Bioscience, Piscataway, N.J., US). The 50-mer oligonucleotides, forming the target sequences, were first suspended in Ultra-pure DDW (Cat # 01-866-1A Kibbutz Beit-Haemek, Israel) to a concentration of 50 μM. Before printing the slides, the oligonucleotides were resuspended in 300 mM sodium phosphate (pH 8.5) to final concentration of 150 mM and printed at 35-40% relative humidity at 21° C.

Each slide contained a total of 9792 features in 32 subarrays. Of these features, 4224 features were sequences of interest according to the present invention and negative controls that were printed in duplicate. An additional 288 features (96 target sequences printed in triplicate) contained housekeeping genes from Human Evaluation Library2, Compugen Ltd, Israel. Another 384 features are E. coli spikes 1-6, which are oligos to E-Coli genes which are commercially available in the Array Control product (Array control-sense oligo spots, Ambion Inc. Austin, Tex. Cat #1781, Lot #112K06).

Post-Coupling Processing of Printed Slides

After the spotting of the oligonucleotides to the glass (CodeLink) slides, the slides were incubated for 24 hours in a sealed saturated NaCl humidification chamber (relative humidity 70-75%).

Slides were treated for blocking of the residual reactive groups by incubating them in blocking solution at 50° C. for 15 minutes (10 ml/slide of buffer containing 0.1M Tris, 50 mM ethanolamine, 0.1% SDS). The slides were then rinsed twice with Ultra-pure DDW (double distilled water). The slides were then washed with wash solution (10 ml/slide. 4×SSC, 0.1% SDS)) at 50° C. for 30 minutes on the shaker. The slides were then rinsed twice with Ultra-pure DDW, followed by drying by centrifugation for 3 minutes at 800 rpm.

Next, in order to assist in automatic operation of the hybridization protocol, the slides were treated with Ventana Discovery hybridization station barcode adhesives. The printed slides were loaded on a Bio-Optica (Milan, Italy) hematology staining device and were incubated for 10 minutes in 50 ml of 3-Aminopropyl Triethoxysilane (Sigma A3648 lot #122K589). Excess fluid was dried and slides were then incubated for three hours in 20 mm/Hg in a dark vacuum desiccator (Pelco 2251, Ted Pella, Inc. Redding Calif.).

The following protocol was then followed with the Genisphere 900-RP (random primer), with mini elute columns on the Ventana Discovery HybStation™, to perform the microarray experiments. Briefly, the protocol was performed as described with regard to the instructions and information provided with the device itself. The protocol included cDNA synthesis and labeling. cDNA concentration was measured with the TBS-380 (Turner Biosystems. Sunnyvale, Calif.) PicoFlour, which is used with the OliGreen ssDNA Quantitation reagent and kit.

Hybridization was performed with the Ventana Hybridization device, according to the provided protocols (Discovery Hybridization Station Tuscon Ariz.).

The slides were then scanned with GenePix 4000B dual laser scanner from Axon Instruments Inc, and analyzed by GenePix Pro 5.0 software.

Schematic summary of the oligonucleotide based microarray fabrication and the experimental flow is presented in FIGS. 4 and 5.

Briefly, as shown in FIG. 4, DNA oligonucleotides at 25 uM were deposited (printed) onto Amersham ‘CodeLink’ glass slides generating a well defined ‘spot’. These slides are covered with a long-chain, hydrophilic polymer chemistry that creates an active 3-D surface that covalently binds the DNA oligonucleotides 5′-end via the C6-amine modification. This binding ensures that the full length of the DNA oligonucleotides is available for hybridization to the cDNA and also allows lower background, high sensitivity and reproducibility.

FIG. 5 shows a schematic method for performing the microarray experiments. It should be noted that stages on the left-hand or right-hand side may optionally be performed in any order, including in parallel, until stage 4 (hybridization). Briefly, on the left-hand side, the target oligonucleotides are being spotted on a glass microscope slide (although optionally other materials could be used) to form a spotted slide (stage 1). On the right hand side, control sample RNA and cancer sample RNA are Cy3 and Cy5 labeled, respectively (stage 2), to form labeled probes. It should be noted that the control and cancer samples come from corresponding tissues (for example, normal prostate tissue and cancerous prostate tissue). Furthermore, the tissue from which the RNA was taken is indicated below in the specific examples of data for particular clusters, with regard to overexpression of an oligonucleotide from a “chip” (microarray), as for example “prostate” for chips in which prostate cancerous tissue and normal tissue were tested as described above. In stage 3, the probes are mixed. In stage 4, hybridization is performed to form a processed slide. In stage 5, the slide is washed and scanned to form an image file, followed by data analysis in stage 6.

Description for Cluster R11723

Cluster R11723 features 6 transcript(s) and 26 segment(s) of interest, the names for which are given in Tables 4 and 5, respectively, the sequences themselves are given at the end of the application. The selected protein variants are given in table 6.

TABLE 4 Transcripts of interest Transcript Name Sequence ID No. R11723_PEA_1_T15 5 R11723_PEA_1_T17 6 R11723_PEA_1_T19 7 R11723_PEA_1_T20 8 R11723_PEA_1_T5 9 R11723_PEA_1_T6 10

TABLE 5 Segments of interest Segment Name Sequence ID No. R11723_PEA_1_node_13 90 R11723_PEA_1_node_16 91 R11723_PEA_1_node_19 92 R11723_PEA_1_node_2 93 R11723_PEA_1_node_22 94 R11723_PEA_1_node_31 95 R11723_PEA_1_node_10 96 R11723_PEA_1_node_11 97 R11723_PEA_1_node_15 98 R11723_PEA_1_node_18 99 R11723_PEA_1_node_20 100 R11723_PEA_1_node_21 101 R11723_PEA_1_node_23 102 R11723_PEA_1_node_24 103 R11723_PEA_1_node_25 104 R11723_PEA_1_node_26 105 R11723_PEA_1_node_27 106 R11723_PEA_1_node_28 107 R11723_PEA_1_node_29 108 R11723_PEA_1_node_3 109 R11723_PEA_1_node_30 110 R11723_PEA_1_node_4 111 R11723_PEA_1_node_5 112 R11723_PEA_1_node_6 113 R11723_PEA_1_node_7 114 R11723_PEA_1_node_8 115

TABLE 6 Proteins of interest Protein Name Sequence ID No. R11723_PEA_1_P2 331 R11723_PEA_1_6 332 R11723_PEA_1_P7 333 R11723_PEA_1_P13 334 R11723_PEA_1_P10 335

Cluster R11723 can be used as a diagnostic marker according to overexpression of transcripts of this cluster in cancer. Expression of such transcripts in normal tissues is also given according to the previously described methods. The term “number” in the right hand column of the table and the numbers on the y-axis of FIG. 6 refer to weighted expression of ESTs in each category, as “parts per million” (ratio of the expression of ESTs for a particular cluster to the expression of all ESTs in that category, according to parts per million).

Overall, the following results were obtained as shown with regard to the histograms in FIG. 6 and Table 7. This cluster is overexpressed (at least at a minimum level) in the following pathological conditions: epithelial malignant tumors, a mixture of malignant tumors from different tissues and kidney malignant tumors.

TABLE 7 Normal tissue distribution Name of Tissue Number Adrenal 0 Brain 30 Epithelial 3 General 17 head and neck 0 Kidney 0 Lung 0 Breast 0 Ovary 0 Pancreas 10 Skin 0 Uterus 0

TABLE 8 P values and ratios for expression in cancerous tissue Name of Tissue P1 P2 SP1 R3 SP2 R4 Adrenal 4.2e−01 4.6e−01 4.6e−01 2.2 5.3e−01 1.9 Brain 2.2e−01 2.0e−01 1.2e−02 2.8 5.0e−02 2.0 Epithelial 3.0e−05 6.3e−05 1.8e−05 6.3 3.4e−06 6.4 General 7.2e−03 4.0e−02 1.3e−04 2.1 1.1e−03 1.7 head and neck 1 5.0e−01 1 1.0 7.5e−01 1.3 Kidney 1.5e−01 2.4e−01 4.4e−03 5.4 2.8e−02 3.6 Lung 1.2e−01 1.6e−01 1 1.6 1 1.3 Breast 5.9e−01 4.4e−01 1 1.1 6.8e−01 1.5 Ovary 1.6e−02 1.3e−02 1.0e−01 3.8 7.0e−02 3.5 Pancreas 5.5e−01 2.0e−01 3.9e−01 1.9 1.4e−01 2.7 Skin 1 4.4e−01 1 1.0 1.9e−02 2.1 Uterus 1.5e−02 5.4e−02 1.9e−01 3.1 1.4e−01 2.5

As noted above, cluster R11723 features 6 transcript(s), which were listed in Table 4 above. A description of each variant protein according to the present invention is now provided.

Variant protein R11723_PEA1_P2 (SEQ ID NO:331) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) R11723_PEA1_T6 (SEQ ID NO:10). The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein R11723_PEA1_P2 (SEQ ID NO:331) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 9, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P2 (SEQ ID NO:331) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 9 Amino acid mutations SNP position(s) on amino acid Alternative sequence amino acid(s) Previously known SNP? 107 H -> P Yes 70 G -> No 70 G -> C No

Variant protein R11723_PEA1_P2 (SEQ ID NO:331) is encoded by the following transcript(s): R11723_PEA1_T6 (SEQ ID NO:10), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript R11723_PEA1_T6 (SEQ ID NO:10) is shown in bold; this coding portion starts at position 1716 and ends at position 2051. The transcript also has the following SNPs as listed in Table 10 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P2 (SEQ ID NO:331) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 10 Nucleic acid SNPs SNP position on nucleotide Previously sequence Alternative nucleic acid known SNP? 1231 C -> T Yes 1278 G -> C Yes 1923 G -> No 1923 G -> T No 2035 A -> C Yes 2048 A -> C No 2057 A -> G Yes

Variant protein R11723_PEA1_P6 (SEQ ID NO:332) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) R11723_PEA1_T15 (SEQ ID NO:5). One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between R11723_PEA1_P6 (SEQ ID NO:332) and Q8IXM0 (SEQ ID NO:485):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P6 (SEQ ID NO:332), comprising a first amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAGIMYRKSCASSAACLIASAGSPCRGLAPGREEQRALHKAGAVGGGVR (SEQ ID NO: 534) corresponding to amino acids 1-110 of R11723_PEA1_P6 (SEQ ID NO:332), and a second amino acid sequence being at least 90% homologous to MYAQALLVVGVLQRQAAAQHLHEHPPKLLRGHRVQERVDDRAEVEKRLREGEEDHV RPEVGPRPVVLGFGRSHDPPNLVGHPAYGQCHNNQPWADTSRRERQRKEKHSMRTQ corresponding to amino acids 1-112 of Q8IXM0, which also corresponds to amino acids 111-222 of R11723_PEA1_P6 (SEQ ID NO:332), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a head of R11723_PEA1_P6 (SEQ ID NO:332), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence

(SEQ ID NO: 534) MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV QDMCQKEVMEQSAGIMYRKSCASSAACLIASAGSPCRGLAPGREEQRALH KAGAVGGGVR of (SEQ ID NO: 332) R11723_PEA_1_P6.

Comparison report between R11723_PEA1_P6 (SEQ ID NO:332) and Q96AC2 (SEQ ID NO:486):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P6 (SEQ ID NO:332), comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAGIMYRKSCASSAACLIASAG corresponding to amino acids 1-83 of Q96AC2, which also corresponds to amino acids 1-83 of R11723_PEA1_P6 (SEQ ID NO:332), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SPCRGLAPGREEQRALHKAGAVGGGVRMYAQALLVVGVLQRQAAAQHLHEHPPKLL RGHRVQERVDDRAEVEKRLREGEEDHVRPEVGPRPVVLGFGRSHDPPNLVGHPAYGQ CHNNQPWADTSRRERQRKEKHSMRTQ (SEQ ID NO: 535) corresponding to amino acids 84-222 of R11723_PEA1_P6 (SEQ ID NO:332), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P6 (SEQ ID NO:332) comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence

(SEQ ID NO: 535) SPCRGLAPGREEQRALHKAGAVGGGVRMYAQALLVVGVLQRQAAAQHLHE HPPKLLRGHRVQERVDDRAEVEKRLREGEEDHVRPEVGPRPVVLGFGRSH DPPNLVGHPAYGQCHNNQPWADTSRRERQRKEKHSMRTQ (SEQ ID NO: 332) in R11723_PEA_1_P6.

Comparison report between R11723_PEA1_P6 (SEQ ID NO:332) and Q8N2G4 (SEQ ID NO:487):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P6 (SEQ ID NO:332), comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAGIMYRKSCASSAACLIASAG corresponding to amino acids 1-83 of Q8N2G4, which also corresponds to amino acids 1-83 of R11723_PEA1_P6 (SEQ ID.NO:332), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SPCRGLAPGREEQRALHKAGAVGGGVRMYAQALLVVGVLQRQAAAQHLHEHPPKLL RGHRVQERVDDRAEVEKRLREGEEDHVRPEVGPRPVVLGFGRSHDPPNLVGHPAYGQ CHNNQPWADTSRRERQRKEKHSMRTQ (SEQ ID NO: 535) corresponding to amino acids 84-222 of R11723_PEA1_P6 (SEQ ID NO:332), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P6 (SEQ ID NO:332) comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence

(SEQ ID NO: 535) SPCRGLAPGREEQRALHKAGAVGGGVRMYAQALLVVGVLQRQAAAQHLHE HPPKLLRGHRVQERVDDRAEVEKRLREGEEDHVRPEVGPRPVVLGFGRSH DPPNLVGHPAYGQCHNNQPWADTSRRERQRKEKHSMRTQ (SEQ ID NO: 332) in R11723_PEA_1_P6

Comparison report between R11723_PEA1_P6 (SEQ ID NO:332) and BAC85518 (SEQ ID NO:488):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P6 (SEQ ID NO:332), comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAGIMYRKSCASSAACLIASAG corresponding to amino acids 24-106 of BAC85518, which also corresponds to amino acids 1-83 of R11723_PEA1_P6 (SEQ ID NO:332), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SPCRGLAPGREEQRALHKAGAVGGGVRMYAQALLVVGVLQRQAAAQHLHEHPPKLL RGHRVQERVDDRAEVEKRLREGEEDHVRPEVGPRPVVLGFGRSHDPPNLVGHPAYGQ CHNNQPWADTSRRERQRKEKHSMRTQ (SEQ ID NO: 535) corresponding to amino acids 84-222 of R11723_PEA1_P6 (SEQ ID NO:332), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P6 (SEQ ID NO:332), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence

(SEQ ID NO: 535) SPCRGLAPGREEQRALHKAGAVGGGVRMYAQALLVVGVLQRQAAAQHLHE HPPKLLRGHRVQERVDDRAEVEKRLREGEEDHVRPEVGPRPVVLGFGRSH DPPNLVGHPAYGQCHNNQPWADTSRRERQRKEKHSMRTQ (SEQ ID NO: 332) in R11723_PEA_1_P6.

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein R11723_PEA1_P6 (SEQ ID NO:332) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 11, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P6 (SEQ ID NO:332) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 11 Amino acid mutations SNP position(s) on amino acid Alternative sequence amino acid(s) Previously known SNP? 180 G -> No 180 G -> C No 217 H -> P Yes

Variant protein R11723_PEA1_P6 (SEQ ID NO:332) is encoded by the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript R11723_PEA1_T15 (SEQ ID NO:5) is shown in bold; this coding portion starts at position 434 and ends at position 1099. The transcript also has the following SNPs as listed in Table 12 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P6 (SEQ ID NO:332) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 12 Nucleic acid SNPs SNP position on nucleotide Previously sequence Alternative nucleic acid known SNP? 971 G -> No 971 G -> T No 1083 A -> C Yes 1096 A -> C No 1105 A -> G Yes

Variant protein R11723_PEA_l_P7 (SEQ ID NO:333) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) R11723_PEA1_T17 (SEQ ID NO:6). One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between R11723_PEA1_P7 (SEQ ID NO:333) and Q96AC2 (SEQ ID NO:486):

1. An isolated chimeric polypeptide encoding for R11723_PEA1P7 (SEQ ID NO:333), comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAG corresponding to amino acids 1-64 of Q96AC2 (SEQ ID NO:486), which also corresponds to amino acids 1-64 of R11723_PEA1_P7 (SEQ ID NO:333), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) corresponding to amino acids 65-93 of R11723_PEA1_P7 (SEQ ID NO:333), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P7 (SEQ ID NO:333), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) in R11723_PEA1_P7 (SEQ ID NO:333).

Comparison report between R11723_PEA1_P7 (SEQ ID NO:333) and Q8N2G4 (SEQ ID NO:487):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P7 (SEQ ID NO:333), comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAG corresponding to amino acids 1-64 of Q8N2G4 (SEQ ID NO:487), which also corresponds to amino acids 1-64 of R11723_PEA1P7 (SEQ ID NO:333), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) corresponding to amino acids 65-93 of R11723_PEA1_P7 (SEQ ID NO:333), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P7 (SEQ ID NO:333), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) in R11723_PEA1_P7 (SEQ ID NO:333).

Comparison report between R11723_PEA1_P7 (SEQ ID NO:333) and BAC85273 (SEQ ID NO:489):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P7 (SEQ ID NO:333), comprising a first amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence MWVLG (SEQ ID NO: 537) corresponding to amino acids 1-5 of R11723_PEA1_P7 (SEQ ID NO:333), second amino acid sequence being at least 90% homologous to IAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEVMEQSAG corresponding to amino acids 22-80 of BAC85273, which also corresponds to amino acids 6-64 of R11723_PEA1_P7 (SEQ ID NO:333), and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) corresponding to amino acids 65-93 of R11723_PEA1_P7 (SEQ ID NO:333), wherein said first, second and third amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a head of R11723_PEA1_P7 (SEQ ID NO:333), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence MWVLG (SEQ ID NO: 537) of R 1723_PEA1_P7 (SEQ ID NO:333).

3. An isolated polypeptide encoding for a tail of R11723_PEA1P7 (SEQ ID NO:333), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) in R11723_PEA1_P7 (SEQ ID NO:333).

Comparison report between R11723_PEA1_P7 (SEQ ID NO:333) and BAC85518 (SEQ ID NO:489):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P7 (SEQ ID NO:333), comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSAG corresponding to amino acids 24-87 of BAC85518, which also corresponds to amino acids 1-64 of R11723_PEA1_P7 (SEQ ID NO:333), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) corresponding to amino acids 65-93 of R11723_PEA1_P7 (SEQ ID NO:333), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P7 (SEQ ID NO:333), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence SHCVTRLECSGTISAHCNLCLPGSNDHPT (SEQ ID NO: 536) in R11723_PEA1_P7 (SEQ ID NO:333).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein R11723_PEA1_P7 (SEQ ID NO:333) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 13, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P7 (SEQ ID NO:333) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 13 Amino acid mutations SNP position(s) on amino acid Alternative sequence amino acid(s) Previously known SNP? 67 C -> S Yes

Variant protein R11723_PEA1_P7 (SEQ ID NO:333) is encoded by the following transcript(s): R11723_PEA1_T17 (SEQ ID NO:6), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript R11723_PEA1_T17 (SEQ ID NO:6) is shown in bold; this coding portion starts at position 434 and ends at position 712. The transcript also has the following SNPs as listed in Table 14 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P7 (SEQ ID NO:333) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 14 Nucleic acid SNPs SNP position on nucleotide Alternative sequence nucleic acid Previously known SNP? 625 G -> T Yes 633 G -> C Yes 1303 C -> T Yes

Variant protein R11723_PEA1_P13 (SEQ ID NO:334) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) R11723_PEA1_T19 (SEQ ID NO:7) and R11723_PEA1_T5 (SEQ ID NO:9). One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between R11723_PEA1_P13 (SEQ ID NO:334) and Q96AC2:

1. An isolated chimeric polypeptide encoding for R11723_PEA_-1P13 (SEQ ID NO:334) comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSA corresponding to amino acids 1-63 of Q96AC2, which also corresponds to amino acids 1-63 of R11723_PEA1_P13 (SEQ ID NO:334), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence DTKRTNTLLFEMRHFAKQLTT (SEQ ID NO: 538) corresponding to amino acids 64-84 of R11723_PEA1_P13 (SEQ ID NO:334), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P13 (SEQ ID NO:334), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence DTKRTNTLLFEMRHFAKQLTT (SEQ ID NO: 538) in R11723_PEA1_P13 (SEQ ID NO:334).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein R11723_PEA1_P13 (SEQ ID NO:334) is encoded by the following transcript(s): R11723_PEA1_T19 (SEQ ID NO:7), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript R11723_PEA1_T19 (SEQ ID NO:7) is shown in bold; this coding portion starts at position 434 and ends at position 685. The transcript also has the following SNPs as listed in Table 15 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P13 (SEQ ID NO:334) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 15 Nucleic acid SNPs SNP position on nucleotide Alternative sequence nucleic acid Previously known SNP? 778 G -> T Yes 786 G -> C Yes 1456 C -> T Yes

Variant protein R11723_PEA1_P10 (SEQ ID NO:335) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) R11723_PEA1_T20 (SEQ ID NO:8). One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between R11723_PEA1_P10 (SEQ ID NO:335) and Q96AC2:

1. An isolated chimeric polypeptide encoding for R11723_PEA1P10 (SEQ ID NO:335), comprising a first amino acid sequence being at least 90% homologous to MWVLGLAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSA corresponding to amino acids 1-63 of Q96AC2, which also corresponds to amino acids 1-63 of R11723_PEA_lP10 (SEQ ID NO:335), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) corresponding to amino acids 64-90 of R11723_PEA1_P10 (SEQ ID NO:335), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P10 (SEQ ID NO:335), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) in R11723_PEA1_P10 (SEQ ID NO:335).

Comparison report between R11723_PEA1_P10 (SEQ ID NO:335) and Q8N2G4 (SEQ ID NO:487):

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P10 (SEQ ID NO:335) comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSA corresponding to amino acids 1-63 of Q8N2G4, which also corresponds to amino acids 1-63 of R11723_PEA1P10 (SEQ ID NO:335), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) corresponding to amino acids 64-90 of R11723_PEA1_P10 (SEQ ID NO:335), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P10 (SEQ ID NO:335), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) in R11723_PEA1_P10 (SEQ ID NO:335).

Comparison report between R11723_PEA1_P10 (SEQ ID NO:335) and BAC85273:

1. An isolated chimeric polypeptide encoding for R11723_PEA1_P10 (SEQ ID NO:335), comprising a first amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence MWVLG (SEQ ID NO: 537) corresponding to amino acids 1-5 of R11723_PEA1 P10 (SEQ ID NO:335), second amino acid sequence being at least 90% homologous to IAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEVMEQSA corresponding to amino acids 22-79 of BAC85273, which also corresponds to amino acids 6-63 of R11723_PEA1P10 (SEQ ID NO:335), and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) corresponding to amino acids 64-90 of R11723_PEA1_P10 (SEQ ID NO:335), wherein said first, second and third amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a head of R11723_PEA1_P10 (SEQ ID NO:335), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence MWVLG (SEQ ID NO: 537) of R11723_PEA1_P10 (SEQ ID NO:335).

3. An isolated polypeptide encoding for a tail of R11723_PEA1_P10 (SEQ ID NO:335), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) in R11723_PEA1_P10 (SEQ ID NO:335).

Comparison report between R11723_PEA1_P10 (SEQ ID NO:335) and BAC85518:

1. An isolated chimeric polypeptide encoding for R11723_PEA1P10 (SEQ ID NO:335) comprising a first amino acid sequence being at least 90% homologous to MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEV MEQSA corresponding to amino acids 24-86 of BAC85518, which also corresponds to amino acids 1-63 of R11723_PEA1P10 (SEQ ID NO:335), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) corresponding to amino acids 64-90 of R11723_PEA1_P10 (SEQ ID NO:335), wherein said first and second amino acid sequences are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of R11723_PEA1_P10 (SEQ ID NO:335), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence DRVSLCHEAGVQWNNFSTLQPLPPRLK (SEQ ID NO: 539) in R11723_PEA1_P10 (SEQ ID NO:335).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein R11723_PEA1_P10 (SEQ ID NO:335) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 16, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P10 (SEQ ID NO:335) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 16 Amino acid mutations SNP position(s) on amino acid Alternative sequence amino acid(s) Previously known SNP? 66 V -> F Yes

Variant protein R11723_PEA1_P10 (SEQ ID NO:335) is encoded by the following transcript(s): R11723_PEA1_T20 (SEQ ID NO:8), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript R11723_PEA1_T20 (SEQ ID NO:8) is shown in bold; this coding portion starts at position 434 and ends at position 703. The transcript also has the following SNPs as listed in Table 17 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein R11723_PEA1_P10 (SEQ ID NO:335) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 17 Nucleic acid SNPs SNP position on nucleotide Alternative sequence nucleic acid Previously known SNP? 629 G -> T Yes 637 G -> C Yes 1307 C -> T Yes

As noted above, cluster R11723 features 26 segment(s), which were listed in Table 49 above and for which the sequence(s) are given at the end of the application. These segment(s) are portions of nucleic acid sequence(s) which are described herein separately because they are of particular interest. A description of each segment according to the present invention is now provided.

Segment cluster R11723_PEA1_node13 (SEQ ID NO:90) according to the present invention is supported by 5 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1IT19 (SEQ ID NO:7), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 18 below describes the starting and ending position of this segment on each transcript.

TABLE 18 Segment location on transcripts Segment Segment Transcript name starting position ending position R11723_PEA_1_T19 (SEQ 624 776 ID NO: 7) R11723_PEA_1_T5 (SEQ ID 624 776 NO: 9) R11723_PEA_1_T6 (SEQ ID 658 810 NO: 10)

Segment cluster R11723_PEA1_node16 (SEQ ID NO:91) according to the present invention is supported by 3 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1IT17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7) and R11723_PEA1_T20 (SEQ ID NO:8). Table 19 below describes the starting and ending position of this segment on each transcript.

TABLE 19 Segment location on transcripts Segment Segment Transcript name starting position ending position R11723_PEA_1_T17 (SEQ 624 1367 ID NO: 6) R11723_PEA_1_T19 (SEQ 777 1520 ID NO: 7) R11723_PEA_1_T20 (SEQ 628 1371 ID NO: 8)

Segment cluster R11723_PEA1_node19 (SEQ ID NO:92) according to the present invention is supported by 45 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 20 below describes the starting and ending position of this segment on each transcript.

TABLE 20 Segment location on transcripts Segment Segment Transcript name starting position ending position R11723_PEA_1_T5 (SEQ ID 835 1008 NO: 9) R11723_PEA_1_T6 (SEQ ID 869 1042 NO: 10)

Segment cluster R11723_PEA1_node2 (SEQ ID NO:93) according to the present invention is supported by 29 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7) R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 21 below describes the starting and ending position of this segment on each transcript.

TABLE 21 Segment location on transcripts Segment Segment Transcript name starting position ending position R11723_PEA_1_T15 (SEQ 1 309 ID NO: 5) R11723_PEA_1_T17 (SEQ 1 309 ID NO: 6) R11723_PEA_1_T19 (SEQ 1 309 ID NO: 7) R11723_PEA_1_T20 (SEQ 1 309 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 1 309 NO: 9) R11723_PEA_1_T6 (SEQ ID 1 309 NO: 10)

Segment cluster R11723_PEA1_node22 (SEQ ID NO:94) according to the present invention is supported by 65 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO: 10). Table 22 below describes the starting and ending position of this segment on each transcript.

TABLE 22 Segment location on transcripts Segment Segment Transcript name starting position ending position R11723_PEA_1_T5 (SEQ ID 1083 1569 NO: 9) R11723_PEA_1_T6 (SEQ ID 1117 1603 NO: 10)

Segment cluster R11723_PEA1_node31 (SEQ ID NO:95) according to the present invention is supported by 70 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1IT6 (SEQ ID NO:10). Table 23 below describes the starting and ending position of this segment on each transcript (it should be noted that these transcripts show alternative polyadenylation).

TABLE 23 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 1060 1295 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1978 2213 NO: 9) R11723_PEA_1_T6 (SEQ ID 2012 2247 NO: 10)

According to an optional embodiment of the present invention, short segments related to the above cluster are also provided. These segments are up to about 120 bp in length, and so are included in a separate description.

Segment cluster R11723_PEA1_node10 (SEQ ID NO:96) according to the present invention is supported by 38 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7), R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and

R11723PEA1_T6 (SEQ ID NO:10). Table 24 below describes the starting and ending position of this segment on each transcript.

TABLE 24 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 486 529 ID NO: 5) R11723_PEA_1_T17 (SEQ 486 529 ID NO: 6) R11723_PEA_1_T19 (SEQ 486 529 ID NO: 7) R11723_PEA_1_T20 (SEQ 486 529 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 486 529 NO: 9) R11723_PEA_1_T6 (SEQ ID 520 563 NO: 10)

Segment cluster R11723_PEA1_node11 (SEQ ID NO:97) according to the present invention is supported by 42 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7), R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 25 below describes the starting and ending position of this segment on each transcript.

TABLE 25 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 530 623 ID NO: 5) R11723_PEA_1_T17 (SEQ 530 623 ID NO: 6) R11723_PEA_1_T19 (SEQ 530 623 ID NO: 7) R11723_PEA_1_T20 (SEQ 530 623 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 530 623 NO: 9) R11723_PEA_1_T6 (SEQ ID 564 657 NO: 10)

Segment cluster R11723_PEA1_node15 (SEQ ID NO:98) according to the present invention can be found in the following transcript(s): R11723_PEA1_T20 (SEQ ID NO:8). Table 26 below describes the starting and ending position of this segment on each transcript.

TABLE 26 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T20 (SEQ 624 627 ID NO: 8)

Segment cluster R11723_PEA1_node18 (SEQ ID NO:99) according to the present invention is supported by 40 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723-PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 27 below describes the starting and ending position of this segment on each transcript.

TABLE 27 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 624 681 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 777 834 NO: 9) R11723_PEA_1_T6 (SEQ ID 811 868 NO: 10)

Segment cluster R11723_PEA1_node20 (SEQ ID NO:100) according to the present invention can be found in the following transcript(s): R11723_PEA1_T5 (SEQ ID NO:9) and

R11723_PEA1T6 (SEQ ID NO:10). Table 28 below describes the starting and ending position of this segment on each transcript.

TABLE 28 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T5 (SEQ ID 1009 1019 NO: 9) R11723_PEA_1_T6 (SEQ ID 1043 1053 NO: 10)

Segment cluster R11723_PEA1_node21 (SEQ ID NO:101) according to the present invention is supported by 36 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 29 below describes the starting and ending position of this segment on each transcript.

TABLE 29 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T5 (SEQ ID 1020 1082 NO: 9) R11723_PEA_1_T6 (SEQ ID 1054 1116 NO: 10)

Segment cluster R11723_PEA1_node23 (SEQ ID NO: 102) according to the present invention is supported by 39 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1T6 (SEQ ID NO:10). Table 30 below describes the starting and ending position of this segment on each transcript.

TABLE 30 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T5 (SEQ ID 1570 1599 NO: 9) R11723_PEA_1_T6 (SEQ ID 1604 1633 NO: 10)

Segment cluster R11723_PEA1_node24 (SEQ ID NO:103) according to the present invention is supported by 51 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1IT6 (SEQ ID NO:10). Table 31 below describes the starting and ending position of this segment on each transcript.

TABLE 31 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 682 765 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1600 1683 NO: 9) R11723_PEA_1_T6 (SEQ ID 1634 1717 NO: 10)

Segment cluster R11723_PEA1_node25 (SEQ ID NO: 104) according to the present invention is supported by 54 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 32 below describes the starting and ending position of this segment on each transcript.

TABLE 32 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 766 791 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1684 1709 NO: 9) R11723_PEA_1_T6 (SEQ ID 1718 1743 NO: 10)

Segment cluster R11723_PEA1_node26 (SEQ ID NO:105) according to the present invention is supported by 62 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 33 below describes the starting and ending position of this segment on each transcript.

TABLE 33 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 792 904 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1710 1822 NO: 9) R11723_PEA_1_T6 (SEQ ID 1744 1856 NO: 10)

Segment cluster R11723_PEA1_node 27 (SEQ ID NO: 106) according to the present invention is supported by 67 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1IT6 (SEQ ID NO:10) Table 34 below describes the starting and ending position of this segment on each transcript.

TABLE 34 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 905 986 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1823 1904 NO: 9) R11723_PEA_1_T6 (SEQ ID 1857 1938 NO: 10)

Segment cluster R11723_PEA1_node28 (SEQ ID NO:107) according to the present invention can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1IT6 (SEQ ID NO:10). Table 35 below describes the starting and ending position of this segment on each transcript.

TABLE 35 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 987 1010 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1905 1928 NO: 9) R11723_PEA_1_T6 (SEQ ID 1939 1962 NO: 10)

Segment cluster R11723_PEA1_node29 (SEQ ID NO:108) according to the present invention is supported by 69 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723-PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 36 below describes the starting and ending position of this segment on each transcript.

TABLE 36 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 1011 1038 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1929 1956 NO: 9) R11723_PEA_1_T6 (SEQ ID 1963 1990 NO: 10)

Segment cluster R11723_PEA1_node3 (SEQ ID NO:109) according to the present invention can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7), R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 37 below describes the starting and ending position of this segment on each transcript.

TABLE 37 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 310 319 ID NO: 5) R11723_PEA_1_T17 (SEQ 310 319 ID NO: 6) R11723_PEA_1_T19 (SEQ 310 319 ID NO: 7) R11723_PEA_1_T20 (SEQ 310 319 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 310 319 NO: 9) R11723_PEA_1_T6 (SEQ ID 310 319 NO: 10)

Segment cluster R11723_PEA1_node30 (SEQ ID NO: 110) according to the present invention can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1T6 (SEQ ID NO:10). Table 38 below describes the starting and ending position of this segment on each transcript.

TABLE 38 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 1039 1059 ID NO: 5) R11723_PEA_1_T5 (SEQ ID 1957 1977 NO: 9) R11723_PEA_1_T6 (SEQ ID 1991 2011 NO: 10)

Segment cluster R11723_PEA1_node4 (SEQ ID NO:111) according to the present invention is supported by 25 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7) R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 39 below describes the starting and ending position of this segment on each transcript.

TABLE 39 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 320 371 ID NO: 5) R11723_PEA_1_T17 (SEQ 320 371 ID NO: 6) R11723_PEA_1_T19 (SEQ 320 371 ID NO: 7) R11723_PEA_1_T20 (SEQ 320 371 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 320 371 NO: 9) R11723_PEA_1_T6 (SEQ ID 320 371 NO: 10)

Segment cluster R11723_PEA1_node5 (SEQ ID NO:112) according to the present invention is supported by 26 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1T19 (SEQ ID NO:7), R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723PEA1T6 (SEQ ID NO:10). Table 40 below describes the starting and ending position of this segment on each transcript.

TABLE 40 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 372 414 ID NO: 5) R11723_PEA_1_T17 (SEQ 372 414 ID NO: 6) R11723_PEA_1_T19 (SEQ 372 414 ID NO: 7) R11723_PEA_1_T20 (SEQ 372 414 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 372 414 NO: 9) R11723_PEA_1_T6 (SEQ ID 372 414 NO: 10)

Segment cluster R11723_PEA1_node6 (SEQ ID NO:113) according to the present invention is supported by 27 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7), R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 41 below describes the starting and ending position of this segment on each transcript.

TABLE 41 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 415 446 ID NO: 5) R11723_PEA_1_T17 (SEQ 415 446 ID NO: 6) R11723_PEA_1_T19 (SEQ 415 446 ID NO: 7) R11723_PEA_1_T20 (SEQ 415 446 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 415 446 NO: 9) R11723_PEA_1_T6 (SEQ ID 415 446 NO: 10)

Segment cluster R11723_PEA1_node7 (SEQ ID NO:114) according to the present invention is supported by 29 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1_T15 (SEQ ID NO:5), R11723_PEA1_T17 (SEQ ID NO:6), R11723_PEA1_T19 (SEQ ID NO:7), R11723_PEA1_T20 (SEQ ID NO:8), R11723_PEA1_T5 (SEQ ID NO:9) and R11723_PEA1_T6 (SEQ ID NO:10). Table 42 below describes the starting and ending position of this segment on each transcript.

TABLE 42 Segment location on transcripts Segment starting Segment ending Transcript name position position R11723_PEA_1_T15 (SEQ 447 485 ID NO: 5) R11723_PEA_1_T17 (SEQ 447 485 ID NO: 6) R11723_PEA_1_T19 (SEQ 447 485 ID NO: 7) R11723_PEA_1_T20 (SEQ 447 485 ID NO: 8) R11723_PEA_1_T5 (SEQ ID 447 485 NO: 9) R11723_PEA_1_T6 (SEQ ID 447 485 NO: 10)

Segment cluster R11723_PEA1_node8 (SEQ ID NO:115) according to the present invention is supported by 2 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): R11723_PEA1T6 (SEQ ID NO: 10). Table 43 below describes the starting and ending position of this segment on each transcript.

TABLE 43 Segment location on transcripts Segment Segment ending Transcript name starting position position R11723_PEA_1_T6 (SEQ ID 486 519 NO: 10)

It should be noted that the variants of this cluster are variants of the hypothetical protein PSEC0181 (referred to herein as “PSEC”). Furthermore, use of the known protein (WT protein) for detection of ovarian cancer, alone or in combination with one or more variants of this cluster and/or of any other cluster and/or of any known marker, also comprises an embodiment of the present invention. It should be noted that the nucleotide transcript sequence of known protein (PSEC, also referred to herein as the “wild type” or WT protein) features at least one SNP that appears to affect the coding region, in addition to certain silent SNPs. This SNP does not have an effect on the R11723_PEA1_T5 (SEQ ID NO:9) splice variant sequence): “G->” resulting in a missing nucleotide (affects amino acids from position 91 onwards). The missing nucleotide creates a frame shift, resulting in a new protein. This SNP was not previously identified and is supported by 5 ESTs out of ˜70 ESTs in this exon.

Variant protein alignment to the previously known protein: Sequence name: /tmp/gp6eQTLWqk/mFtjupUzhb: Q8IXM0 Sequence documentation: Alignment of: R11723_PEA_1_P6 (SEQ ID NO: 332) × Q8IXM0 . . Alignment segment 1/1: Quality: 1128.00 Escore: 0 Matching length: 112 Total length: 112 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 111 MYAQALLVVGVLQRQAAAQHLHEHPPKLLRGHRVQERVDDRAEVEKRLRE 160 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MYAQALLVVGVLQRQAAAQHLHEHPPKLLRGHRVQERVDDRAEVEKRLRE 50          .         .         .         .         . 161 GEEDHVRPEVGPRPVVLGFGRSHDPPNLVGHPAYGQCHNNQPWADTSRRE 210 |||||||||||||||||||||||||||||||||||||||||||||||||| 51 GEEDHVRPEVGPRPVVLGFGRSHDPPNLVGHPAYGQCHNNQPWADTSRRE 100          . 211 RQRKEKHSMRTQ 222 |||||||||||| 101 RQRKEKHSMRTQ 112 Sequence name: /tmp/gp6eQTLWqk/mFtjUpUzhb: Q96AC2 Sequence documentation: Alignment of: R11723_PEA_1_P6 (SEQ ID NO: 332) × Q96AC2 . . Alignment segment 1/1: Quality: 835.00 Escore: 0 Matching length: 83 Total length: 83 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          .         .         . 51 QDMCQKEVMEQSAGIMYRKSCASSAACLIASAG 83 ||||||||||||||||||||||||||||||||| 51 QDMCQKEVMEQSAGIMYRKSCASSAACLIASAG 83 Sequence name: /tmp/gp6eQTLWqk/mFtjUpUzhb: Q8N2G4 Sequence documentation: Alignment of: R11723_PEA_1_P6 (SEQ ID NO: 332) × Q8N2G4 . . Alignment segment 1/1: Quality: 835.00 Escore: 0 Matching length: 83 Total length: 83 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          .         .         . 51 QDMCQKEVMEQSAGIMYRKSCASSAACLIASAG 83 ||||||||||||||||||||||||||||||||| 51 QDMCQKEVMEQSAGIMYRKSCASSAACLIASAG 83 Sequence name: /tmp/gp6eQTLWqk/mFtjUpUzhb: BAC85518 Sequence documentation: Alignment of: R11723_PEA_1_P6 (SEQ ID NO: 332) × BAC85518 . . Alignment segment 1/1: Quality: 835.00 Escore: 0 Matching length: 83 Total length: 83 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 24 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 73          .         .         . 51 QDMCQKEVMEQSAGIMYRKSCASSAACLIASAG 83 ||||||||||||||||||||||||||||||||| 74 QDMCQKEVMEQSAGIMYRKSCASSAACLIASAG 106 Sequence name: /tmp/VXjdFlzdBX/bexTxTh0Th: Q96AC2 Sequence documentation: Alignment of: R11723_PEA_1_P7 (SEQ ID NO: 333) × Q96AC2 . . Alignment segment 1/1: Quality: 654.00 Escore: 0 Matching length: 64 Total length: 64 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          . 51 QDMCQKEVMEQSAG 64 |||||||||||||| 51 QDMCQKEVMEQSAG 64 Sequence name: /tmp/VXjdFlzdBX/bexTxTh0Th: Q8N2G4 Sequence documentation: Alignment of: R11723_PEA_1_P7 (SEQ ID NO: 333) × Q8N2G4 . . Alignment segment 1/1: Quality: 654.00 Escore: 0 Matching length: 64 Total length: 64 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          . 51 QDMCQKEVMEQSAG 64 |||||||||||||| 51 QDMCQKEVMEQSAG 64 Sequence name: /tmp/VXjdFlzdBX/bexTxm0Th: BAC85273 Sequence documentation: Alignment of: R11723_PEA_1_P7 (SEQ ID NO: 333) × BAC85273 . . Alignment segment 1/1: Quality: 600.00 Escore: 0 Matching length: 59 Total length: 59 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 6 IAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQ 55 |||||||||||||||||||||||||||||||||||||||||||||||||| 22 IAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQ 71          . 56 KEVMEQSAG 64 ||||||||| 72 KEVMEQSAG 80 Sequence name: /tmp/VXjdFlzdBX/bexTxTh0Th: BACS5518 Sequence documentation: Alignment of: R11723_PEA_1_P7 (SEQ ID NO: 333) × BAC85518 . . Alignment segment 1/1: Quality: 654.00 Escore: 0 Matching length: 64 Total length: 64 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 24 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 73          . 51 QDMCQKEVMEQSAG 64 |||||||||||||| 74 QDMCQKEVMEQSAG 87 Sequence name: /tmp/OLMSexEmIh/pc7Z7Xm1YR: Q96AC2 Sequence documentation: Alignment of: R11723_PEA_1_P10 (SEQ ID NO: 335) × Q96AC2 . . Alignment segment 1/1: Quality: 645.00 Escore: 0 Matching length: 63 Total length: 63 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          . 51 QDMCQKEVMEQSA 63 ||||||||||||| 51 QDMCQKEVMEQSA 63 Sequence name: /tmp/OLMSexEmIh/pc7Z7Xm1YR: Q8N2G4 Sequence documentation: Alignment of: R11723_PEA_1_P10 (SEQ ID NO: 335) × Q8N2G4 . . Alignment segment 1/1: Quality: 645.00 Escore: 0 Matching length: 63 Total length: 63 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          . 51 QDMCQKEVMEQSA 63 ||||||||||||| 51 QDMCQKEVMEQSA 63 Sequence name: /tmp/OLMSexEmIh/pc7Z7Xm1YR: BAC85273 Sequence documentation: Alignment of: R11723_PEA_1_P10 (SEQ ID NO: 335) × BAC85273 . . Alignment segment 1/1: Quality: 591.00 Escore: 0 Matching length: 58 Total length: 58 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 6 IAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQ 55 |||||||||||||||||||||||||||||||||||||||||||||||||| 22 IAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQ 71 56 KEVMEQSA 63 |||||||| 72 KEVMEQSA 79 Sequence name: /tmp/OLMSexEmIh/pc7Z7Xm1YR: BAC85518 Sequence documentation: Alignment of: R11723_PEA_1_P10 (SEQ ID NO: 335) × BAC85518 . . Alignment segment 1/1: Quality: 645.00 Escore: 0 Matching length: 63 Total length: 63 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 24 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 73          . 51 QDMCQKEVMEQSA 63 ||||||||||||| 74 QDMCQKEVMEQSA 86 Alignment of: R11723_PEA_1_P13 (SEQ ID NO: 334) × Q96AC2 . . Alignment segment 1/1: Quality: 645.00 Escore: 0 Matching length: 63 Total length: 63 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MWVLGIAATFCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNV 50          . 51 QDMCQKEVMEQSA 63 ||||||||||||| 51 QDMCQKEVMEQSA 63

Expression of R11723 Transcripts which are Detectable by Amplicon as Depicted in Sequence Name R11723 seg13 in Normal and Cancerous Prostate Tissues

Expression of transcripts detectable by or according to seg13, R11732seg13 (SEQ ID NO:492) amplicon (s) and R11732seg13F (SEQ ID NO:490) and R11732seg13R (SEQ ID NO:491) primers was measured by real time PCR. In parallel the expression of four housekeeping genes—PBGD (GenBank Accession No. BC019323 (SEQ ID NO:509); amplicon—PBGD—amplicon, HPRT1 (GenBank Accession No. NM000194 (SEQ ID NO:510); amplicon—HPRT1-amplicon (SEQ ID NO:401), SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SDHA-, RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); RPL19 amplicon (SEQ ID NO:410) was measured similarly. For each RT sample, the expression of the above amplicon was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the normal post-mortem (PM) samples (Sample Nos. 42, 48-53, 59-63, Table 1 above, “Tissue samples in testing panel”), to obtain a value of fold up-regulation for each sample relative to median of the normal PM samples.

FIG. 7 is a histogram showing over expression of the above-indicated transcripts in cancerous prostate samples relative to the normal samples. Values represent the average of duplicate experiments. Error bars indicate the minimal and maximal values obtained).

As is evident from FIG. 7, the expression of transcripts detectable by the above amplicon in cancer samples was higher than in the non-cancerous samples (Sample Nos. 42, 48-53, 59-63, Table 1 above, “Tissue samples in testing panel”). Notably an over-expression of at least 5 fold was found in 4 out of 19 adenocarcinoma samples Statistical analysis was applied to verify the significance of these results, as described below.

The P value for the difference in the expression levels of transcripts detectable by the above amplicon (s) in prostate cancer samples versus the normal tissue samples was determined by T test as 7.57E-02.

The above values demonstrate statistical significance of the results. Primer pairs are also optionally and preferably encompassed within the present invention; for example, for the above experiment, the following primer pair was used as a non-limiting illustrative example only of a suitable primer pair: R11732seg13F forward primer (SEQ ID NO:490); and R11732seg13R reverse primer (SEQ ID NO:491).

The present invention also preferably encompasses any amplicon obtained through the use of any suitable primer pair; for example, for the above experiment, the following amplicon was obtained as a non-limiting illustrative example only of a suitable amplicon:R11732seg13 (SEQ ID NO:492)

R11732seg13F (SEQ ID NO: 490)- ACACTAAAAGAACAAACACCTTGCTC R11732seg13R (SEQ ID NO: 491)- TCCTCAGAAGGCACATGAAAGA R11732seg13 (SEQ ID NO: 492) (SEQ ID NO: 492) - ACACTAAAAGAACAAACACCTTGCTCTTCGAGATGAGACATTTTGCCAAG CAGTTGACCACTTAGTTCTCAAGAAGCAACTATCTCTTTCATGTGCCTTC TGAGGA

Expression of R11723 Transcripts which are Detectable by Amplicon as Depicted in Sequence Name R11723seg13 (SEQ ID NO:492) in Different Normal Tissues

Expression of R11723 transcripts detectable by or according to R11723seg13 (SEQ ID NO:492) amplicon and R11723seg13F (SEQ ID NO:490), R11723seg13R (SEQ ID NO:491) was measured by real time PCR. In parallel the expression of four housekeeping genes: RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); RPL19 amplicon (SEQ ID NO:410), TATA box (GenBank Accession No. NM003194 (SEQ ID NO:512); TATA amplicon (SEQ ID NO:515), Ubiquitin (GenBank Accession No. BC000449 (SEQ ID NO:516); amplicon—Ubiquitin-amplicon (SEQ ID NO:519)) and SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SDHA-amplicon was measured similarly. For each RT sample, the expression of the above amplicon was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the ovary samples (Sample Nos. 18-20 Table 2 “Tissue samples in normal panel”, above), to obtain a value of relative expression of each sample relative to median of the ovary samples. Primers and amplicon are as above.

The results are presented in FIG. 8, demonstrating the expression of R11723 transcripts which are detectable by amplicon as depicted in sequence name R11723seg13 (SEQ ID NO:492) in different normal tissues.

Expression of R11723 Transcripts, which are Detectable by Amplicon as Depicted in Sequence Name R11723junc11-18 (SEQ ID NO:495) in Normal and Cancerous Prostate Tissues.

Expression of transcripts detectable by or according to junc11-18 R11732junc11-18 amplicon (SEQ ID NO:495) and R11732junc11-18F (SEQ ID NO:493) and R11732junc11-18R (SEQ ID NO:494) primers was measured by real time PCR (this junction is found in the known protein sequence or “wild type” (WT) sequence, also termed herein the PSEC sequence). In parallel the expression of four housekeeping genes—PBGD (GenBank Accession No. BC019323 (SEQ ID NO:509); amplicon—PBGD-amplicon (SEQ ID NO:404)), HPRT1 (GenBank Accession No. NM000194 (SEQ ID NO:510); amplicon—HPRT1-amplicon (SEQ ID NO:401)), SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SDHA-amplicon, and RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); RPL19 amplicon (SEQ ID NO:410) was measured similarly. For each RT sample, the expression of the above amplicon was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the normal post-mortem (PM) samples (Sample Nos. 42, 48-53, 59-63 Table 1, above “Tissue samples in prostate cancer testing panel”), to obtain a value of fold up-regulation for each sample relative to median of the normal PM samples.

FIG. 9A is a histogram showing over expression of the above-indicated transcripts in cancerous prostate samples relative to the normal samples.

As is evident from FIG. 9A, the expression of transcripts detectable by the above amplicon in a few cancer samples was higher than in the non-cancerous samples (Sample Nos. 42, 48-53, 59-63, Table 1, above: “Tissue samples in prostate cancer testing panel”). Notably an over-expression of at least 5 fold was found in 2 out of 19 adenocarcinoma samples

Primer pairs are also optionally and preferably encompassed within the present invention; for example, for the above experiment, the following primer pair was used as a non-limiting illustrative example only of a suitable primer pair: R11732junc11-18F forward primer (SEQ ID NO:493); and R11732 junc11-18R reverse primer (SEQ ID NO:494).

The present invention also preferably encompasses any amplicon obtained through the use of any suitable primer pair; for example, for the above experiment, the following amplicon was obtained as a non-limiting illustrative example only of a suitable amplicon: R11732 junc11-18 (SEQ ID NO:495)

R11723junc11-18F (SEQ ID NO: 493)- AGTGATGGAGCAAAGTGCCG R11723 junc11-18R (SEQ ID NO: 494)- CAGCAGCTGATGCAAACTGAG R11723 junc11-18 (SEQ ID NO: 495)- AGTGATGGAGCAAAGTGCCGGGATCATGTACCGCAAGTCCTGTGCATCAT CAGCGGCCTGTCTCATCGCCTCTGCCGGGTACCAGTCCTTCTGCTCCCCA GGGAAACTGAACTCAGTTTGCATCAGCTGCTG

Expression of R11723 Transcripts, which were Detected by Amplicon as Depicted in the Sequence Name R11723 junc11-18 (SEQ ID NO:495) in Different Normal Tissues.

Expression of R11723 transcripts detectable by or according to R1723seg13 amplicon (SEQ ID NO:495) and R11723junc11-18F (SEQ ID NO:493), R11723junc11-18R (SEQ ID NO:494) was measured by real time PCR. In parallel the expression of four housekeeping genes RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); RPL19 amplicon (SEQ ID NO:410), TATA box (GenBank Accession No. NM003194 (SEQ ID NO:512); TATA amplicon (SEQ ID NO:515), UBC (GenBank Accession No. BC000449 (SEQ ID NO:516); amplicon—Ubiquitin-amplicon (SEQ ID NO:519) and SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SDHA-amplicon (SEQ ID NO:407) was measured similarly. For each RT sample, the expression of the above amplicon was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the ovary samples (Sample Nos. 18-20, Table 2, “Tissue samples in normal panel”, above), to obtain a value of relative expression of each sample relative to median of the ovary samples. Results are shown in FIG. 9B; primers and amplicon are as above.

The expression of variant transcripts relating to the R11723 cluster (also known as PSEC) was found to be similar to that of the WT (known or wild type) protein; however in some cancers, expression of one or more variant transcripts was found to be higher (R11723_T5 for example in certain tissues).

DESCRIPTION FOR CLUSTER HUMTREFAC

Cluster HUMTREFAC features 2 transcript(s) and 7 segment(s) of interest, the names for which are given in Tables 44 and 45, respectively, the sequences themselves are given at the end of the application. The selected protein variants are given in table 46.

TABLE 44 Transcripts of interest Transcript Name Sequence ID No. HUMTREFAC_PEA_2_T4 13 HUMTREFAC_PEA_2_T5 14

TABLE 45 Segments of interest Segment Name Sequence ID No. HUMTREFAC_PEA_2_node_0 136 HUMTREFAC_PEA_2_node_9 137 HUMTREFAC_PEA_2_node_2 138 HUMTREFAC_PEA_2_node_3 139 HUMTREFAC_PEA_2_node_4 140 HUMTREFAC_PEA_2_node_5 141 HUMTREFAC_PEA_2_node_8 142

TABLE 46 Proteins of interest Sequence Protein Name ID No. Corresponding Transcript(s) HUMTREFAC_PEA_2_P7 338 HUMTREFAC_PEA_2_T5 (SEQ ID NO: 14) HUMTREFAC_PEA_2_P8 339 HUMTREFAC_PEA_2_T4 (SEQ ID NO: 13)

These sequences are variants of the known protein Trefoil factor 3 precursor (SEQ ID NO:387) (SwissProt accession identifier TFF3—HUMAN (SEQ ID NO 387); known also according to the synonyms Intestinal trefoil factor; hP1.B), SEQ ID NO: 387, referred to herein as the previously known protein.

Protein Trefoil factor 3 precursor (SEQ ID NO:387) is known or believed to have the following function(s): May have a role in promoting cell migration (motogen). The sequence for protein Trefoil factor 3 precursor (SEQ ID NO:387) is given at the end of the application, as “Trefoil factor 3 precursor (SEQ ID NO:387) amino acid sequence”. Known polymorphisms for this sequence are as shown in Table 47.

TABLE 47 Amino acid mutations for Known Protein SNP position(s) on amino acid sequence Comment 74-76 QEA -> TRKT

Protein Trefoil factor 3 precursor (SEQ ID NO:387) localization is believed to be Secreted.

The following GO Annotation(s) apply to the previously known protein. The following annotation(s) were found: defense response; digestion, which are annotation(s) related to Biological Process; and extracellular, which are annotation(s) related to Cellular Component.

The GO assignment relies on information from one or more of the SwissProt/TremBl Protein knowledgebase, available from <http://www.expasy.ch/sprot/>; or Locuslink, available from <http://www.ncbi.nlm.nih.gov/projects/LocusLink/>.

Cluster HUMTREFAC can be used as a diagnostic marker according to overexpression of transcripts of this cluster in cancer. Expression of such transcripts in normal tissues is also given according to the previously described methods. The term “number” in the left hand column of the table and the numbers on the y-axis of FIG. 10 refer to weighted expression of ESTs in each category, as “parts per million” (ratio of the expression of ESTs for a particular cluster to the expression of all ESTs in that category, according to parts per million).

Overall, the following results were obtained as shown with regard to the histograms in FIG. 10 and Table 48. This cluster is overexpressed (at least at a minimum level) in the following pathological conditions: a mixture of malignant tumors from different tissues, breast malignant tumors, pancreas carcinoma and prostate cancer.

TABLE 48 Normal tissue distribution Name of Tissue Number Adrenal 40 Colon 797 Epithelial 95 General 39 Liver 0 Lung 57 Lymph nodes 3 Breast 0 Muscle 3 Pancreas 2 Prostate 16 Stomach 0 Thyroid 257 Uterus 54

TABLE 49 P values and ratios for expression in cancerous tissue Name of Tissue P1 P2 SP1 R3 SP2 R4 Adrenal 6.4e−01 6.9e−01 7.1e−01 1.1 7.8e−01 0.9 Colon 4.6e−01 5.7e−01 9.7e−01 0.5 1 0.4 Epithelial 2.4e−02 3.4e−01 9.5e−10 2.0 5.3e−02 1.1 General 2.5e−04 3.9e−02 1.4e−28 3.6 1.9e−10 1.9 Liver 1 6.8e−01 1 1.0 6.9e−01 1.4 Lung 4.8e−01 7.6e−01 2.2e−03 1.0 1.6e−01 0.5 Lymph nodes 5.1e−01 8.0e−01 2.3e−02 5.0 1.9e−01 2.1 Breast 7.6e−02 1.2e−01 3.1e−06 12.0 1.1e−03 6.5 Muscle 9.2e−01 4.8e−01 1 0.8 3.9e−01 2.1 Pancreas 1.2e−01 2.4e−01 5.7e−03 6.5 2.1e−02 4.6 Prostate 1.5e−01 2.7e−01 9.9e−10 8.1 3.1e−07 5.7 Stomach 3.0e−01 1.3e−01 5.0e−01 2.0 6.7e−02 2.8 Thyroid 6.4e−01 6.4e−01 9.6e−01 0.5 9.6e−01 0.5 Uterus 4.1e−01 7.3e−01 7.5e−02 1.3 4.0e−01 0.8

As noted above, cluster HUMTREFAC features 2 transcript(s), which were listed in Table 44 above. These transcript(s) encode for protein(s) which are variant(s) of protein Trefoil factor 3 precursor (SEQ ID NO:387). A description of each variant protein according to the present invention is now provided.

Variant protein HUMTREFAC_PEA2_P7 (SEQ ID NO:338) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HUMTREFAC_PEA2_T5 (SEQ ID NO:14). The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HUMTREFAC_PEA2_P7 (SEQ ID NO:338) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 50, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HUMTREFAC_PEA2 P7 (SEQ ID NO:338) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 50 Amino acid mutations SNP position(s) on amino acid Previously sequence Alternative amino acid(s) known SNP? 5 A -> S No 5 A -> T No 14 A -> V Yes 43 L -> M No 60 P -> S Yes 123 S -> * Yes

Variant protein HUMTREFAC_PEA2_P7 (SEQ ID NO:338) is encoded by the following transcript(s): HUMTREFAC_PEA2_T5 (SEQ ID NO:14), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript HUMTREFAC_PEA2_T5 (SEQ ID NO:14) is shown in bold; this coding portion starts at position 278 and ends at position 688. The transcript also has the following SNPs as listed in Table 51 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HUMTREFAC_PEA2_P7 (SEQ ID NO:338) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 51 Nucleic acid SNPs SNP position on nucleotide Previously sequence Alternative nucleic acid known SNP? 233 A -> G Yes 290 G -> A No 290 G -> T No 318 C -> T Yes 404 C -> A No 404 C -> T No 455 C -> T Yes 645 C -> A Yes 685 C -> T No

Variant protein HUMTREFAC_PEA2_P8 (SEQ ID NO:339) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HUMTREFAC_PEA-2-T4 (SEQ ID NO: 13). An alignment is given to the known protein (Trefoil factor 3 precursor (SEQ ID NO:387)) at the end of the application. One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between HUMTREFAC_PEA2_P8 (SEQ ID NO:339) and

TFF3—HUMAN:

1. An isolated chimeric polypeptide encoding for HUMTREFAC_PEA2_P8 (SEQ ID NO:339), comprising a first amino acid sequence being at least 90% homologous to MAARALCMLGLVLALLSSSSAEEYVGL corresponding to amino acids 1-27 of TFF3—HUMAN, which also corresponds to amino acids 1-27 of HUMTREFAC_PEA2_P8 (SEQ ID NO:339), and a second amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence WKVHLPKGEGFSSG (SEQ ID NO: 543) corresponding to amino acids 28-41 of HUMTREFAC_PEA2_P8 (SEQ ID NO:339) wherein said first amino acid sequence and second amino acid sequence are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of HUMTREFAC_PEA2_P8 (SEQ ID NO:339), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence WKVHLPKGEGFSSG (SEQ ID NO: 543) in HUMTREFAC_PEA2_P8 (SEQ ID NO:339).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HUMTREFAC_PEA2_P8 (SEQ ID NO:339) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 52, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HUMTREFAC_PEA2-P8 (SEQ ID NO:339) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 52 Amino acid mutations SNP position(s) on amino acid Previously sequence Alternative amino acid(s) known SNP? 5 A -> S No 5 A -> T No 14 A -> V Yes

Variant protein HUMTREFAC_PEA2_P8 (SEQ ID NO:339) is encoded by the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO:13), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript HUMTREFAC_PEA 2_T4 (SEQ ID NO:13) is shown in bold; this coding portion starts at position 278 and ends at position 400. The transcript also has the following SNPs as listed in Table 53 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HUMTREFAC_PEA2_P8 (SEQ ID NO:339) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 53 Nucleic acid SNPs SNP position on nucleotide Previously sequence Alternative nucleic acid known SNP? 233 A -> G Yes 290 G -> A No 290 G -> T No 318 C -> T Yes 515 C -> A No 515 C -> T No 566 C -> T Yes 756 C -> A Yes 796 C -> T No 1265 A -> C No 1266 A -> T No

As noted above, cluster HUMTREFAC features 7 segment(s), which were listed in Table 121 above and for which the sequence(s) are given at the end of the application. These segment(s) are portions of nucleic acid sequence(s) which are described herein separately because they are of particular interest. A description of each segment according to the present invention is now provided.

Segment cluster HUMTREFAC_PEA2_node0 (SEQ ID NO:136) according to the present invention is supported by 188 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO:13) and HUMTREFAC_PEA2_T5 (SEQ ID NO: 14). Table 54 below describes the starting and ending position of this segment on each transcript.

TABLE 54 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 1 359 ID NO: 13) HUMTREFAC_PEA_2_T5 (SEQ 1 359 ID NO: 14)

Segment cluster HUMTREFAC_PEA2_node9 (SEQ ID NO:137) according to the present invention is supported by 150 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO: 13) and HUMTREFAC_PEA2_T5 (SEQ ID NO: 14). Table 55 below describes the starting and ending position of this segment on each transcript.

TABLE 55 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 681 1266 ID NO: 13) HUMTREFAC_PEA_2_T5 (SEQ 570 747 ID NO: 14)

According to an optional embodiment of the present invention, short segments related to the above cluster are also provided. These segments are up to about 120 bp in length, and so are included in a separate description.

Segment cluster HUMTREFAC_PEA2_node2 (SEQ ID NO: 138) according to the present invention is supported by 4 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO: 13). Table 56 below describes the starting and ending position of this segment on each transcript.

TABLE 56 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 360 470 ID NO: 13)

Segment cluster HUMTREFAC_PEA2_node3 (SEQ ID NO:139) according to the present invention is supported by 10 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO: 13) and HUMTREFAC_PEA2_T5 (SEQ ID NO: 14). Table 57 below describes the starting and ending position of this segment on each transcript.

TABLE 57 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 471 514 ID NO: 13) HUMTREFAC_PEA_2_T5 (SEQ 360 403 ID NO: 14)

Segment cluster HUMTREFAC_PEA2_node4 (SEQ ID NO:140) according to the present invention is supported by 197 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO:13) and HUMTREFAC_PEA2_T5 (SEQ ID NO:14). Table 58 below describes the starting and ending position of this segment on each transcript.

TABLE 58 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 515 611 ID NO: 13) HUMTREFAC_PEA_2_T5 (SEQ 404 500 ID NO: 14)

Segment cluster HUMTREFAC_PEA2_node5 (SEQ ID NO:141) according to the present invention is supported by 187 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO: 13) and HUMTREFAC_PEA2_T5 (SEQ ID NO: 14). Table 59 below describes the starting and ending position of this segment on each transcript.

TABLE 59 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 612 661 ID NO: 13) HUMTREFAC_PEA_2_T5 (SEQ 501 550 ID NO: 14)

Segment cluster HUMTREFAC_PEA2_node8 (SEQ ID NO:142) according to the present invention can be found in the following transcript(s): HUMTREFAC_PEA2_T4 (SEQ ID NO:13) and HUMTREFAC_PEA2_T5 (SEQ ID NO:14). Table 60 below describes the starting and ending position of this segment on each transcript.

TABLE 60 Segment location on transcripts Segment Segment Transcript name starting position ending position HUMTREFAC_PEA_2_T4 (SEQ 662 680 ID NO: 13) HUMTREFAC_PEA_2_T5 (SEQ 551 569 ID NO: 14)

Variant protein alignment to the previously known protein:

Sequence name: TFF3_HUMAN Sequence documentation: Alignment of: HUMTREFAC_PEA_2_P8 (SEQ ID NO: 339) × TFF3_HUMAN . . Alignment segment 1/1: Quality: 246.00 Escore: 0 Matching length: 27 Total length: 27 Matching Percent 100.00 Matching Percent 100.00 Similarity: Identity: Total Percent 100.00 Total Percent 100.00 Similarity: Identity: Gaps: 0 Alignment:          .         . 1 MAARALCMLGLVLALLSSSSAEEYVGL 27 ||||||||||||||||||||||||||| 1 MAARALCMLGLVLALLSSSSAEEYVGL 27

Description for Cluster HSSTROL3

Cluster HSSTROL3 features 6 transcript(s) and 16 segment(s) of interest, the names for which are given in Tables 61 and 62, respectively, the sequences themselves are given at the end of the application. The selected protein variants are given in table 63.

TABLE 61 Transcripts of interest Transcript Name Sequence ID No. HSSTROL3_T5 34 HSSTROL3_T8 35 HSSTROL3_T9 36 HSSTROL3_T10 37 HSSTROL3_T11 38 HSSTROL3_T12 39

TABLE 62 Segments of interest Segment Name Sequence ID No. HSSTROL3_node_6 222 HSSTROL3_node_10 223 HSSTROL3_node_13 224 HSSTROL3_node_15 225 HSSTROL3_node_19 226 HSSTROL3_node_21 227 HSSTROL3_node_24 228 HSSTROL3_node_25 229 HSSTROL3_node_26 230 HSSTROL3_node_28 231 HSSTROL3_node_29 232 HSSTROL3_node_11 233 HSSTROL3_node_17 234 HSSTROL3_node_18 235 HSSTROL3_node_20 236 HSSTROL3_node_27 237

TABLE 63 Proteins of interest Protein Name Sequence ID No. Corresponding Transcript(s) HSSTROL3_P4 359 HSSTROL3_T5 (SEQ ID NO: 34) HSSTROL3_P5 360 HSSTROL3_T8 (SEQ ID NO: 35); HSSTROL3_T9 (SEQ ID NO: 36) HSSTROL3_P7 361 HSSTROL3_T10 (SEQ ID NO: 37) HSSTROL3_P8 362 HSSTROL3_T11 (SEQ ID NO: 38) HSSTROL3_P9 363 HSSTROL3_T12 (SEQ ID NO: 39)

These sequences are variants of the known protein Stromelysin-3 precursor (SEQ ID NO:391) (SwissProt accession identifier MM11_HUMAN (SEQ ID NO: 391); known also according to the synonyms EC 3.4.24.-; Matrix metalloproteinase-11; MMP-11; ST3; SL-3), SEQ ID NO: 391) referred to herein as the previously known protein.

Protein Stromelysin-3 precursor (SEQ ID NO:391) is known or believed to have the following function(s): May play an important role in the progression of epithelial malignancies. The sequence for protein Stromelysin-3 precursor (SEQ ID NO:391) is given at the end of the application, as “Stromelysin-3 precursor (SEQ ID NO:391) amino acid sequence”.

The following GO Annotation(s) apply to the previously known protein. The following annotation(s) were found: proteolysis and peptidolysis; developmental processes; morphogenesis, which are annotation(s) related to Biological Process; stromelysin 3; calcium binding; zinc binding; hydrolase, which are annotation(s) related to Molecular Function; and extracellular matrix, which are annotation(s) related to Cellular Component.

The GO assignment relies on information from one or more of the SwissProt/TremBl Protein knowledgebase, available from <http://www.expasy.ch/sprot/>; or Locuslink, available from <http://www.ncbi.nlm.nih.gov/projects/LocusLink/>.

Cluster HSSTROL3 can be used as a diagnostic marker according to overexpression of transcripts of this cluster in cancer. Expression of such transcripts in normal tissues is also given according to the previously described methods. The term “number” in the left hand column of the table and the numbers on the y-axis of FIG. 11 refer to weighted expression of ESTs in each category, as “parts per million” (ratio of the expression of ESTs for a particular cluster to the expression of all ESTs in that category, according to parts per million).

Overall, the following results were obtained as shown with regard to the histograms in FIG. 11 and Table 64. This cluster is overexpressed (at least at a minimum level) in the following pathological conditions: transitional cell carcinoma, epithelial malignant tumors, a mixture of malignant tumors from different tissues and pancreas carcinoma.

TABLE 64 Normal tissue distribution Name of Tissue Number Adrenal 0 Bladder 0 Brain 1 Colon 63 Epithelial 33 General 13 head and neck 101 Kidney 0 Lung 11 Breast 8 Ovary 14 Pancreas 0 Prostate 2 Skin 99 Thyroid 0 Uterus 181

TABLE 65 P values and ratios for expression in cancerous tissue Name of Tissue P1 P2 SP1 R3 SP2 R4 Adrenal 1 4.6e−01 1 1.0 5.3e−01 1.9 Bladder 2.7e−01 3.4e−01 3.3e−03 4.9 2.1e−02 3.3 Brain 3.5e−01 2.6e−01 1 1.7 3.3e−01 2.8 Colon 7.7e−02 1.5e−01 3.1e−01 1.4 5.2e−01 1.0 Epithelial 1.2e−04 1.2e−02 1.3e−06 2.7 4.6e−02 1.4 General 5.4e−09 3.1e−05 1.8e−16 5.0 3.1e−07 2.6 head and neck 4.6e−01 4.3e−01 1 0.6 9.4e−01 0.7 Kidney 2.5e−01 3.5e−01 1.1e−01 4.0 2.4e−01 2.8 Lung 1.8e−01 4.5e−01 1.9e−01 2.7 5.1e−01 1.4 Breast 2.0e−01 3.4e−01 7.3e−02 3.3 2.5e−01 2.0 Ovary 2.6e−01 3.2e−01 2.2e−02 2.0 7.0e−02 1.6 Pancreas 9.5e−02 1.8e−01 1.8e−04 7.8 1.6e−03 5.5 Prostate 8.2e−01 7.8e−01 4.5e−01 1.8 5.6e−01 1.5 Skin 5.2e−01 5.8e−01 7.1e−01 0.8 1 0.3 Thyroid 2.9e−01 2.9e−01 1 1.1 1 1.1 Uterus 4.2e−01 8.0e−01 7.5e−01 0.6 9.9e−01 0.4

As noted above, cluster HSSTROL3 features 6 transcript(s), which were listed in Table 65 above. These transcript(s) encode for protein(s) which are variant(s) of protein Stromelysin-3 precursor (SEQ ID NO:391). A description of each variant protein according to the present invention is now provided.

Variant protein HSSTROL3_P4 (SEQ ID NO:359) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HSSTROL3_T5 (SEQ ID NO:34). An alignment is given to the known protein (Stromelysin-3 precursor (SEQ ID NO:391)) at the end of the application. One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between HSSTROL3_P4 (SEQ ID NO:359) and MM11_HUMAN:

1. An isolated chimeric polypeptide encoding for HSSTROL3_P4 (SEQ ID NO:359) comprising a first amino acid sequence being at least 90% homologous to MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQPWHAALPSS PAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVLSGGRWEKTDLTYRILRFP WQLVQEQVRQTMAEALKVWSDVTPLTFTEVHEGRADIMIDFARYW corresponding to amino acids 1-163 of MM11_HUMAN, which also corresponds to amino acids 1-163 of HSSTROL3_P4 (SEQ ID NO:359), a bridging amino acid H corresponding to amino acid 164 of HSSTROL3_P4 (SEQ ID NO:359), a second amino acid sequence being at least 90% homologous to GDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWTIGDDQGTDLLQVAAHEFGHVLG LQHTTAAKALMSAFYTFRYPLSLSPDDCRGVQHLYGQPWPTVTSRTPALGPQAGIDTN EIAPLEPDAPPDACEASFDAVSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGL PSPVDAAFEDAQGHIWFFQGAQYWVYDGEKPVLGPAPLTELGLVRFPVHAALVWGPE KNKIYFFRGRDYWRFHPSTRRVDSPVPRRATDWRGVPSEIDAAFQDADG corresponding to amino acids 165-445 of MM11_HUMAN, which also corresponds to amino acids 165-445 of HSSTROL3_P4 (SEQ ID NO:359), and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence ALGVRQLVGGGHSSRFSHLVVAGLPHACHRKSGSSSQVLCPEPSALLSVAG (SEQ ID NO: 564) corresponding to amino acids 446-496 of HSSTROL3_P4 (SEQ ID NO:359), wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of HSSTROL3_P4 (SEQ ID NO:359), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence ALGVRQLVGGGHSSRFSHLVVAGLPHACHRKSGSSSQVLCPEPSALLSVAG in (SEQ ID NO: 564) HSSTROL3_P4 (SEQ ID NO:359).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HSSTROL3_P4 (SEQ ID NO:359) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 66, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P4 (SEQ ID NO:359) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 66 Amino acid mutations SNP position(s) on Alternative Previously amino acid sequence amino acid(s) known SNP? 38 V -> A Yes 104 R -> P Yes 214 A -> No 323 Q -> H Yes

Variant protein HSSTROL3_P4 (SEQ ID NO:359) is encoded by the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript HSSTROL3_T5 (SEQ ID NO:34) is shown in bold; this coding portion starts at position 24 and ends at position 1511. The transcript also has the following SNPs as listed in Table 67 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P4 (SEQ ID NO:359) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 67 Nucleic acid SNPs SNP position on Alternative Previously nucleotide sequence nucleic acid known SNP? 136 T -> C Yes 334 G -> C Yes 663 G -> No 699 -> T No 992 G -> C Yes 1528 A -> G Yes 1710 A -> G Yes 2251 A -> G Yes 2392 C -> No 2444 C -> A Yes 2470 A -> T Yes 2687 -> G No 2696 -> G No 2710 C -> No 2729 -> A No 2755 T -> C No 2813 A -> No 2813 A -> C No 2963 A -> No 2963 A -> C No 2993 T -> C Yes 3140 -> T No

Variant protein HSSTROL3_P5 (SEQ ID NO:360) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HSSTROL3_T8 (SEQ ID NO:35) and HSSTROL3_T9 (SEQ ID NO:36). An alignment is given to the known protein (Stromelysin-3 precursor (SEQ ID NO:391) at the end of the application. One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between HSSTROL3_P5 (SEQ ID NO:360) and MM11_HUMAN:

1. An isolated chimeric polypeptide encoding for HSSTROL3_P5 (SEQ ID NO:360), comprising a first amino acid sequence being at least 90% homologous to MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQPWHAALPSS PAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVLSGGRWEKTDLTYRILRFP WQLVQEQVRQTMAEALKVWSDVTPLTFTEVHEGRADIMIDFARYW corresponding to amino acids 1-163 of MM11_HUMAN, which also corresponds to amino acids 1-163 of HSSTROL3_P5 (SEQ ID NO:360), a bridging amino acid H corresponding to amino acid 164 of HSSTROL3_P5 (SEQ ID NO:360), a second amino acid sequence being at least 90% homologous to GDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWTIGDDQGTDLLQVAAHEFGHVLG LQHTTAAKALMSAFYTFRYPLSLSPDDCRGVQHLYGQPWPTVTSRTPALGPQAGIDTN EIAPLEPDAPPDACEASFDAVSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGL PSPVDAAFEDAQGHIWFFQ corresponding to amino acids 165-358 of MM11_HUMAN, which also corresponds to amino acids 165-358 of HSSTROL3_P5 (SEQ ID NO:360), and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence ELGFPSSTGRDESLEHCRCQGLHK (SEQ ID NO: 565) corresponding to amino acids 359-382 of HSSTROL3_P5 (SEQ ID NO:360), wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of HSSTROL3_P5 (SEQ ID NO:360), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence ELGFPSSTGRDESLEHCRCQGLHK (SEQ ID NO: 565) in HSSTROL3_P5 (SEQ ID NO:360).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HSSTROL3_P5 (SEQ ID NO:360) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 68, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P5 (SEQ ID NO:360) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 68 Amino acid mutations SNP position(s) on Alternative Previously amino acid sequence amino acid(s) known SNP? 38 V -> A Yes 104 R -> P Yes 214 A -> No 323 Q -> H Yes

Variant protein HSSTROL3_P5 (SEQ ID NO:360) is encoded by the following transcript(s): HSSTROL3_T8 (SEQ ID NO:35) and HSSTROL3_T9 (SEQ ID NO:36), for which the sequence(s) is/are given at the end of the application.

The coding portion of transcript HSSTROL3_T8 (SEQ ID NO:35) is shown in bold; this coding portion starts at position 24 and ends at position 1169. The transcript also has the following SNPs as listed in Table 69 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P5 (SEQ ID NO:360) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 69 Nucleic acid SNPs SNP position on Alternative Previously nucleotide sequence nucleic acid known SNP? 136 T -> C Yes 334 G -> C Yes 663 G -> No 699 -> T No 992 G -> C Yes 1903 C -> No 1955 C -> A Yes 1981 A -> T Yes 2198 -> G No 2207 -> G No 2221 C -> No 2240 -> A No 2266 T -> C No 2324 A -> No 2324 A -> C No 2474 A -> No 2474 A -> C No 2504 T -> C Yes 2651 -> T No

The coding portion of transcript HSSTROL3_T9 (SEQ ID NO:36) is shown in bold; this coding portion starts at position 24 and ends at position 1169. The transcript also has the following SNPs as listed in Table 70 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P5 (SEQ ID NO:360) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 70 Nucleic acid SNPs SNP position on Alternative Previously nucleotide sequence nucleic acid known SNP? 136 T -> C Yes 334 G -> C Yes 663 G -> No 699 -> T No 992 G -> C Yes 1666 A -> G Yes 1848 A -> G Yes 2389 A -> G Yes 2530 C -> No 2582 C -> A Yes 2608 A -> T Yes 2825 -> G No 2834 -> G No 2848 C -> No 2867 -> A No 2893 T -> C No 2951 A -> No 2951 A -> C No 3101 A -> No 3101 A -> C No 3131 T -> C Yes 3278 -> T No

Variant protein HSSTROL3_P7 (SEQ ID NO:361) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HSSTROL3_T10 (SEQ ID NO:37). An alignment is given to the known protein (Stromelysin-3 precursor (SEQ ID NO:391)) at the end of the application. One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between HSSTROL3_P7 (SEQ ID NO:361) and MM11_HUMAN:

1. An isolated chimeric polypeptide encoding for HSSTROL3_P7 (SEQ ID NO:361) comprising a first amino acid sequence being at least 90% homologous to MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQPWHAALPSS PAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVLSGGRWEKTDLTYRILRFP WQLVQEQVRQTMAEALKVWSDVTPLTFTEVHEGRADIMIDFARYW corresponding to amino acids 1-163 of MM11_HUMAN, which also corresponds to amino acids 1-163 of HSSTROL3_P7 (SEQ ID NO:361), a bridging amino acid H corresponding to amino acid 164 of HSSTROL3_P7 (SEQ ID NO:361), a second amino acid sequence being at least 90% homologous to GDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWTIGDDQGTDLLQVAAHEFGHVLG LQHTTAAKALMSAFYTFRYPLSLSPDDCRGVQHLYGQPWPTVTSRTPALGPQAGIDTN EIAPLEPDAPPDACEASFDAVSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGL PSPVDAAFEDAQGHIWFFQG corresponding to amino acids 165-359 of MM11_HUMAN, which also corresponds to amino acids 165-359 of HSSTROL3_P7 (SEQ ID NO:361), and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence TTGVSTPAPGV (SEQ ID NO: 566) corresponding to amino acids 360-370 of HSSTROL3_P7 (SEQ ID NO:361), wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of HSSTROL3_P7 (SEQ ID NO:361) comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence TTGVSTPAPGV (SEQ ID NO: 566) in HSSTROL3_P7 (SEQ ID NO:361).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HSSTROL3_P7 (SEQ ID NO:361) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 71, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P7 (SEQ ID NO:361) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 71 Amino acid mutations SNP position(s) on Alternative Previously amino acid sequence amino acid(s) known SNP? 38 V -> A Yes 104 R -> P Yes 214 A -> No 323 Q -> H Yes

Variant protein HSSTROL3_P7 (SEQ ID NO:361) is encoded by the following transcript(s): HSSTROL3_T10 (SEQ ID NO:37), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript HSSTROL3_T10 (SEQ ID NO:37) is shown in bold; this coding portion starts at position 24 and ends at position 1133. The transcript also has the following SNPs as listed in Table 72 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P7 (SEQ ID NO:361) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 72 Nucleic acid SNPs SNP position on Alternative Previously nucleotide sequence nucleic acid known SNP? 136 T -> C Yes 334 G -> C Yes 663 G -> No 699 -> T No 992 G -> C Yes 1386 A -> G Yes 1568 A -> G Yes 2109 A -> G Yes 2250 C -> No 2302 C -> A Yes 2328 A -> T Yes 2545 -> G No 2554 -> G No 2568 C -> No 2587 -> A No 2613 T -> C No 2671 A -> No 2671 A -> C No 2821 A -> No 2821 A -> C No 2851 T -> C Yes 2998 -> T No

Variant protein HSSTROL3_P8 (SEQ ID NO:362) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HSSTROL3_T11 (SEQ ID NO:38). An alignment is given to the known protein (Stromelysin-3 precursor (SEQ ID NO:391)) at the end of the application. One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between HSSTROL3_P8 (SEQ ID NO:362) and MM11_HUMAN:

1. An isolated chimeric polypeptide encoding for HSSTROL3_P8 (SEQ ID NO:362) comprising a first amino acid sequence being at least 90% homologous to MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQPWHAALPSS PAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVLSGGRWEKTDLTYRILRFP WQLVQEQVRQTMAEALKVWSDVTPLTFTEVHEGRADIMIDFARYW corresponding to amino acids 1-163 of MM 11_HUMAN, which also corresponds to amino acids 1-163 of HSSTROL3_P8 (SEQ ID NO:362), a bridging amino acid H corresponding to amino acid 164 of HSSTROL3_P8 (SEQ ID NO:362), a second amino acid sequence being at least 90% homologous to GDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWTIGDDQGTDLLQVAAHEFGHVLG LQHTTAAKALMSAFYTFRYPLSLSPDDCRGVQHLYGQPWPTVTSRTPALGPQAGIDTN EIAPLE corresponding to amino acids 165-286 of MM11_HUMAN, which also corresponds to amino acids 165-286 of HSSTROL3_P8 (SEQ ID NO:362), and a third amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence VRPCLPVPLLLCWPL (SEQ ID NO: 567) corresponding to amino acids 287-301 of HSSTROL3_P8 (SEQ ID NO:362), wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order.

2. An isolated polypeptide encoding for a tail of HSSTROL3_P8 (SEQ ID NO:362), comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence VRPCLPVPLLLCWPL (SEQ ID NO: 567) in HSSTROL3_P8 (SEQ ID NO:362).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HSSTROL3_P8 (SEQ ID NO:362) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 73, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P8 (SEQ ID NO:362) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 73 Amino acid mutations SNP position(s) on Alternative Previously amino acid sequence amino acid(s) known SNP? 38 V -> A Yes 104 R -> P Yes 214 A -> No

Variant protein HSSTROL3_P8 (SEQ ID NO:362) is encoded by the following transcript(s): HSSTROL3_T11 (SEQ ID NO:38), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript HSSTROL3_T11 (SEQ ID NO:38) is shown in bold; this coding portion starts at position 24 and ends at position 926. The transcript also has the following SNPs as listed in Table 74 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P8 (SEQ ID NO:362) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 74 Nucleic acid SNPs SNP position on Alternative Previously nucleotide sequence nucleic acid known SNP? 136 T -> C Yes 334 G -> C Yes 663 G -> No 699 -> T No 935 G -> A Yes 948 G -> A Yes 1084 G -> C Yes 1557 C -> No 1609 C -> A Yes 1635 A -> T Yes 1852 -> G No 1861 -> G No 1875 C -> No 1894 -> A No 1920 T -> C No 1978 A -> No 1978 A -> C No 2128 A -> No 2128 A -> C No 2158 T -> C Yes 2305 -> T No

Variant protein HSSTROL3_P9 (SEQ ID NO:363) according to the present invention has an amino acid sequence as given at the end of the application; it is encoded by transcript(s) HSSTROL3_T12 (SEQ ID NO:39). An alignment is given to the known protein (Stromelysin-3 precursor (SEQ ID NO:391) at the end of the application. One or more alignments to one or more previously published protein sequences are given at the end of the application. A brief description of the relationship of the variant protein according to the present invention to each such aligned protein is as follows:

Comparison report between HSSTROL3_P9 (SEQ ID NO:363) and MM11_HUMAN:

1. An isolated chimeric polypeptide encoding for HSSTROL3_P9 (SEQ ID NO:363) comprising a first amino acid sequence being at least 90% homologous to MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQPWHAALPSS PAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQK corresponding to amino acids 1-96 of MM11_HUMAN, which also corresponds to amino acids 1-96 of HSSTROL3_P9 (SEQ ID NO:363), a second amino acid sequence being at least 90% homologous to RILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHEGRADIMIDFARYW corresponding to amino acids 113-163 of MM11_HUMAN, which also corresponds to amino acids 97-147 of HSSTROL3_P9 (SEQ ID NO:363), a bridging amino acid H corresponding to amino acid 148 of HSSTROL3_P9 (SEQ ID NO:363), a third amino acid sequence being at least 90% homologous to GDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWTIGDDQGTDLLQVAAHEFGHVLG LQHTTAAKALMSAFYTFRYPLSLSPDDCRGVQHLYGQPWPTVTSRTPALGPQAGIDTN EIAPLEPDAPPDACEASFDAVSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGL PSPVDAAFEDAQGHIWFFQG corresponding to amino acids 165-359 of MM11_HUMAN, which also corresponds to amino acids 149-343 of HSSTROL3_P9 (SEQ ID NO:363), and a fourth amino acid sequence being at least 70%, optionally at least 80%, preferably at least 85%, more preferably at least 90% and most preferably at least 95% homologous to a polypeptide having the sequence TTGVSTPAPGV (SEQ ID NO: 566) corresponding to amino acids 344-354 of HSSTROL3_P9 (SEQ ID NO:363), wherein said first amino acid sequence, second amino acid sequence, bridging amino acid, third amino acid sequence and fourth amino acid sequence are contiguous and in a sequential order.

2. An isolated chimeric polypeptide encoding for an edge portion of HSSTROL3_P9 (SEQ ID NO:363), comprising a polypeptide having a length “n”, wherein n is at least about 10 amino acids in length, optionally at least about 20 amino acids in length, preferably at least about 30 amino acids in length, more preferably at least about 40 amino acids in length and most preferably at least about 50 amino acids in length, wherein at least two amino acids comprise KR, having a structure as follows: a sequence starting from any of amino acid numbers 96-x to 96; and ending at any of amino acid numbers 97+((n-2)−x), in which x varies from 0 to n-2.

3. An isolated polypeptide encoding for a tail of HSSTROL3_P9 (SEQ ID NO:363) comprising a polypeptide being at least 70%, optionally at least about 80%, preferably at least about 85%, more preferably at least about 90% and most preferably at least about 95% homologous to the sequence TTGVSTPAPGV (SEQ ID NO: 566) in HSSTROL3_P9 (SEQ ID NO:363).

The location of the variant protein was determined according to results from a number of different software programs and analyses, including analyses from SignalP and other specialized programs. The variant protein is believed to be located as follows with regard to the cell: secreted. The protein localization is believed to be secreted because both signal-peptide prediction programs predict that this protein has a signal peptide, and neither trans-membrane region prediction program predicts that this protein has a trans-membrane region.

Variant protein HSSTROL3_P9 (SEQ ID NO:363) also has the following non-silent SNPs (Single Nucleotide Polymorphisms) as listed in Table 75, (given according to their position(s) on the amino acid sequence, with the alternative amino acid(s) listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P9 (SEQ ID NO:363) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 75 Amino acid mutations SNP position(s) on amino acid Previously sequence Alternative amino acid(s) known SNP? 38 V -> A Yes 198 A -> No 307 Q -> H Yes

Variant protein HSSTROL3_P9 (SEQ ID NO:363) is encoded by the following transcript(s): HSSTROL3_T12 (SEQ ID NO:39), for which the sequence(s) is/are given at the end of the application. The coding portion of transcript HSSTROL3_T12 (SEQ ID NO:39) is shown in bold; this coding portion starts at position 24 and ends at position 1085. The transcript also has the following SNPs as listed in Table 76 (given according to their position on the nucleotide sequence, with the alternative nucleic acid listed; the last column indicates whether the SNP is known or not; the presence of known SNPs in variant protein HSSTROL3_P9 (SEQ ID NO:363) sequence provides support for the deduced sequence of this variant protein according to the present invention).

TABLE 76 Nucleic acid SNPs SNP position on nucleotide Previously sequence Alternative nucleic acid known SNP? 136 T -> C Yes 615 G -> No 651 -> T No 944 G -> C Yes 1275 C -> No 1327 C -> A Yes 1353 A -> T Yes 1570 -> G No 1579 -> G No 1593 C -> No 1612 -> A No 1638 T -> C No 1696 A -> No 1696 A -> C No 1846 A -> No 1846 A -> C No 1876 T -> C Yes 2023 -> T No

As noted above, cluster HSSTROL3 features 16 segment(s), which were listed in Table 63 above and for which the sequence(s) are given at the end of the application. These segment(s) are portions of nucleic acid sequence(s) which are described herein separately because they are of particular interest. A description of each segment according to the present invention is now provided.

Segment cluster HSSTROL3_node6 (SEQ ID NO:222) according to the present invention is supported by 14 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 77 below describes the starting and ending position of this segment on each transcript.

TABLE 77 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 1 131 HSSTROL3_T8 (SEQ ID NO: 35) 1 131 HSSTROL3_T9 (SEQ ID NO: 36) 1 131 HSSTROL3_T10 (SEQ ID NO: 37) 1 131 HSSTROL3_T11 (SEQ ID NO: 38) 1 131 HSSTROL3_T12 (SEQ ID NO: 39) 1 131

Segment cluster HSSTROL3_node10 (SEQ ID NO:223) according to the present invention is supported by 21 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 78 below describes the starting and ending position of this segment on each transcript.

TABLE 78 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 132 313 HSSTROL3_T8 (SEQ ID NO: 35) 132 313 HSSTROL3_T9 (SEQ ID NO: 36) 132 313 HSSTROL3_T10 (SEQ ID NO: 37) 132 313 HSSTROL3_T11 (SEQ ID NO: 38) 132 313 HSSTROL3_T12 (SEQ ID NO: 39) 132 313

Segment cluster HSSTROL3_node13 (SEQ ID NO:224) according to the present invention is supported by 36 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36) HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 79 below describes the starting and ending position of this segment on each transcript.

TABLE 79 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 362 505 HSSTROL3_T8 (SEQ ID NO: 35) 362 505 HSSTROL3_T9 (SEQ ID NO: 36) 362 505 HSSTROL3_T10 (SEQ ID NO: 37) 362 505 HSSTROL3_T11 (SEQ ID NO: 38) 362 505 HSSTROL3_T12 (SEQ ID NO: 39) 314 457

Segment cluster HSSTROL3_node15 (SEQ ID NO:225) according to the present invention is supported by 47 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 80 below describes the starting and ending position of this segment on each transcript.

TABLE 80 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 506 639 HSSTROL3_T8 (SEQ ID NO: 35) 506 639 HSSTROL3_T9 (SEQ ID NO: 36) 506 639 HSSTROL3_T10 (SEQ ID NO: 37) 506 639 HSSTROL3_T11 (SEQ ID NO: 38) 506 639 HSSTROL3_T12 (SEQ ID NO: 39) 458 591

Segment cluster HSSTROL3_node19 (SEQ ID NO:226) according to the present invention is supported by 63 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 81 below describes the starting and ending position of this segment on each transcript.

TABLE 81 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 699 881 HSSTROL3_T8 (SEQ ID NO: 35) 699 881 HSSTROL3_T9 (SEQ ID NO: 36) 699 881 HSSTROL3_T10 (SEQ ID NO: 37) 699 881 HSSTROL3_T11 (SEQ ID NO: 38) 699 881 HSSTROL3_T12 (SEQ ID NO: 39) 651 833

Segment cluster HSSTROL3_node21 (SEQ ID NO:227) according to the present invention is supported by 61 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 82 below describes the starting and ending position of this segment on each transcript.

TABLE 82 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 882 1098 HSSTROL3_T8 (SEQ ID NO: 35) 882 1098 HSSTROL3_T9 (SEQ ID NO: 36) 882 1098 HSSTROL3_T10 (SEQ ID NO: 37) 882 1098 HSSTROL3_T11 (SEQ ID NO: 38) 974 1190 HSSTROL3_T12 (SEQ ID NO: 39) 834 1050

Segment cluster HSSTROL3_node24 (SEQ ID NO:228) according to the present invention is supported by 7 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T8 (SEQ ID NO:35) and HSSTROL3_T9 (SEQ ID NO:36). Table 83 below describes the starting and ending position of this segment on each transcript.

TABLE 83 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T8 (SEQ ID NO: 35) 1099 1236 HSSTROL3_T9 (SEQ ID NO: 36) 1099 1236

Segment cluster HSSTROL3_node25 (SEQ ID NO:229) according to the present invention is supported by 13 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T8 (SEQ ID NO:35). Table 84 below describes the starting and ending position of this segment on each transcript.

TABLE 84 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T8 (SEQ ID NO: 35) 1237 1536

Segment cluster HSSTROL3_node26 (SEQ ID NO:230) according to the present invention is supported by 55 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36) and HSSTROL3_T111 (SEQ ID NO:38). Table 85 below describes the starting and ending position of this segment on each transcript.

TABLE 85 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 1099 1240 HSSTROL3_T8 (SEQ ID NO: 35) 1537 1678 HSSTROL3_T9 (SEQ ID NO: 36) 1237 1378 HSSTROL3_T11 (SEQ ID NO: 38) 1191 1332

Segment cluster HSSTROL3_node28 (SEQ ID NO:231) according to the present invention is supported by 10 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T9 (SEQ ID NO:36) and HSSTROL3_T10 (SEQ ID NO:37). Table 86 below describes the starting and ending position of this segment on each transcript.

TABLE 86 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 1357 2283 HSSTROL3_T9 (SEQ ID NO: 36) 1495 2421 HSSTROL3_T10 (SEQ ID NO: 37) 1215 2141

Segment cluster HSSTROL3_node29 (SEQ ID NO:232) according to the present invention is supported by 109 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 87 below describes the starting and ending position of this segment on each transcript.

TABLE 87 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 2284 3194 HSSTROL3_T8 (SEQ ID NO: 35) 1795 2705 HSSTROL3_T9 (SEQ ID NO: 36) 2422 3332 HSSTROL3_T10 (SEQ ID NO: 37) 2142 3052 HSSTROL3_T11 (SEQ ID NO: 38) 1449 2359 HSSTROL3_T12 (SEQ ID NO: 39) 1167 2077

According to an optional embodiment of the present invention, short segments related to the above cluster are also provided. These segments are up to about 120 bp in length, and so are included in a separate description.

Segment cluster HSSTROL3_node11 (SEQ ID NO:233) according to the present invention is supported by 25 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37) and HSSTROL3_T11 (SEQ ID NO:38). Table 88 below describes the starting and ending position of this segment on each transcript.

TABLE 88 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 314 361 HSSTROL3_T8 (SEQ ID NO: 35) 314 361 HSSTROL3_T9 (SEQ ID NO: 36) 314 361 HSSTROL3_T10 (SEQ ID NO: 37) 314 361 HSSTROL3_T11 (SEQ ID NO: 38) 314 361

Segment cluster HSSTROL3_node17 (SEQ ID NO:234) according to the present invention is supported by 45 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 89 below describes the starting and ending position of this segment on each transcript.

TABLE 89 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 640 680 HSSTROL3_T8 (SEQ ID NO: 35) 640 680 HSSTROL3_T9 (SEQ ID NO: 36) 640 680 HSSTROL3_T10 (SEQ ID NO: 37) 640 680 HSSTROL3_T11 (SEQ ID NO: 38) 640 680 HSSTROL3_T12 (SEQ ID NO: 39) 592 632

Segment cluster HSSTROL3_node18 (SEQ ID NO:235) according to the present invention can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 90 below describes the starting and ending position of this segment on each transcript.

TABLE 90 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T5 (SEQ ID NO: 34) 681 698 HSSTROL3_T8 (SEQ ID NO: 35) 681 698 HSSTROL3_T9 (SEQ ID NO: 36) 681 698 HSSTROL3_T10 (SEQ ID NO: 37) 681 698 HSSTROL3_T11 (SEQ ID NO: 38) 681 698 HSSTROL3_T12 (SEQ ID NO: 39) 633 650

Segment cluster HSSTROL3_node20 (SEQ ID NO:236) according to the present invention is supported by 1 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T11 (SEQ ID NO:38). Table 91 below describes the starting and ending position of this segment on each transcript.

TABLE 91 Segment location on transcripts Segment Segment Transcript name starting position ending position HSSTROL3_T11 (SEQ ID NO: 38) 882 973

Segment cluster HSSTROL3_node27 (SEQ ID NO:237) according to the present invention is supported by 50 libraries. The number of libraries was determined as previously described. This segment can be found in the following transcript(s): HSSTROL3_T5 (SEQ ID NO:34), HSSTROL3_T8 (SEQ ID NO:35), HSSTROL3_T9 (SEQ ID NO:36), HSSTROL3_T10 (SEQ ID NO:37), HSSTROL3_T11 (SEQ ID NO:38) and HSSTROL3_T12 (SEQ ID NO:39). Table 92 below describes the starting and ending position of this segment on each transcript.

TABLE 92 Segment location on transcripts Segment Segment Transcript name Starting position ending position HSSTROL3_T5 (SEQ ID NO:34) 1241 1356 HSSTROL3_T8 (SEQ ID NO:35) 1679 1794 HSSTROL3_T9 (SEQ ID NO:36) 1379 1494 HSSTROL3_T10 (SEQ ID NO:37) 1099 1214 HSSTROL3_T11 (SEQ ID NO:38) 1333 1448 HSSTROL3_T12 (SEQ ID NO:39) 1051 1166

Variant protein alignment to the previously known protein: Sequence name: MM11_HUMAN Sequence documentation: Alignment of: HSSTROL3_P4 (SEQ ID NO: 359) × MM11_HUMAN . . Alignment segment 1/1: Quality: 4444.00 Escore: 0 Matching length: 445 Total length: 445 Matching Percent 99.78 Matching Percent 99.78 Similarity: Identity: Total Percent 99.78 Total Percent 99.78 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50          .         .         .         .         . 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100 |||||||||||||||||||||||||||||||||||||||||||||||||| 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100          .         .         .         .         . 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150 |||||||||||||||||||||||||||||||||||||||||||||||||| 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150          .         .         .         .         . 151 GRADIMIDFARYWHGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200 |||||||||||||||||||||||||||||||||||||||||||||||||| 151 GRADIMIDFARYWDGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200          .         .         .         .         . 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250 |||||||||||||||||||||||||||||||||||||||||||||||||| 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250          .         .         .         .         . 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300 |||||||||||||||||||||||||||||||||||||||||||||||||| 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300          .         .         .         .         . 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350 |||||||||||||||||||||||||||||||||||||||||||||||||| 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350          .         .         .         .         . 351 QGHIWFFQGAQYWVYDGEKPVLGPAPLTELGLVRFPVHAALVWGPEKNKI 400 |||||||||||||||||||||||||||||||||||||||||||||||||| 351 QGHIWFFQGAQYWVYDGEKPVLGPAPLTELGLVRFPVHAALVWGPEKNKI 400          .         .         .         . 401 YFFRGRDYWRFHPSTRRVDSPVPRRATDWRGVPSEIDAAFQDADG 445 ||||||||||||||||||||||||||||||||||||||||||||| 401 YFFRGRDYWRFHPSTRRVDSPVPRRATDWRGVPSEIDAAFQDADG 445 Sequence name: MM11_HUMAN Sequence documentation: Alignment of: HSSTROL3_P5 (SEQ ID NO: 360) × MM11_HUMAN . . Alignment segment 1/1: Quality: 3566.00 Escore: 0 Matching length: 358 Total length: 358 Matching Percent 99.72 Matching Percent 99.72 Similarity: Identity: Total Percent 99.72 Total Percent 99.72 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50          .         .         .         .         . 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100 |||||||||||||||||||||||||||||||||||||||||||||||||| 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100          .         .         .         .         . 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150 |||||||||||||||||||||||||||||||||||||||||||||||||| 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150          .         .         .         .         . 151 GRADIMIDFARYWHGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200 |||||||||||||||||||||||||||||||||||||||||||||||||| 151 GRADIMIDFARYWDGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200          .         .         .         .         . 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250 |||||||||||||||||||||||||||||||||||||||||||||||||| 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250          .         .         .         .         . 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300 |||||||||||||||||||||||||||||||||||||||||||||||||| 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300          .         .         .         .         . 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350 |||||||||||||||||||||||||||||||||||||||||||||||||| 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350 351 QGHIWFFQ 358 |||||||| 351 QGHIWFFQ 358 Sequence name: MM11_HUMAN Sequence documentation: Alignment of: HSSTROL3_P7 (SEQ ID NO: 361) × MM11_HUMAN . . Alignment segment 1/1: Quality: 3575.00 Escore: 0 Matching length: 359 Total length: 359 Matching Percent 99.72 Matching Percent 99.72 Similarity: Identity: Total Percent 99.72 Total Percent 99.72 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MAPAAWLRSAAAPALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MAPAAWLRSAAAPALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50          .         .         .         .         . 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100 |||||||||||||||||||||||||||||||||||||||||||||||||| 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100          .         .         .         .         . 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150 |||||||||||||||||||||||||||||||||||||||||||||||||| 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150          .         .         .         .         . 151 GRADIMIDFARYWHGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200 |||||||||||||||||||||||||||||||||||||||||||||||||| 151 GRADIMIDFARYWDGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200          .         .         .         .         . 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250 |||||||||||||||||||||||||||||||||||||||||||||||||| 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250          .         .         .         .         . 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300 |||||||||||||||||||||||||||||||||||||||||||||||||| 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300          .         .         .         .         . 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350 |||||||||||||||||||||||||||||||||||||||||||||||||| 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350 351 QGHIWFFQG 359 ||||||||| 351 QGHIWFFQG 359 Sequence name: MM11_HUMAN Sequence documentation: Alignment of: HSSTROL3_P8 (SEQ ID NO: 362) × MM11_HUMAN . . Alignment segment 1/1: Quality: 2838.00 Escore: 0 Matching length: 286 Total length: 286 Matching Percent 99.65 Matching Percent 99.65 Similarity: Identity: Total Percent 99.65 Total Percent 99.65 Similarity: Identity: Gaps: 0 Alignment:          .         .         .         .         . 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50          .         .         .         .         . 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100 |||||||||||||||||||||||||||||||||||||||||||||||||| 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100          .         .         .         .         . 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150 |||||||||||||||||||||||||||||||||||||||||||||||||| 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150          .         .         .         .         . 151 GPADIMIDFARYWHGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200 |||||||||||||||||||||||||||||||||||||||||||||||||| 151 GRADIMIDFARYWDGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200          .         .         .         .         . 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250 |||||||||||||||||||||||||||||||||||||||||||||||||| 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250          .         .         . 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLE 286 |||||||||||||||||||||||||||||||||||| 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLE 286 Sequence name: MM11_HUMAN Sequence documentation: Alignment of: HSSTROL3_P9 (SEQ ID NO: 363) × MM11_HUMAN . . Alignment segment 1/1: Quality: 3316.00 Escore: 0 Matching length: 343 Total length: 359 Matching Percent 99.71 Matching Percent 99.71 Similarity: Identity: Total Percent 95.26 Total Percent 95.26 Similarity: Identity: Gaps: 1 Alignment:          .         .         .         .         . 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50 |||||||||||||||||||||||||||||||||||||||||||||||||| 1 MAPAAWLRSAAARALLPPMLLLLLQPPPLLARALPPDVHHLHAERRGPQP 50          .         .         .         .         . 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQK.... 96 |||||||||||||||||||||||||||||||||||||||||||||| 51 WHAALPSSPAPAPATQEAPRPASSLRPPRCGVPDPSDGLSARNRQKRFVL 100          .         .         .         .         . 97 ............RILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 134             |||||||||||||||||||||||||||||||||||||| 101 SGGRWEKTDLTYRILRFPWQLVQEQVRQTMAEALKVWSDVTPLTFTEVHE 150          .         .         .         .         . 135 GRADIMIDFARYWHGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 184 ||||||||||||| |||||||||||||||||||||||||||||||||||| 151 GRADIMIDFARYWDGDDLPFDGPGGILAHAFFPKTHREGDVHFDYDETWT 200          .         .         .         .         . 185 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 234 |||||||||||||||||||||||||||||||||||||||||||||||||| 201 IGDDQGTDLLQVAAHEFGHVLGLQHTTAAKALMSAFYTFRYPLSLSPDDC 250          .         .         .         .         . 235 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 284 |||||||||||||||||||||||||||||||||||||||||||||||||| 251 RGVQHLYGQPWPTVTSRTPALGPQAGIDTNEIAPLEPDAPPDACEASFDA 300          .         .         .         .         . 285 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 334 |||||||||||||||||||||||||||||||||||||||||||||||||| 301 VSTIRGELFFFKAGFVWRLRGGQLQPGYPALASRHWQGLPSPVDAAFEDA 350 335 QGHIWFFQG 343 ||||||||| 351 QGHIWFFQG 359

Expression of Stromelysin-3 precursor (SEQ ID NO:391) (EC 3.4.24.-) (Matrix metalloproteinase-11) (MMP-11) (ST3) (SL-3HSSTROL3) transcripts which are detectable by amplicon as depicted in sequence name HSSTROL3 seg24 (SEQ ID NO:499) in normal and cancerous Prostate tissues

Expression of Stromelysin-3 precursor (SEQ ID NO:391) (EC 3.4.24.-) (Matrix metalloproteinase-11) (MMP-11) (ST3) (SL-3) transcripts detectable by or according to seg24, HSSTROL3 seg24 (SEQ ID NO:499) amplicon(s) and HSSTROL3 seg24F (SEQ ID NO:497) and HSSTROL3 seg24R (SEQ ID NO:498) primers was measured by real time PCR. In parallel the expression of four housekeeping genes —PBGD (GenBank Accession No. BC019323 (SEQ ID NO:509); amplicon—PBGD-amplicon (SEQ ID NO:404), HPRT1 (GenBank Accession No. NM000194 (SEQ ID NO:510); amplicon—HPRT1-amplicon (SEQ ID NO:401), SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SDHA-amplicon (SEQ ID NO:407), and RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); RPL19 amplicon (SEQ ID NO:410) was measured similarly. For each RT sample, the expression of the above amplicon was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the normal post-mortem (PM) samples (Sample Nos. 42, 48-53, 59-63, Table 1, “Tissue samples in testing panel”, above), to obtain a value of fold up-regulation for each sample relative to median of the normal PM samples.

FIG. 12 is a histogram showing over expression of the above-indicated Stromelysin-3 precursor (SEQ ID NO:391) transcripts in cancerous Prostate samples relative to the normal samples. Values represent the average of duplicate experiments. Error bars indicate the minimal and maximal values obtained.)

As is evident from FIG. 12, the expression of Stromelysin-3 precursor (SEQ ID NO:391) transcripts detectable by the above amplicon(s) in cancer samples was higher than in several non-cancerous samples (Sample Nos. 42, 48-53, 59-63, Table 1, “Tissue samples in testing panel”). Notably an over-expression of at least 3 fold was found in 4 out of 19 adenocarcinoma samples.

Statistical analysis was applied to verify the significance of these results, as described below.

The P value for the difference in the expression levels of Stromelysin-3 precursor (SEQ ID NO:391) transcripts detectable by the above amplicon(s) in Prostate cancer samples versus the normal tissue samples was determined by T test as 2.34E-03.

The above value demonstrate statistical significance of the results.

Primer pairs are also optionally and preferably encompassed within the present invention; for example, for the above experiment, the following primer pair was used as a non-limiting illustrative example only of a suitable primer pair: HSSTROL3 seg24F forward primer (SEQ ID NO:497); and HSSTROL3 seg24R reverse primer (SEQ ID NO:498).

The present invention also preferably encompasses any amplicon obtained through the use of any suitable primer pair; for example, for the above experiment, the following amplicon was obtained as a non-limiting illustrative example only of a suitable amplicon: HSSTROL seg24 (SEQ ID NO:499).

HSSTROL seg24 Forward primer (SEQ ID NO: 497): ATTTCCATCCTCAACTGGCAGA HSSTROL seg24 Reverse primer (SEQ ID NO: 498): TGCCCTGGAACCCACG HSSTROL seg24 Amplicon (SEQ ID NO: 499): ATTTCCATCCTCAACTGGCAGAGATGAGAGCCTGGAGCATTGCAGATGCC AGGGACTTCACAAATGAAGGCACAGCATGGGAAACCTGCGTGGGTTCCAG GGCA

Expression of Stromelysin-3 precursor (SEQ ID NO:391) transcripts which are detectable by amplicon as depicted in sequence name HSSTROL3 seg24 (SEQ ID NO:499) in different normal tissues

Expression of Stromelysin-3 precursor (SEQ ID NO:391) transcripts detectable by or according to HSSTROL3 seg24 (SEQ ID NO:499) amplicon(s) and HSSTROL3 seg24F (SEQ ID NO:497) and HSSTROL3 seg24R (SEQ ID NO:498) was measured by real time PCR. In parallel the expression of four housekeeping genes Ubiquitin (GenBank Accession No. BC000449 (SEQ ID NO:516); amplicon—Ubiquitin-amplicon (SEQ ID NO:519) and SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SDHA-amplicon (SEQ ID NO:407), RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); RPL19 amplicon (SEQ ID NO:410)), TATA box (GenBank Accession No. NM003194 (SEQ ID NO:512); TATA amplicon (SEQ ID NO:515)) was measured similarly. For each RT sample, the expression of the above amplicon was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the lung samples (Sample Nos. 15-17, Table 2 “Tissue samples in normal panel”, above), to obtain a value of relative expression of each sample relative to median of the lung samples.

The results are shown in FIG. 13, demonstrating the expression of Stromelysin-3 transcripts which are detectable by amplicon as depicted in sequence name HSSTROL3 seg24 (SEQ ID NO:499) in different normal tissues.

Expression of Thrombospondin 1 (THBS1) Transcripts which are Detectable by SEQ ID NO:421 in Normal, Benign and Cancerous Prostate Tissues

Expression of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO:421, segment 24 (e.g., variants no. 10,11 and 30; SEQ ID NOs: 441, 442, and 451) was measured by real time PCR, according to the exemplary marker HUMTHROM-segment 24 (SEQ ID NO:425). In parallel the expression of four housekeeping genes—PBGD (GenBank Accession No. BC019323 (SEQ ID NO:509); amplicon—SEQ ID NO:404), HPRT1 (GenBank Accession No. NM000194 (SEQ ID NO:510); amplicon—SEQ ID NO:401), RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); amplicon—SEQ ID NO:410) and SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SEQ ID NO:407), was measured similarly. For each RT sample, the expression of SEQ ID NO:421 was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the normal post-mortem (PM) samples (Sample Nos. 42, 48-53, 59-63, Table 2, above), to obtain a value of fold up-regulation for each sample relative to median of the normal PM samples.

FIG. 14 is a histogram showing over expression of the above-indicated Thrombospondin 1 (THBS1) transcripts in cancerous and benign (BPH) prostate samples relative to the normal samples. The number and percentage of cancer samples that exhibit at least 3 fold over-expression, out of the total number of samples tested is indicated in the bottom.

As is evident from FIG. 14, the expression of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO:421 in cancer samples was significantly higher than in the normal PM samples (Sample Nos. 42, 48-53, 59-63, Table 2). Notably an over-expression of at least 3 fold was found in 10 out of 19 adenocarcinoma samples. Over expression of at least 3 fold was observed also in 7 out of the 8 BPH samples, and in the 2 matched normal samples. Since matched samples are histologically non-cancerous tissue that surrounds the tumor, such samples could have been contaminated with cancer or pre-cancer cells.

Statistical analysis was applied to verify the significance of these results, as described below.

The P value for the difference in the expression levels of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO:421 in prostate cancer samples versus the normal prostate samples was determined by T test as 9.92E-03.

Threshold of 3 fold overexpression was found to differentiate between cancer and normal samples with P value of 2.08E-03 as checked by exact fisher test.

The above value demonstrates statistical significance of the results.

According to the present invention, HUMTHROM is a non-limiting example of a marker for diagnosing prostate cancer. The HUMTHROM marker of the present invention, can be used alone or in combination, for prognosis, prediction, screening, early diagnosis, therapy selection and treatment monitoring of prostate cancer. Although optionally any method may be used to detect overexpression and/or differential expression of this marker, preferably a NAT-based technology is used. Therefore, optionally and preferably, any nucleic acid molecule capable of selectively hybridizing to HUMTHROM as previously defined is also encompassed within the present invention. Primer pairs are also optionally and preferably encompassed within the present invention; for example, for the above experiment, the following primer pair was used as a non-limiting illustrative example only of a suitable primer pair: HUMTHROM-seg24-forward (SEQ ID NO:419): CTGCAGGCTCAGCAACTTCTT; and HUMTHROM-seg24-reverse (SEQ ID NO:420): TTTCAAATCCCTCCCTTGTCA.

The present invention also preferably encompasses any amplicon obtained through the use of any suitable primer pair; for example, for the above experiment, the following amplicon was obtained as a non-limiting illustrative example only of a suitable amplicon:Amplicon from seg 24: (SEQ ID NO:421)

     CTGCAGGCTCAGCAACTTCTTTTAATGAAAAACAAACTCACCCTC TTCCCCAGCATTCTTTCCATGTGTCAGAGAAGCAGAGGTTTCTTGAACGG GCTTAGGAGAGTCTATGACAAGGGAGGGATTTGAAA.

According to other preferred embodiments of the present invention, HUMTHROM or a fragment thereof comprises a biomarker for detecting prostate cancer. Optionally and more preferably, the fragment of HUMTHROM comprises HUMTHROM-seg24 (SEQ ID NO:425). Also optionally and more preferably, any suitable method may be used for detecting a fragment such as HUMTHROM-seg24 for example. Most preferably, NAT-based technology used, such as any nucleic acid molecule capable of specifically hybridizing with the fragment. Optionally and most preferably, a primer pair is used for obtaining the fragment.

Optionally the HUMTHROM transcript could (additionally or alternatively) comprise any one or more of the following sequences: SEQ ID NOs: 435-440; 443-445; 447-450.

According to still other preferred embodiments, the present invention optionally and preferably encompasses any amino acid sequence or fragment thereof encoded by a nucleic acid sequence corresponding to HUMTHROM as described above or below. Any oligopeptide or peptide relating to such an amino acid sequence or fragment thereof may optionally also (additionally or alternatively) be used as a biomarker. The present invention also optionally encompasses antibodies capable of recognizing, and/or being elicited by, such an oligopeptide or peptide.

Also, optionally and preferably HUMTHROM could be detected by detection of an amino acid sequence according to any of SEQ ID NOs 452-463, for which the unique regions relating to the splice variants are given separately and additionally in SEQ ID NOs 464-472. The present invention also encompasses these amino acid sequences as a biomarker for detecting prostate cancer.

The present invention also optionally and preferably encompasses any nucleic acid sequence or fragment thereof, or amino acid sequence or fragment thereof, corresponding to HUMTHROM as described above or below, optionally for any application.

Expression of Thrombospondin 1 (THBS1) Transcripts which are Detectable by SEQ ID NO:418 in Normal, Benign and Cancerous Prostate Tissues

Expression of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO:418, segment 19 (e.g., variant no. 18; SEQ ID NO: 446) was measured by real time PCR, according to the exemplary, illustrative marker HUMTHROM-segment 19 (SEQ ID NO: 423). In parallel the expression of four housekeeping genes—PBGD (GenBank Accession No. BC019323 (SEQ ID NO:509); amplicon—SEQ ID NO:404), HPRT1 (GenBank Accession No. NM000194 (SEQ ID NO:510); amplicon—SEQ ID NO:401), RPL19 (GenBank Accession No. NM000981 (SEQ ID NO:511); amplicon—SEQ ID NO:410) and SDHA (GenBank Accession No. NM004168 (SEQ ID NO:508); amplicon—SEQ ID NO:407), was measured similarly. For each RT sample, the expression of SEQ ID NO:418 was normalized to the geometric mean of the quantities of the housekeeping genes. The normalized quantity of each RT sample was then divided by the median of the quantities of the normal post-mortem (PM) samples (Sample Nos. 42, 48-53, 59-63, Table 2, above), to obtain a value of fold up-regulation for each sample relative to median of the normal PM samples.

FIG. 15 is a histogram showing over expression of the above-indicated Thrombospondin 1 (THBS1) transcripts in cancerous and benign (BPH) prostate samples relative to the normal samples. The number and percentage of cancer samples that exhibit at least 3 fold over-expression, out of the total number of samples tested is indicated in the bottom.

As is evident from FIG. 15, the expression of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO: 418 in cancer samples was significantly higher than in the normal PM samples (Sample Nos. 42, 48-53, 59-63, Table 2). Notably an over-expression of at least 3 fold was found in 17 out of 19 adenocarcinoma samples. Over expression of at least 3 fold was observed also in 6 out of the 8 BPH samples, and in the 2 matched normal samples. Since matched samples are histologically non-cancerous tissue that surrounds the tumor, such samples could have been contaminated with cancer or pre-cancer cells. These samples were purchased commercially with the matching non-cancerous tissue samples, as described above.

Statistical analysis was applied to verify the significance of these results, as described below.

The P value for the difference in the expression levels of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO: 418 in prostate cancer samples versus the normal prostate samples was determined by T test as 1.17E-04.

Threshold of 3 fold overexpression was found to differentiate between cancer and normal samples with P value of 1.00E-05 as checked by exact fisher test.

The P value for the difference between the expression levels of Thrombospondin 1 (THBS1) transcripts detectable by SEQ ID NO: 418 in the prostate cancer samples versus the BPH and normal prostate samples was determined by T test as 7.36E-02. Threshold of 3 fold overexpression was found to differentiate between cancer sample and BPH and normal sample with P value of 5.42E-04 as checked by exact fisher test.

All the above values demonstrate statistical significance of the results.

According to the present invention, HUMTHROM is a non-limiting example of a marker for diagnosing prostate cancer. Although optionally any method may be used to detect overexpression and/or differential expression of this marker, preferably a NAT-based technology is used. Therefore, optionally and preferably, any nucleic acid molecule capable of selectively hybridizing to HUMTHROM as previously defined is also encompassed within the present invention. Primer pairs are also optionally and preferably encompassed within the present invention; for example, for the above experiment, the following primer pair was used as a non-limiting illustrative example only of a suitable primer pair:HUMTHROM-seg19-forward (SEQ ID NO:416): AAAGCATCCGATTACCCCACT and HUMTHROM-segl9-reverse (SEQ ID NO:417): CCGGCACAAAGTTGCAGTTA.

The present invention also preferably encompasses any amplicon obtained through the use of any suitable primer pair; for example, for the above experiment, the following amplicon was obtained as a non-limiting illustrative example only of a suitable amplicon:Amplicon from seg 19:

Kits and Diagnostic Assays and Methods

The markers described with regard to any of Examples 1-6 above can be used alone, in combination with other markers described above, and/or with other entirely different markers (including but not limited to DD3, PSA or prostate specific membrane antigen) to aid in the diagnosis of prostate cancer, benign prostate hyperplasia or a negative diagnosis. These markers can be used in combination with other markers for a number of uses, including but not limited to, prognosis, prediction, screening, early diagnosis, therapy selection and treatment monitoring of prostate cancer, and also optionally including staging of the disease. Used together, they tend to provide more information for the diagnostician, increasing the percentage of true positive and true negative diagnoses and decreasing the percentage of false positive or false negative diagnoses, than a single marker alone.

Assays and methods according to the present invention, as described above, include but are not limited to, immunoassays, hybridization assays and NAT-based assays. The combination of the markers of the present invention with other markers described above, and/or with other entirely different markers to aid in the diagnosis of prostate cancer could be carried out as a mix of NAT-based assays, immunoassays and hybridization assays. According to preferred embodiments of the present invention, the assays are NAT-based assays, as described for example with regard to the Examples above.

In yet another aspect, the present invention provides kits for aiding a diagnosis of prostate cancer, wherein the kits can be used to detect the markers of the present invention. For example, the kits can be used to detect any one or combination of markers described above, which markers are differentially present in samples of a prostate cancer patient, BPH and normal patients. The kits of the invention have many applications. For example, the kits can be used to differentiate if a subject has prostate cancer, BPH or has a negative diagnosis, thus aiding a prostate cancer diagnosis. In another example, the kits can be used to identify compounds that modulate expression of the markers in in vitro prostate cells or in vivo animal models for prostate cancer.

In one embodiment, a kit comprises: (a) a substrate comprising an adsorbent thereon, wherein the adsorbent is suitable for binding a marker, and (b) a washing solution or instructions for making a washing solution, wherein the combination of the adsorbent and the washing solution allows detection of the marker as previously described.

Optionally, the kit can further comprise instructions for suitable operational parameters in the form of a label or a separate insert. For example, the kit may have standard instructions informing a consumer/kit user how to wash the probe after a sample of seminal plasma or other tissue sample is contacted on the probe.

In another embodiment, a kit comprises (a) an antibody that specifically binds to a marker; and (b) a detection reagent. Such kits can be prepared from the materials described above.

In either embodiment, the kit may optionally further comprise a standard or control information, and/or a control amount of material, so that the test sample can be compared with the control information standard and/or control amount to determine if the test amount of a marker detected in a sample is a diagnostic amount consistent with a diagnosis of prostate cancer.

It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.

Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims. All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention.

Claims

1. An isolated polynucleotide comprising the polynucleotide sequence set forth in a member selected from the group consisting of 5, 6, 7, 8, 9, 10, 34, 35, 36, 37, 38, 39, 13, 14, 435, 436, 437-450 and 451.

2. An isolated polynucleotide, comprising the polynucleotide sequence set forth in a member selected from the group consisting of 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 136, 137, 138, 139, 140, 141, 142, 422, 423, 424-433 and 434.

3. An isolated polypeptide comprising the polypeptide sequence set forth in a member selected from the group consisting of 331, 332, 333, 334, 335, 359, 360, 361, 362, 363, 338, 339, 452, 453-462 and 463.

4. An isolated polypeptide, comprising the polypeptide selected from the group consisting of:

a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-63 of SEQ ID NO:334, and a second amino acid sequence being at least about 95% homologous to amino acids 64-84 of SEQ ID NO:334, wherein said first and second amino acid sequences are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least 95% homologous to amino acids 1-163 of SEQ ID NO. 359, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO. 359, a second amino acid sequence being at least 95% homologous to amino acids 165-445 of SEQ ID NO. 359, and a third amino acid sequence being at least about 95% homologous to amino acids 446-496 of SEQ ID NO. 359, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-163 of SEQ ID NO:360, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO. 360, a second amino acid sequence being at least about 95% homologous to amino acids 165-358 of SEQ ID NO. 360, and a third amino acid sequence being at least about 95% homologous to amino acids 359-382 of SEQ ID NO. 360, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-163 of SEQ ID NO. 361, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO. 361, a second amino acid sequence being at least about 95% homologous to amino acids 165-359 of SEQ ID NO. 361, and a third amino acid sequence being at least about 95% homologous to amino acids 360-370 of SEQ ID NO. 361, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-163 of SEQ ID NO. 362, a bridging amino acid H corresponding to amino acid 164 of SEQ ID NO. 362, a second amino acid sequence being at least about 95% homologous to amino acids 165-286 of SEQ ID NO. 362, and a third amino acid sequence being at least about 95% homologous to amino acids 287-301 of SEQ ID NO. 362, wherein said first amino acid sequence, bridging amino acid, second amino acid sequence and third amino acid sequence are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-96 of SEQ ID NO. 363, a second amino acid sequence being at least about 95% homologous to amino acids 97-147 of SEQ ID NO. 363, a bridging amino acid H corresponding to amino acid 148 of SEQ ID NO. 363, a third amino acid sequence being at least about 95% homologous to amino acids 149-343 of SEQ ID NO. 363, and a fourth amino acid sequence being at least about 95% homologous to amino acids 344-354 of SEQ ID NO. 363, wherein said first amino acid sequence, second amino acid sequence, bridging amino acid, third amino acid sequence and fourth amino acid sequence are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-27 of SEQ ID NO. 339, and a second amino acid sequence being at least about 95% homologous to amino acids 28-41 of SEQ ID NO. 339, wherein said first amino acid sequence and second amino acid sequence are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-110 of SEQ ID NO. 332, and a second amino acid sequence being at least about 95% homologous to amino acids 111-222 of SEQ ID NO. 332, wherein said first and second amino acid sequences are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-5 of SEQ ID NO. 333, a second amino acid sequence being at least about 95% homologous to amino acids 6-64 of SEQ ID NO. 333, and a third amino acid sequence being at least about 95% homologous to amino acids 65-93 of SEQ ID NO. 333, wherein said first, second and third amino acid sequences are contiguous and in a sequential order;
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-63 of SEQ ID NO. 334, and a second amino acid sequence being at least about 95% homologous to amino acids 64-84 of SEQ ID NO. 334, wherein said first and second amino acid sequences are contiguous and in a sequential order; and
a polypeptide comprising a first amino acid sequence being at least about 95% homologous to amino acids 1-63 of SEQ ID NO. 335, and a second amino acid sequence being at least about 95% homologous to a polypeptide sequence corresponding to amino acids 64-90 of SEQ ID NO. 335, wherein said first and second amino acid sequences are contiguous and in a sequential order.

5. The isolated polypeptide of claim 4, comprising a member selected from the group consisting of

a polypeptide comprising an amino acid sequence being at least about 95% about homologous to SEQ ID NO: 538;
a polypeptide comprising an amino acid sequence being at least about 95% about homologous to amino acids 446-496 in SEQ ID NO. 359;
a polypeptide comprising an amino acid sequence being at least about 95% about homologous to amino acids 359-382 in SEQ ID NO. 360;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 360-370 in SEQ ID NO. 361;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 287-301 in SEQ ID NO. 362;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to the sequence in SEQ ID NO. 363;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 28-41 in SEQ ID NO. 339;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 84-222 in SEQ ID NO. 332;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 65-93 in SEQ ID NO. 333;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 64-84 in SEQ ID NO. 334;
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 64-90 in SEQ ID NO. 335; and
a polypeptide comprising an amino acid sequence being at least about 95% homologous to amino acids 1-110 of SEQ ID NO. 332.

6. An isolated peptide, the peptide having a length “n”, wherein n is selected from the group consisting of 10 amino acids in length, 20 amino acids in length, 30 amino acids in length, 40 amino acids in length and 50 amino acids in length, wherein at least two amino acids comprise KR, having a structure as follows: a sequence starting from any of amino acid numbers 96-x to 96 of SEQ ID NO:363; and ending at any of amino acid numbers 97+((n-2)−x) of SEQ ID NO:363, in which x varies from 0 to n-2.

7. An isolated peptide comprising the amino acid sequence set forth in a 50 amino acid long segment of SEQ ID NO:363 anywhere from amino acid position 48 to amino acid position 145, and including amino acids at positions 96 and 97.

8. An isolated peptide comprising the amino acid sequence set forth in a 40 amino acid long segment of SEQ ID NO:363 anywhere from amino acid position 58 to amino acid position 135, and including amino acids at positions 96 and 97.

9. An isolated peptide comprising the amino acid sequence set forth in a 30 amino acid long segment of SEQ ID NO:363 anywhere from amino acid position 68 to amino acid position 125, and including amino acids at positions 96 and 97.

10. An isolated peptide comprising the amino acid sequence set forth in a 20 amino acid long segment of SEQ ID NO:363 anywhere from amino acid position 78 to amino acid position 115, and including amino acids at positions 96 and 97.

11. An isolated peptide comprising the amino acid sequence set forth in a member selected from the group consisting of SEQ ID NOs 464, 465-471 and 472.

12. An isolated primer pair, comprising the pair of nucleic acid sequences selected from the group consisting of: SEQ NOs 490 and 491; 41.6 and 417; 419 and 420.

13. An antibody to specifically bind to the amino acid sequence of claim 3.

14. An antibody to specifically bind to the amino acid sequence of claim 4.

15. An antibody to specifically bind to the amino acid sequence of claim 5.

16. A kit for detecting prostate cancer, comprising at least one primer pair of claim 12.

17. A kit for detecting prostate cancer, comprising the antibody of claim 13.

18. The kit of claim 17, wherein said kit further comprises at least one ELISA reagent or at least one Western blot reagent.

19. A kit for detecting prostate cancer, comprising the antibody of claim 14.

20. The kit of claim 19, wherein said kit further comprises at least one ELISA reagent or at least one Western blot reagent.

21. A kit for detecting prostate cancer, comprising the antibody of claim 15.

22. The kit of claim 21, wherein said kit further comprises at least one ELISA reagent or at least one Western blot reagent.

23. A method for detecting prostate cancer, comprising detecting overexpression of the polynucleotide of claim 1 in a sample from a patient.

24. The method of claim 23, wherein said detecting overexpression comprises performing nucleic acid amplification.

25. A method for detecting prostate cancer, comprising detecting overexpression of the polynucleotide of claim 2 in a sample from a patient.

26. The method of claim 25, wherein said detecting overexpression comprises performing nucleic acid amplification.

27. A method for detecting prostate cancer, comprising detecting overexpression of the polypeptide of claim 3 in a sample from a patient.

28. The method of claim 27, wherein said detecting comprises detecting binding of the antibody of claim 13 to the polypeptide of claim 3 in a sample from a patient.

29. A method for detecting prostate cancer, comprising detecting overexpression of the polypeptide of claim 4 in a sample from a patient.

30. The method of claim 29, wherein said detecting comprises detecting binding of the antibody of claim 14 to the polypeptide of claim 4 in a sample from a patient.

31. A method for detecting prostate cancer, comprising detecting overexpression of the polypeptide of claim 5 in a sample from a patient.

32. The method of claim 31, wherein said detecting comprises detecting binding of the antibody of claim 15 to the polypeptide of claim 5 in a sample from a patient.

33. A biomarker to detect prostate cancer, comprising an amino acid sequence of claim 3, marked with a label.

34. A biomarker to detect prostate cancer, comprising an amino acid sequence of claim 4, marked with a label.

35. A biomarker to detect prostate cancer, comprising an amino acid sequence of claim 5, marked with a label.

36. A method to screen for prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 33.

37. A method to screen for prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 34.

38. A method to screen for prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 35.

39. A method to diagnose prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 33.

40. A method to diagnose prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 34.

41. A method to diagnose prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 35.

42. A method for monitoring disease progression, treatment efficacy or relapse of prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 33.

43. A method for monitoring disease progression, treatment efficacy or relapse of prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 34.

44. A method for monitoring disease progression, treatment efficacy or relapse of prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 35.

45. A method of selecting a therapy for prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 33 and selecting a therapy according to said detection.

46. A method of selecting a therapy for prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 34 and selecting a therapy according to said detection.

47. A method of selecting a therapy for prostate cancer, comprising detecting prostate cancer cells with the biomarker of claim 35 and selecting a therapy according to said detection.

Patent History
Publication number: 20080014590
Type: Application
Filed: Jun 19, 2007
Publication Date: Jan 17, 2008
Applicant: COMPUGEN LTD. (Tel Aviv)
Inventors: Dvir Dahary (Tel-Aviv), Sarah Pollock (Tel-Aviv), Zurit Levine (Herzlia), Rotern Sorek (Rechovot), Michal Avalon-Soffer (Ramat-HaSharon), Pinchas Akiva (Ramat-Gan), Amir Toporik (Azur), Osnat Sella-Tavor (Kfar Kish), Shirley Sameah-Greenwald (Kfar-Saba)
Application Number: 11/812,518
Classifications
Current U.S. Class: 435/6.000; 435/7.100; 530/324.000; 530/326.000; 530/328.000; 530/350.000; 530/387.900; 536/23.500; 536/24.330
International Classification: G01N 33/574 (20060101); C07H 21/04 (20060101); C07K 14/82 (20060101); C07K 16/30 (20060101); C12Q 1/68 (20060101);