Methods of diagnosis and prognosis of ovarian cancer

Information

  • Patent Application
  • 20070054268
  • Publication Number
    20070054268
  • Date Filed
    September 05, 2003
    20 years ago
  • Date Published
    March 08, 2007
    17 years ago
Abstract
The present invention provides novel genes and proteins for diagnosing ovarian cancer and/or a likelihood for survival, or recurrence of disease, wherein the expression of the genes and proteins is up-regulated or down-regulated or associated with the occurrence or recurrence of a specific scanner sub-type. The ovarian cancer-associated genes and proteins of the invention are specifically exemplified by the genes and proteins set forth in Tables 1 to 3 and the Sequence Listing.
Description
FIELD OF THE INVENTION

The present invention relates to the identification of nucleic acid and protein expression profiles and nucleic acids, products, and antibodies thereto that are involved in ovarian cancer; and to the use of such expression profiles and compositions in the diagnosis, prognosis and therapy of ovarian cancer. More particularly, this invention relates to novel genes that are expressed at elevated or reduced levels in malignant tissues and uses therefor in the diagnosis of cancer or malignant tumors in human subjects. This Invention also relates to the use of nucleic acid or antibody probes to specifically detect ovarian cancer cells, such as, for example, in the ovarian surface epithelium, wherein over-expression or reduced expression of nucleic acids hybridizing to the probes is highly associated with the occurrence and/or recurrence of an ovarian tumor, and/or the likelihood of patient survival. The diagnostic and prognostic test of the present invention is particularly useful for the early detection of ovarian cancer or metastases thereof, or other cancers, and for monitoring the progress of disease, such as, for example, during remission or following surgery or chemotherapy. The present invention is also directed to methods of therapy wherein the activity of a protein encoded by a diagnostic/prognostic gene described herein is modulated.


BACKGROUND OF THE INVENTION

1. General


As used herein the term “derived from” shall be taken to indicate that a specified integer are obtained from a particular source albeit not necessarily directly from that source.


Unless the context requires otherwise or specifically stated to the contrary, integers, steps, or elements of the invention recited herein as singular integers, steps or elements clearly encompass both singular and plural forms of the recited integers, steps or elements.


The embodiments of the invention described herein with respect to any single embodiment shall be taken to apply mutatis mutandis to any other embodiment of the invention described herein.


Throughout this specification, unless the context requires otherwise, the word “comprise”, or variations such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated step or element or integer or group of steps or elements or integers but not the exclusion of any other step or element or integer or group of elements or integers.


Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations or any two or more of said steps or features.


The present invention is not to be limited in scope by the specific examples described herein. Functionally equivalent products, compositions and methods are clearly within the scope of the invention, as described herein.


The present invention is performed without undue experimentation using, unless otherwise indicated, conventional techniques of molecular biology, microbiology, virology, recombining DNA technology, peptide synthesis in solution, solid phase peptide synthesis, and immunology. Such procedures are described, for example, in the following texts that are incorporated herein by reference:

  • 1. Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratories, New York, Second Edition (1989), whole of Vols I, II, and II;
  • 2. DNA Cloning: A Practical Approach, Vols. I and II (D. N. Glover, ed., 1985), IRL Press, Oxford, whole of text;
  • 3. Oligonucleotide Synthesis: A Practical Approach (M. J. Gait, ed., 1984) IRL Press, Oxford, whole of text, and particularly the papers therein by Gait, pp 1-22; Atkinson et al., pp 35-81; Sproat et al., pp 83-115; and Wu et al., pp 135-151;
  • 4. Nucleic Acid Hybridization: A Practical Approach (B. D. Hames & S. J. Higgins, eds., 1985) IRL Press, Oxford, whole of text;
  • 5. Perbal, B., A Practical Guide to Molecular Cloning (1984);
  • 6. Wunsch, E., ed. (1974) Synthese von Peptiden in Houben-Weyls Metoden der Organischen Chemie (Müler, E., ed.), vol. 15, 4th edn., Parts 1 and 2, Thieme, Stuttgart.
  • 7. Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C. Blackwell, eds., 1986, Blackwell Scientific Publications).


This specification contains nucleotide and amino acid sequence information prepared using Patentin Version 3.1, presented herein after the claims. Each nucleotide sequence is identified in the sequence listing by the numeric indicator <210> followed by the sequence identifier (e.g. <210>1, <210>2, <210>3, etc). The length and type of sequence (DNA, protein (PRT), etc), and source organism for each nucleotide sequence, are indicated by information provided in the numeric indicator fields <211>, <212> and <213>, respectively. Nucleotide sequences referred to in the specification are defined by the term “SEQ ID NO:”, followed by the sequence identifier (eg. SEQ ID NO: 1 refers to the sequence in the sequence listing designated as <400>1).


The designation of nucleotide residues referred to herein are those recommended by the IUPAC-IUB Biochemical Nomenclature Commission, wherein A represents Adenine, C represents Cytosine, G represents Guanine, T represents thymine, Y represents a pyrimidine residue, R represents a purine residue, M represents Adenine or Cytosine, K represents Guanine or Thymine, S represents Guanine or Cytosine, W represents Adenine or Thymine, H represents a nucleotide other than Guanine, B represents a nucleotide other than Adenine, V represents a nucleotide other than Thymine, D represents a nucleotide other than Cytosine and N represents any nucleotide residue.


2. Description of the Related Art


Cancer is a multi-factorial disease and major cause of morbidity in humans and other animals, and deaths resulting from cancer in humans are increasing and expected to surpass deaths from heart disease in future. Carcinomas of the lung, prostate, breast, colon, pancreas, and ovary are major contributing factors to total cancer death in humans. For example, prostate cancer is the fourth most prevalent cancer and the second leading cause of cancer death in males. Similarly, cancer of the ovary is the second most common cancer of the female reproductive organs and the fourth most common cause of cancer death among females. With few exceptions, metastatic disease from carcinoma is fatal. Even if patients survive their primary cancers, recurrence or metastases are common.


It is widely recognized that simple and rapid tests for solid cancers or tumors have considerable clinical potential. Not only can such tests be used for the early diagnosis of cancer but they also allow the detection of tumor recurrence following surgery and chemotherapy. A number of cancer-specific blood tests have been developed which depend upon the detection of tumor-specific antigens in the circulation (Catalona, W. J., et al., 1991, “Measurement of prostate-specific antigen in serum as a screening test for prostate cancer”, N. Engl. J. Med. 324, 1156-1161; Barrenetxea, G., et al., 1998, “Use of serum tumor markers for the diagnosis and follow-up of breast cancer”, Oncology, 55, 447-449; Cairns, P., and Sidreansky, D., 1999, “Molecular methods for the diagnosis of cancer”. Biochim. Biophys. Acta. 1423, C 11-C 18).


Ovarian cancer is the fourth most frequent cause of cancer death in females and in the United States, and accounts for approximately 13,000 deaths annually. Furthermore, ovarian cancer remains the number one killer of women with gynaecological malignant hyperplasia and the incidence is rising in industrialized countries. The etiology of the neoplastic transformation remains unknown although there is epidemiological evidence for an association with disordered endocrine function. The incidence of ovarian carcinoma is higher in nulliparous females and in those with early menopause.


Most ovarian cancers are thought to arise from the ovarian surface of epithelium (OSE). Epithelial ovarian cancer is seldom encountered in women less than 35 years of age. Its incidence increases sharply with advancing age and peaks at ages 75 to 80, with the median age being 60 years. The single most important known risk factor is a strong familial history of breast or ovarian cancer. To date, little is known about the structure and function of the OSE cells. It is known that the OSE is highly dynamic tissue that undergoes morphogenic changes, and has proliferative properties sufficient to cover the ovulatory site following ovulation. Morphological and histochemical studies suggest that the OSE has secretory, endocytotic and transport functions which are hormonally-controlled (Blaustein and Lee, Oncol. 8, 34-43, 1979; Nicosia and Johnson, Int J. Gynecol. Pathol., 3, 249-260, 1983; Papadaki and Beilby, J. Cell Sci. 8, 445-464, 1971; Anderson et al., J. Morphol., 150, 135-164, 1976).


Ovarian cancers are not readily detectable by diagnostic techniques (Siemens et al., J. Cell. Physiol., 134: 347-356, 1988). In fact, the diagnosis of carcinoma of the ovary is generally only possible when the disease has progressed to a late stage of development. Approximately 75% of women diagnosed with ovarian cancer are already at an advanced stage (III and IV) of the disease at their initial diagnosis. During the past 20 years, neither diagnosis nor five year survival rates have greatly improved for these patients. This is substantially due to the high percentage of high-stage initial detection of the disease. There is therefore a need to develop new markers that improve early diagnosis and thereby reduce the percentage of high-stage initial diagnoses.


A number of proteinaceous ovarian tumor markers were evaluated several years ago, however these were found to be non-specific, and determined to be of low value as markers for primary ovarian cancer (Kudlacek et al., Gyn. One. 35, 323-329, 1989; Rustin et al., J. Clin. One., 7, 1667-1671, 1989; Sevelda et al., Am. J. Obstet. Gynecol., 161, 1213-1216, 1989; Omar et al., Tumor Biol., 10, 316-323, 1989). Several monoclonal antibodies were also shown to react with ovarian tumor associated antigens, however they were not specific for ovarian cancer and merely recognize determinants associated with high molecular weight mucin-like glycoproteins (Kenemans et al., Eur. J. Obstet Gynecol. Repod. Biol, 29, 207-218, 1989; McDuffy, Ann. Clin. Biochem., 26, 379-387, 1989). More recently, oncogenes associated with ovarian cancers have been identified, including HER-21neu (c-erbB-2) which is over-expressed in one-third of ovarian cancers (U.S. Pat. No. 6,075,122 by Cheever et al, issued Jun. 13, 2000), the fms oncogene, and abnormalities in the p53 gene, which are seen in about half of ovarian cancers.


Whilst previously identified markers for carcinomas of the ovary have facilitated efforts to diagnose and treat these serious diseases, there is a clear need for the identification of additional markers and therapeutic targets. The identification of tumor markers that are amenable to the early-stage detection of localized tumors is critical for more effective management of carcinomas of the ovary.


SUMMARY OF THE INVENTION

In work leading up to the present invention, the inventors sought to identify nucleic acid markers that were diagnostic of ovarian cancers generally, or diagnostic of specific ovarian cancers such as, for example, serous ovarian cancer (SOC), mucinous ovarian cancer (MOC), non-invasive (borderline ovarian cancer or low malignant potential ovarian cancer), mixed phenotype ovarian cancer, endometrioid ovarian cancer (EnOC) and clear cell ovarian cancer (ClCA), papillary serous ovarian cancer, Brenner cell or undifferentiated adenocarcinoma, by virtue of their modulated expression in cancer tissues derived from a patient cohort compared to their expression in healthy or non-cancerous cells and tissues. Additionally, the inventors sought to determine whether any correlation exists between the expression of any particular gene in a subject having ovarian cancer and the survival, or likelihood for survival, of the subject during the medium to long term (i.e. in the period between about 1-2 years from primary diagnosis, or longer). The inventors also sought to to determine whether any correlation exists between the expression of any particular gene in a subject following treatment for ovarian cancer and the recurrence, or likelihood for recurrence, of ovarian cancer in the subject during the medium to long term (i.e. in the period between about 1-2 years from primary diagnosis, or longer).


As exemplified herein, the inventors identified a number of genes whose expression is altered (up-regulated or down-regulated) in individuals with ovarian cancer compared to healthy individuals, eg., subjects who do not have ovarian cancer. The particular genes are identified in Tables 1 and 2. Preferably, the genes are selected from the group of candidate genes set forth in Table 3.


The list of genes and proteins exemplified herein by Table 1 were identified by a statistical analysis as outlined in the examples which gave a P-value, eg., by comparison of expression to the expression of that gene in normal ovaries.


Accordingly, one aspect of the present invention provides a method of detecting an ovarian cancer-associated transcript in a biological sample, the method comprising contacting the biological sample with a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Table 1 or 2 or 3. Preferably the percentage identity to a sequence disclosed in any one of Tables 1-3 is at least about 85% or 90% or 95%, and still more preferably at least about 98% or 99%.


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a modified level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 47, 49, 51, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a modified level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 47, 49, 51, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


Even more preferably, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a modified level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 5, 7, 9, 11, 13, 15, 17, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 45, 46, 48, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 5, 7, 9, 11, 13, 15, 17, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 45, 46, 48, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 5, 7, 9, 11, 13, 15, 17, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 45, 46, 48, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 6, 8, 10, 12, 14, 16, 18, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 47, 49, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84; and
  • (v) a sequence that is complementary to (i) or (ii) or (iii) or (iv).


As used herein, the term “modified level” includes an enhanced, increased or elevated level of an integer being assayed, or alternatively, a reduced or decreased level of an integer being assayed.


In one embodiment an elevated, enhanced or increased level of expression of the nucleic acid is detected. In accordance with this embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an enhanced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii);
  • (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an enhanced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 7, 9, 11, 13, 15, 17, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 45, 46, 48, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 7, 9, 11, 13, 15, 17, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 45, 46, 48, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 7, 9, 11, 13, 15, 17, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 45, 46, 48, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 8, 10, 12, 14, 16, 18, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 47, 49, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In an alternative preferred embodiment, a reduced level of a diagnostic marker is indicative of ovarian cancer. In accordance with this embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a reduced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii);
  • (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a reduced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, and 5;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, and 5;
  • (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 3, and 5;
  • (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, and 6; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


Preferably, the ovarian cancer that is diagnosed according to the present invention is an epithelial ovarian cancer, such as, for example, serous ovarian cancer, non-invasive ovarian cancer, mixed phenotype ovarian cancer, mucinous ovarian cancer, endometrioid ovarian cancer, clear cell ovarian cancer, papillary serous ovarian cancer, Brenner cell or undifferentiated adenocarcinoma. As will be apparent from the preferred embodiments described below, certain of the genes represented in Table 1, Table 2 and Table 3 are expressed at modified levels in subjects having serous or mucinous ovarian cancers. Data presented in FIGS. 1-4 also exemplify novel diagnostics and prognostics for serous ovarian cancer, mucinous ovarian cancer, endometrioid ovarian cancer and clear cell ovarian cancer.


As exemplified herein by Table 2, the present inventors have identified those genes having an elevated or reduced average ratio of expression of specific genes between ovarian cancer patients vs non-ovarian cancer patients, wherein a high ratio in Table 2 indicates an enhanced expression in an ovarian cancer patients and wherein a negative ratio indicates that a reduced expression in an ovarian cancer patient.


In an alternative preferred embodiment, the present invention provides a method of diagnosing a serous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a modified level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a serous ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, and AB018305;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii);
  • (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In a further alternative preferred embodiment, the present invention provides a method of diagnosing a mucinous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a mucinous ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM006149, AA315933, U47732, NM005588, AW503395, NM004063, AI073913, AI928445, NM022454, W40460, AA132961 and AF111856;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM006149, AA315933, U47732, NM005588, AW503395, NM004063, AI073913, AI928445, NM022454, W40460, AA132961 and AF111856;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii);
  • (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM006149, AA315933, U47732, NM005588, AW503395, NM004063, AI073913, AI928445, NM022454, W40460, AA132961 and AF1111856; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In a preferred embodiment, the present invention provides a method of diagnosing a mucinous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an enhanced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from SEQ ID NO: 57 or 59 or 61;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from SEQ ID NO: 57 or 59 or 61;
  • (iii) a sequence that is at least about 80% identical to SEQ ID NO: 57 or 59 or 61;
  • (iv) a sequence that encodes the amino acid sequence set forth in SEQ ID NO: 58 or 60 or 62; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


Those skilled in the art will be aware that as a carcinoma progresses, metastases occur in organs and tissues outside the site of the primary tumor. For example, in the case of ovarian cancer, metastases commonly appear in a tissue selected from the group consisting of omentum, abdominal fluid, lymph nodes, lung, liver, brain, and bone. Accordingly, the term “ovarian cancer” as used herein shall be taken to include an early or developed tumor of the ovary, such as, for example, any one or more of a number of cancers of epithelial origin, such as serous, mucinous, endometrioid, clear cell, papillary serous, Brenner cell or undifferentiated adenocarcinoma, non-invasive ovarian cancer such as borderline ovarian cancer or low-malignant potential ovarian cancer, or a mixed phenotype ovarian cancer, and optionally, any metastases outside the ovary that occurs in a subject having a primary tumor of the ovary.


As used herein, the term “diagnosis”, and variants thereof, such as, but not limited to “diagnose”, “diagnosed” or “diagnosing” shall not be limited to a primary diagnosis of a clinical state, however should be taken to include any primary diagnosis or prognosis of a clinical state. For example, the “diagnostic assay” formats described herein are equally relevant to assessing the remission of a patient, or monitoring disease recurrence, or tumor recurrence, such as following surgery or chemotherapy, or determining the appearance of metastases of a primary tumor. All such uses of the assays described herein are encompassed by the present invention.


Both classical hybridization and amplification formats, and combinations thereof, are encompassed by the invention. In one embodiment, the hybridization comprises performing a nucleic acid hybridization reaction between a labeled probe and a second nucleic acid in the biological sample from the subject being tested, and detecting the label. In another embodiment, the hybridization comprising performing a nucleic acid amplification reaction eg., polymerase chain reaction (PCR), wherein the probe consists of a nucleic acid primer and nucleic acid copies of the nucleic acid in the biological sample are amplified. As will be known to the skilled artisan, amplification may proceed classical nucleic acid hybridization detection systems, to enhance specificity of detection, particularly in the case of less abundant mRNA species in the sample.


In a preferred embodiment, the polynucleotide is immobilised on a solid surface.


The present invention clearly encompasses nucleic acid-based methods and protein-based methods for diagnosing cancer in humans and other mammals.


Accordingly, in a related embodiment, the present invention provides a method of detecting an ovarian cancer-associated polypeptide in a biological sample the method comprising contacting the biological sample with an antibody that binds specifically to an ovarian cancer-associated polypeptide in the biological sample, the polypeptide being encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3.


Preferably the percentage identity to a sequence disclosed in any one of Tables 1-3 is at least about 85% or 90% or 95%, and still more preferably at least about 98% or 99%.


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a modified level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to a sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 47, 49, 51, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84.


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a modified level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to a sequence selected from the group consisting of SEQ ID NOs: 2, 6, 8, 10, 12, 14, 16, 18, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 47, 49, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84.


In one embodiment an elevated, enhanced or increased level of expression of the antigen-antibody complex is detected. In accordance with this embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200.


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to a sequence selected from the group consisting of SEQ ID NOs: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 47, 49, 51, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84.


In an alternative preferred embodiment, a reduced level of a diagnostic marker is indicative of ovarian cancer. In accordance with this embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a reduced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM022117, NM005460, NM002387, AI745249 and AI694200.


In a preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a reduced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to a sequence selected from the group consisting of SEQ ID NOs: 2, 4, and 6.


Preferably, the ovarian cancer that is diagnosed according to the present invention is an epithelial ovarian cancer, such as, for example, serous ovarian cancer or mucinous ovarian cancer.


In an alternative preferred embodiment, the present invention provides a method of diagnosing a serous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a modified level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a serous ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305.


In a further alternative preferred embodiment, the present invention provides a method of diagnosing a mucinous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a reduced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a mucinous ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM006149, AA315933, U47732, NM005588, AW503395, NM004063, AI073913, AI928445, NM022454, W40460, AA132961 and AF111856.


In a preferred embodiment, the present invention provides a method of diagnosing a mucinous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a mucinous ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to SEQ ID NO: 58 or 60 or 62.


In a further related embodiment, the present invention provides a method of detecting an ovarian cancer-associated antibody in a biological sample the method comprising contacting the biological sample with a polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3, wherein the polypeptide specifically binds to the ovarian cancer-associated antibody.


Preferably, in the above methods, the biological sample is contacted with a plurality of the polynucleotides, polypeptides or antibodies referred to above.


In a particularly preferred embodiment, the present invention provides an antibody-based mulptiplex assay for determining the likelihood of survival of a subject from an ovarian cancer. In one embodiment, the invention provides a method of determining the likelihood of survival of a subject suffering from a serous ovarian cancer, said method comprising contacting a biological sample from said subject being tested with at least two antibodies for a time and under conditions sufficient for antigen-antibody complexes to form and then detecting the complexes wherein an enhanced level of the antigen-antibody complexes for the subject being tested compared to the amount of the antigen-antibody complexes formed for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein one antibody binds to an sFRP polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 72 and wherein one antibody binds to a SOCS3 polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 74.


The present invention is not to be limited by the source or nature of the biological sample. In one embodiment, the biological sample is from a patient undergoing a therapeutic regimen to treat ovarian cancer. In an alternative preferred embodiment, the biological sample is from a patient suspected of having ovarian cancer.


In addition to providing up-regulated and down-regulated genes, the list of genes and proteins exemplified herein by Table 1 were identified by a statistical analysis as outlined in the examples which gave a P-value, eg., by comparison of expression to clinicopathological parameters for disease recurrence or patent survival. Accordingly, the present invention is particularly useful for prognostic applications, in particular for assessing the medium-to-long term survival of a subject having an ovarian cancer, or alternatively or in addition, for assessing the likelihood of disease recurrence.


Accordingly, a further aspect of the present invention provides a method of monitoring the efficacy of a therapeutic treatment of ovarian cancer, the method comprising:

    • (i) providing a biological sample from a patient undergoing the therapeutic treatment; and
    • (ii) determining the level of a ovarian cancer-associated transcript in the biological sample by contacting the biological sample with a polynucleotide that selectively hybridizes to a sequence having at least about 80% identity to a sequence as shown in any one of Tables 1-3, thereby monitoring the efficacy of the therapy.


Preferably the method further comprises comparing the level of the ovarian cancer-associated transcript to a level of the ovarian cancer-associated transcript in a biological sample from the patient prior to, or earlier in, the therapeutic treatment.


In a related embodiment, the present invention provides a method of monitoring the efficacy of a therapeutic treatment of ovarian cancer, the method comprising:

    • (i) providing a biological sample from a patient undergoing the therapeutic treatment; and
    • (ii) determining the level of a ovarian cancer-associated antibody in the biological sample by contacting the biological sample with a polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3, wherein the polypeptide specifically binds to the ovarian cancer-associated antibody, thereby monitoring the efficacy of the therapy.


Preferably the method further comprises comparing the level of the ovarian cancer-associated antibody to a level of the ovarian cancer-associated antibody in a biological sample from the patient prior to, or earlier in, the therapeutic treatment.


In a further related embodiment, the present invention provides a method of monitoring the efficacy of a therapeutic treatment of ovarian cancer, the method comprising:

    • (i) providing a biological sample from a patient undergoing the therapeutic treatment; and
    • (ii) determining the level of a ovarian cancer-associated polypeptide in the biological sample by contacting the biological sample with an antibody, wherein the antibody specifically binds to a polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3, thereby monitoring the efficacy of the therapy.


Preferably the method further comprises comparing the level of the ovarian cancer-associated polypeptide to a level of the ovarian cancer-associated polypeptide in a biological sample from the patient prior to, or earlier in, the therapeutic treatment.


It will also be apparent from the following preferred embodiments, that the expression of certain genes listed in Table 1 and Table 3 is statistically correlated with survival and death of patients having ovarian cancer, wherein a low P value indicates an enhanced likelihood that a patient having altered expression of the gene will die from the cancer.


Accordingly, in one embodiment, the present invention provides a method of determining the likelihood of survival of a subject suffering from an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM003014, AA046217, NM015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI1796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI1798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM002776, BE261944, NM006379, AI002238, X81789, NM002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM001955, AI680737, AI752666, AA505445, BE246649, and NM003955;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM003014, AA046217, NM015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM002776, BE261944, NM006379, AI002238, X81789, NM002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM001955, AI680737, AI752666, AA505445, BE246649, and NM003955;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii);
  • (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM003014, AA046217, NM015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM002776, BE261944, NM006379, AI002238, X81789, NM002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM001955, AI680737, AI752666, AA505445, BE246649, and NM003955; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In a preferred embodiment, the present invention provides a method of determining the likelihood of survival of a subject suffering from an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83;
  • (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84; and
  • (v) a sequence that is complementary to (i) or (ii) or (iii) or (iv).


In an alternative preferred embodiment, the present invention provides a method of determining the likelihood of survival of a subject suffering from an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has has a poor probability of survival, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM003014, AA046217, NM 015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM002776, BE261944, NM006379, AI002238, X81789, NM002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM001955, AI680737, AI752666, AA505445, BE246649, and NM003955.


In an alternative preferred embodiment, the present invention provides a method of determining the likelihood of survival of a subject suffering from an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has has a poor probability of survival, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to a sequence selected from the group consisting of SEQ ID NOs: 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84.


In a particularly preferred embodiment, the present invention provides a marker for determining the likelihood of a subject surviving from serous cancer. In accordance with this embodiment of the invention, there is provided a method of determining the likelihood of survival of a subject suffering from a serous ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid comprising the nucleotide sequence set forth in SEQ ID NO: 71 or 73;
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid comprising the nucleotide sequence set forth in SEQ ID NO: 71 or 73;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii) and encoding an sFRP protein or a SOCS3 protein;
  • (iv) a sequence that encodes a polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 72 or 74; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In an alternative preferred embodiment, the present invention provides a method of determining the likelihood of survival of a subject suffering from a serous ovarian cancer, said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said antibody binds to an sFRP polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 72 or a SOCS3 polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 74 or.


It will also be apparent from the following preferred embodiments, that the expression of certain genes listed in Table 1 and Table 3 is statistically correlated with recurrence of ovarian cancer, wherein a low P value indicates an enhanced likelihood that a patent having altered expression of the gene will experience recurrence of the disease.


In yet another preferred embodiment, the present invention provides a method of determining the likelihood that a subject will suffer from a recurrence of an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a high probability of recurrence, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of:

  • (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM012317, NM000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, AA569756, AW138190, AF126245, L10343, NM002514, AI863735, NM005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N51357, N80486, NM000954, NM005756, NM016652, R26584, R31178, W05391, W25005, W45393, W68815, X65965, X76732 and Z45051,
  • (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM012317, NM 000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, AA569756, AW138190, AF126245, L10343, NM002514, AI186373, NM005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N513571 N80486, NM000954, NM005756, NM016652, R26584, R31178, W05391, W25005, W45393, W68815, X65965, X76732 and Z45051;
  • (iii) a sequence that is at least about 80% identical to (i) or (ii);
  • (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM012317, NM000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, AA569756, AW138190, AF126245, L10343, NM002514, AI863735, NM005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N51357, N80486, NM000954, NM005756, NM016652, R26584, R31178, W05391, W25005, W45393, W68815, X65965, X76732 and Z45051; and
  • (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).


In an alternative preferred embodiment, the present invention provides a method of determining the likelihood that a subject will suffer from a recurrence of an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a high probability of recurrence, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM012317, NM000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, M569756, AW138190, AF126245, L10343, NM002514, AI863735, NM005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N51357, N80486, NM 000954, NM005756, NM016652, R26584, R31178, W05391, W25005, W45393, W68815, X65965, X76732 and Z45051.


The recurrence of ovarian cancer is a clinical recurrence as determined by the presence of one or more clinical symptoms of an ovarian cancer, such as, for example, a metastases, or alternatively, as determined in a biochemical test, immunological test or serological test such as, for example, a cross-reactivity in a biological sample to a CA125 antibody.


Preferably, the recurrence is capable of being detected at least about 2 years from treatment, more preferably about 2-3 years from treatment, and even more preferably about 4 or 5 or 10 years from treatment.


Preferably, in the above diagnostic and/or prognostic methods, the biological sample is contacted with a plurality of the nucleic acids and/or polypeptides and/or antibodies referred to above. In a particularly preferred embodiment, mulpiplex assays are performed to detect enhanced expression at least of sFRP4 and SOC3 at the protein level (eg., using antigen-based or antibody-based assays) or at the mRNA level (eg., by detecting elevated levels of mRNA transcripts).


A further embodiment of the present invention provides a method of diagnosing epithelial ovarian cancer by detecting aberrant methylation of a promoter that regulates expression of a tumor suppressor gene eg., MCC. In particular, the present invention contemplates the detection of hypermethylation of the promoter of a tumor suppressor gene. Without being bound by any theory or mode of action, such hypermethylation leads to gene inactivation, thereby reducing expression for the tumor suppressor gene and permitting oncogenesis. In one preferred embodiment, the present invention provides a method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising determining aberrant methylation in a promoter sequence that regulates expression of a tumor suppressor gene in a biological sample from said subject compared to the methylation of the promoter in nucleic acid obtained for a control subject not having ovarian cancer wherein said aberrant methylation indicates that the subject being tested has an ovarian ovarian cancer.


In a further aspect, the present invention provides a method for identifying a compound that modulates an ovarian cancer-associated polypeptide, the method comprising:

    • (i) contacting the compound with a ovarian cancer-associated polypeptide, the polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3; and
    • (ii) determining the functional effect of the compound upon the polypeptide.


The functional effect may, for example, be a physical effect or a chemical effect. In one embodiment, the functional effect is determined by measuring ligand binding to the polypeptide. In a particular embodiment, the polypeptide is expressed in a eukaryotic host cell or cell membrane. Preferably the polypeptide is recombinant.


In another aspect, the present invention provides a method of inhibiting proliferation of a ovarian tumour cell, which method comprises contacting said cell with a compound identified using the method supra for identifying a compound that modulates an ovarian cancer-associated polypeptide.


In a further aspect, the present invention provides a method of inhibiting proliferation of a ovarian cancer-associated cell to treat ovarian cancer in a patient, the method comprising the step of administering to the patient a therapeutically effective amount of a compound identified using the method supra for identifying a compound that modulates an ovarian cancer-associated polypeptide.


In a further aspect, the present invention provides a drug screening assay comprising:

    • (i) administering a test compound to a mammal having ovarian cancer or a cell isolated therefrom;
    • (ii) comparing the level of gene expression of a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3 in a treated cell or mammal with the level of gene expression of the polynucleotide in a control cell or mammal, wherein a test compound that modulates the level of expression of the polynucleotide is a candidate for the treatment of ovarian cancer.


Typically, the control is a mammal with ovarian cancer or a cell therefrom that has not been treated with the test compound. Alternatively, the control is a normal cell or mammal.


The present invention also provides a method for treating a mammal having ovarian cancer comprising administering a compound identified the drug screening method supra.


In a further aspect, the present invention provides a pharmaceutical composition for use in treating a mammal having ovarian cancer, the composition comprising a compound identified the screening method supra for identifying a compound that modulates an ovarian cancer-associated polypeptide, or alternatively, using the drug screening method supra, and a physiologically acceptable carrier or diluent.


In a further aspect, the present invention provides an assay device, preferably for use in the diagnosis or prognosis of ovarian cancer, said device comprising a plurality of polynucleotides immobilized to a solid phase, wherein each of said polynucleotides consists of a gene as listed in any one of Tables 1-3. Preferably, the solid phase is a substantially planar chip.


In a related embodiment, the present invention provides an assay device, preferably for use in the diagnosis or prognosis of ovarian cancer, said device comprising a plurality of different antibodies immobilized to a solid phase, wherein each of said antibodies binds to a polypeptide listed in Tables 1-3. Preferably, the solid phase is a substantially planar chip.


Preferably, the assay device supra is used in a method of diagnosis or prognosis as described herein.


Alternatively, the assay device is used to identify modulatory compounds of the expression of one or more genes/proteins listed in any one of Tables 1-3.


In a further aspect, the present invention provides a non-human transgenic animal which is transgenic by virtue of comprising a gene set forth in any one of Tables 1-3 and, in particular, to the use of any such transgenic animal in the performance of a diagnostic or prognostic method of the invention as transgenic “knock-out” animals that have disrupted expression of a gene as set forth in any one of Tables 1-3.


In a further aspect, the present invention provides an isolated polynucleotide selected from the group consisting of;

    • (a) polynucleotides comprising a nucleotide sequence as shown in Tables 1-3, or the complement thereof;
    • (b) polynucleotides comprising a nucleotide sequence capable of selectively hybridizing to a nucleotide sequence as shown in Tables 1-3;
    • (c) polynucleotides comprising a nucleotide sequence capable of selectively hybridizing to the complement of a nucleotide sequence as shown in Tables 1-3; and
    • (d) polynucleotides comprising a polynucleotide sequence which is degenerate as a result of the genetic code to the polynucleotides defined in (a), (b) or (c).


Preferred polynucleotides comprise a polynucleotide sequence as shown in Tables 1-3 or a sequence having at least 80% homology thereto.


Preferably, the isolated polynucleotide is used for the diagnosis or prognosis of ovarian cancer, more preferably by a method as described herein. In a particularly preferred embodiment, the present invention provides for the use of a polynucleotide as set forth in any one of Tables 1-3 in the diagnosis or prognosis of ovarian cancer or for the preparation of a medicament for the treatment of ovarian cancer.


The present invention also provides a nucleic acid vector comprising a polynucleotide of the Invention. In one embodiment, the polynucleotide is operably linked to a regulatory control sequence capable of directing expression of the polynucleotide in a host cell. In a particularly preferred embodiment, the present invention provides for the use of a vector comprising a polynucleotide as set forth in any one of Tables 1-3 In the diagnosis or prognosis of ovarian cancer or for the preparation of a medicament for the treatment of ovarian cancer.


The present invention further provides a host cell comprising a vector as described in the preceding paragraph. In a particularly preferred embodiment, the present invention provides for the use of a host cell comprising an introduced polynucleotide as set forth in any one of Tables 1-3 in the diagnosis or prognosis of ovarian cancer or for the preparation of a medicament for the treatment of ovarian cancer.


In a further aspect, the present invention provides an isolated polypeptide which is encoded by a gene set forth in any one of Tables 1-3. The present invention also provides an isolated polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3. In a particularly preferred embodiment, the present invention provides for the use of an isolated polypeptide as set forth in any one of Tables 1-3 in the diagnosis or prognosis of ovarian cancer or for the preparation of a medicament for the treatment of ovarian cancer.


In a further aspect the present invention provides an antibody that binds specifically a polypeptide listed in Tables 1-3. In a particularly preferred embodiment, the present invention provides for the use of an antibody that binds to an isolated polypeptide as set forth in any one of Tables 1-3 in the diagnosis or prognosis of ovarian cancer or for the preparation of a medicament for the treatment of ovarian cancer.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a photographic representation showing expression of genes as identified by immunohistochemical staining of fixed normal (i.e. non-cancerous or healthy) tissues (panel A) or ovarian cancer tissue (panel B). The inset in panel A shows inclusion cysts. The expression levels of the following genes listed in Table 1 or Table 3 were determined: Claudin-3 (SEQ ID NO: 15); EP-CAM (Accession No. NM002354); and SOX17 (SEQ ID NO: 17). Positive controls CA125, MUC-1 and E-Cadherin were also included for comparison.



FIG. 2 is a graphical representation showing the correlation between expression of different genes in serous ovarian cancer (SOC), mucinous ovarian cancer (MOC), endometroid ovarian cancer (EnOC) and clear cell ovarian cancer (CICA). Genes indicated on the x-axis in each case are as in the legend to FIG. 1.



FIG. 3 is a copy of a photographic representation showing immunohistochemical staining of ovary tissue from a normal healthy subject (normal ovary), a subject diagnosed with mucinous ovarian cancer (MOC) and a subject diagnosed with serous ovarian cancer (SOC), following staining with probes that are specific for L1-Cadherin (top row), meprin alpha (middle row) or galectin-4 (lower row). Magnification is indicated as 20-40×.



FIG. 4
a is a copy of a photographic representation showing immunohistochemical staining of samples from a normal healthy subject (normal) or primary serous ovarian tumor (SOC), following staining with probes that are specific for sFRP4 (top row), or SOCS3 (lower row). Magnification is indicated as 20×.



FIG. 4
b is a copy of a graphical representation showing a Kaplan-Meier survival curve correlating sFRP4 expression to patient survival over the medium term (i.e., from about 12 months to about 48 months) to long term (more than about 48 months), indicating that high expression of sFRP4 is associated with poor survival in patients (n=127) having SOC (p=0.0056).




DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

Ovarian Cancer-Associated Sequences


Ovarian cancer-associated sequences can include both nucleic acid (i.e., “ovarian cancer-associated genes”) and protein (i.e., “ovarian cancer-associated proteins”).


As used herein, the term “ovarian cancer-associated protein” shall be taken to mean any protein that has an expression pattern correlated to an ovarian cancer, the recurrence of an ovarian cancer or the survival of a subject suffering from ovarian cancer.


Similarly, the term “ovarian cancer-associated gene” shall be taken to mean any nucleic acid encoding an ovarian cancer-associated protein or nucleic acid having an expression profile that is correlated to an ovarian cancer, the recurrence of an ovarian cancer or the survival of a subject suffering from ovarian cancer.


As will be appreciated by those in the art and is more fully outlined below, ovarian cancer-associated genes are useful in a variety of applications, including diagnostic applications, which will detect naturally occurring nucleic acids, as well as screening applications; e.g., biochips comprising nucleic acid probes or PCR microtitre plates with selected probes to the ovarian cancer sequences are generated.


For identifying ovarian cancer-associated sequences, the ovarian cancer screen typically includes comparing genes identified in different tissues, e.g., normal and cancerous tissues, or tumour tissue samples from patients who have metastatic disease vs. non metastatic tissue. Other suitable tissue comparisons include comparing ovarian cancer samples with metastatic cancer samples from other cancers, such as lung, breast, gastrointestinal cancers, ovarian, etc. Samples of different stages of ovarian cancer, e.g., survivor tissue, drug resistant states, and tissue undergoing metastasis, are applied to biochips comprising nucleic acid probes. The samples are first microdissected, if applicable, and treated as is known in the art for the preparation of mRNA. Suitable biochips are commercially available, e.g. from Affymetrix. Gene expression profiles as described herein are generated and the data analyzed.


In one embodiment, the genes showing changes in expression as between normal and disease states are compared to genes expressed in other normal tissues, preferably normal ovarian, but also Including, and not limited to lung, heart, brain, liver, breast, kidney, muscle, colon, small intestine, large intestine, spleen, bone and placenta. In a preferred embodiment, those genes identified during the ovarian cancer screen that are expressed in any significant amount in other tissues are removed from the profile, although in some embodiments, this is not necessary. That is, when screening for drugs, it is usually preferable that the target be disease specific, to minimise possible side effects.


In a preferred embodiment, ovarian cancer-associated sequences are those that are up-regulated in ovarian cancer; that is, the expression of these genes is modified (up-regulated or down-regulated) in ovarian cancer tissue as compared to non-cancerous tissue (see Table 1).


“Up-regulation” as used herein means at least about a two-fold change, preferably at least about a three fold change, with at least about five-fold or higher being preferred. All Unigene cluster identification numbers and accession numbers herein are for the GenBank sequence database and the sequences of the accession numbers are hereby expressly incorporated by reference. Sequences are also available in other databases, e.g., European Molecular Biology Laboratory (EMBL) and DNA Database of Japan (DDBJ).


“Down-regulation” as used herein often means at least about a 1.5-fold change more preferably a two-fold change, preferably at least about a three fold change, with at least about five-fold or higher being most preferred.


Particularly preferred sequences are those referred to in Tables 1 or 3 that have a P value of less than 0.05, more preferably a P value of less than about 0.01.


Similarly, preferred sequences are those referred to in Table 2 as having an absolute ratio of expression between ovarian patients and normal patients of at least about ±5.0, more preferably at least about ±6.0 even more preferrably at least about ±7.0 or at least about ±8.0 or at least about ±9.0 or at least about ±0.0.


Detection of Ovarian Cancer Sequences for Diagnostic/Prognostic Applications


In one aspect, the RNA expression levels of genes are determined for different cellular states in the ovarian cancer phenotype. Expression levels of genes in normal tissue (i.e., not undergoing ovarian cancer) and in ovarian cancer tissue (and in some cases, for varying severities of ovarian cancer that relate to prognosis, as outlined below) are evaluated to provide expression profiles. An expression profile of a particular cell state or point of development is essentially a “fingerprint” of the state. While two states may have any particular gene similarly expressed, the evaluation of a number of genes simultaneously allows the generation of a gene expression profile that is reflective of the state of the cell. By comparing expression profiles of cells in different states, information regarding which genes are important (including both up- and down-regulation of genes) in each of these states is obtained. Then, diagnosis are performed or confirmed to determine whether a tissue sample has the gene expression profile of normal or cancerous tissue. This will provide for molecular diagnosis of related conditions.


“Differential expression,” or grammatical equivalents as used herein, refers to qualitative or quantitative differences in the temporal and/or cellular gene expression patterns within and among cells and tissue. Thus, a differentially expressed gene can qualitatively have its expression altered, including an activation or inactivation, in, e.g., normal versus ovarian cancer tissue. Genes are turned on or turned off in a particular state, relative to another state thus permitting comparison of two or more states. A qualitatively regulated gene will exhibit an expression pattern within a state or cell type which is detectable by standard techniques. Some genes will be expressed in one state or cell type, but not in both. Alternatively, the difference in expression are quantitative, e.g., in that expression is increased or decreased; i.e., gene expression is either upregulated, resulting in an increased amount of transcript, or downregulated, resulting in a decreased amount of transcript. The degree to which expression differs need only be large enough to quantify via standard characterization techniques as outlined below, such as by use of Affymetrix GeneChip™ expression arrays, Lockhart, Nature Biotechnology 14:1675-1680 (1996), hereby expressly incorporated by reference. Other techniques include, but are not limited to, quantitative reverse transcriptase PCR, northern analysis and RNase protection. As outlined above, preferably the change in expression (i.e., upregulation or downregulation) is at least about 50%, more preferably at least about 100%, more preferably at least about 150%, more preferably at least about 200%, with from 300 to at least 1000% being especially preferred.


Evaluation are at the gene transcript, or the protein level. The amount of gene expression are monitored using nucleic acid probes to the DNA or RNA equivalent of the gene transcript, and the quantification of gene expression levels, or, alternatively, the final gene product itself (protein) are monitored, e.g., with antibodies to the ovarian cancer-associated protein and standard immunoassays (ELISAS, etc.) or other techniques, including mass spectroscopy assays, 2D gel electrophoresis assays, etc. Proteins corresponding to ovarian cancer genes, i.e., those identified as being important in a ovarian cancer phenotype, are evaluated in a ovarian cancer diagnostic test.


In a preferred embodiment, gene expression monitoring is performed on a plurality of genes. Multiple protein expression monitoring are performed as well. Similarly, these assays are performed on an individual basis as well.


In this embodiment, the ovarian cancer nucleic acid probes are attached to biochips as outlined herein for the detection and quantification of ovarian cancer sequences in a particular cell. The assays are further described below in the example. PCR techniques are used to provide greater sensitivity.


In a preferred embodiment nucleic acids encoding the ovarian cancer-associated protein are detected. Although DNA or RNA encoding the ovarian cancer-associated protein are detected, of particular interest are methods wherein an mRNA encoding a ovarian cancer-associated protein is detected. Probes to detect mRNA are a nucleotide/deoxynucleotide probe that is complementary to and hybridizes with the mRNA and includes, but is not limited to, oligonucleotides, cDNA or RNA. Probes also should contain a detectable label, as defined herein. In one method the mRNA is detected after immobilizing the nucleic acid to be examined on a solid support such as nylon membranes and hybridizing the probe with the sample. Following washing to remove the non-specifically bound probe, the label is detected. In another method detection of the mRNA is performed in situ. In this method permeabilized cells or tissue samples are contacted with a detectably labeled nucleic acid probe for sufficient time to allow the probe to hybridize with the target mRNA. Following washing to remove the non-specifically bound probe, the label is detected. For example a digoxygenin labeled riboprobe (RNA probe) that is complementary to the mRNA encoding a ovarian cancer-associated protein is detected by binding the digoxygenin with an anti-digoxygenin secondary antibody and developed with nitro blue tetrazolium and 5-bromo-4-chloro-3-indoyl phosphate.


In a preferred embodiment, various proteins from the three classes of proteins as described herein (secreted, transmembrane or intracellular proteins) are used in diagnostic assays. The ovarian cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing ovarian cancer sequences are used in diagnostic assays. This are performed on an individual gene or corresponding polypeptide level. In a preferred embodiment, the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes and/or corresponding polypeptides.


As described and defined herein, ovarian cancer-associated proteins, including intracellular, transmembrane or secreted proteins, find use as markers of ovarian cancer. Detection of these proteins in putative ovarian cancer tissue allows for detection or diagnosis of ovarian cancer. In one embodiment, antibodies are used to detect ovarian cancer-associated proteins. A preferred method separates proteins from a sample by electrophoresis on a gel (typically a denaturing and reducing protein gel, but are another type of gel, including isoelectric focusing gels and the like). Following separation of proteins, the ovarian cancer-associated protein is detected, e.g., by immunoblotting with antibodies raised against the ovarian cancer-associated protein. Methods of immunoblotting are well known to those of ordinary skill in the art.


In another preferred method, antibodies to the ovarian cancer-associated protein find use in in situ imaging techniques, e.g., in histology (e.g., Methods in Cell Biology: Antibodies in Cell Biology, volume 37 (Asai, ed. 1993)). In this method cells are contacted with from one to many antibodies to the ovarian cancer-associated protein(s). Following washing to remove non-specific antibody binding, the presence of the antibody or antibodies is detected. In one embodiment the antibody is detected by incubating with a secondary antibody that contains a detectable label. In another method the primary antibody to the ovarian cancer-associated proteins) contains a detectable label, e.g. an enzyme marker that can act on a substrate. In another preferred embodiment each one of multiple primary antibodies contains a distinct and detectable label. This method finds particular use in simultaneous screening for a plurality of ovarian cancer-associated proteins. As will be appreciated by one of ordinary skill in the art, many other histological imaging techniques are also provided by the invention.


In a preferred embodiment the label is detected in a fluorometer which has the ability to detect and distinguish emissions of different wavelengths. In addition, a fluorescence activated cell sorter (FACS) are used in the method. In another preferred embodiment, antibodies find use in diagnosing ovarian cancer from blood, serum, plasma, stool, and other samples. Such samples, therefore, are useful as samples to be probed or tested for, the presence of ovarian cancer-associated proteins. Antibodies are used to detect a ovarian cancer-associated protein by previously described immunoassay techniques including ELISA, immunoblotting (western blotting), immunoprecipitation, BIACORE technology and the like. Conversely, the presence of antibodies may indicate an immune response against an endogenous ovarian cancer-associated protein.


In a preferred embodiment, in situ hybridization of labeled ovarian cancer nucleic acid probes to tissue arrays is done. For example, arrays of tissue samples, including ovarian cancer tissue and/or normal tissue, are made. In situ hybridization (see, e.g., Ausubel, supra) is then performed. When comparing the fingerprints between an individual and a standard, the skilled artisan can make a diagnosis, a prognosis, or a prediction based on the findings. It is further understood that the genes which indicate the diagnosis may differ from those which indicate the prognosis and molecular profiling of the condition of the cells may lead to distinctions between responsive or refractory conditions or are predictive of outcomes.


In a preferred embodiment, the ovarian cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing ovarian cancer sequences are used in prognosis assays. As above, gene expression profiles are generated that correlate to ovarian cancer, in terms of long term prognosis. Again, this are done on either a protein or gene level, with the use of genes being preferred. As above, ovarian cancer probes are attached to biochips for the detection and quantification of ovarian cancer sequences in a tissue or patient. The assays proceed as outlined above for diagnosis. PCR method may provide more sensitive and accurate quantification.


Characteristics of Ovarian Cancer-Associated Proteins and Genes Encoding Same


Ovarian cancer-associated proteins of the present invention are classified as secreted proteins, transmembrane proteins or intracellular proteins. In one embodiment, the ovarian cancer-associated protein is an intracellular protein. Intracellular proteins are found in the cytoplasm and/or in the nucleus. Intracellular proteins are involved in all aspects of cellular function and replication (including, e.g., signaling pathways); aberrant expression of such proteins often results in unregulated or disregulated cellular processes (see, e.g., Molecular Biology of the Cell (Alberts, ed., 3rd ed., 1994). For example, many intracellular proteins have enzymatic activity such as protein kinase activity, protein phosphatase activity, protease activity, nucleotide cyclase activity, polymerase activity and the like. Intracellular proteins also serve as docking proteins that are involved in organizing complexes of proteins, or targeting proteins to various subcellular localizations, and are involved in maintaining the structural integrity of organelles.


An increasingly appreciated concept in characterising proteins is the presence in the proteins of one or more motifs for which defined functions have been attributed. In addition to the highly conserved sequences found in the enzymatic domain of proteins, highly conserved sequences have been identified in proteins that are involved in protein-protein interaction. For example, Src-homology-2 (SH2) domains bind tyrosine-phosphorylated targets in a sequence dependent manner. PTB domains, which are distinct from SH2 domains, also bind tyrosine phosphorylated targets. SH3 domains bind to proline-rich targets. In addition, PH domains, tetratricopeptide repeats and WD domains to name only a few, have been shown to mediate protein-protein interactions. Some of these may also be involved in binding to phospholipids or other second messengers. As will be appreciated by one of ordinary skill in the art, these motifs are identified on the basis of primary sequence; thus, an analysis of the sequence of proteins may provide insight into both the enzymatic potential of the molecule and/or molecules with which the protein may associate. One useful database is Pfam (protein families), which is a large collection of multiple sequence alignments and hidden Markov models covering many common protein domains. Versions are available via the internet from Washington University in St. Louis, the Sanger Center in England, and the Karolinska Institute in Sweden (see, e.g., Bateman et al., 2000, Nuc. Acids Res. 28: 263-266; Sonnhammer et al., 1997, Proteins 28: 405-420; Bateman et al., 1999, Nuc. Acids Res. 27:260-262; and Sonnhammer et al., 1998, Nuc. Acids Res. 26: 320-322.


In another embodiment, the ovarian cancer sequences are transmembrane proteins. Transmembrane proteins are molecules that span a phospholipid bilayer of a cell. They may have an intracellular domain, an extracellular domain, or both. The intracellular domains of such proteins may have a number of functions including those already described for intracellular proteins. For example, the intracellular domain may have enzymatic activity and/or may serve as a binding site for additional proteins. Frequently the intracellular domain of transmembrane proteins serves both roles. For example certain receptor tyrosine kinases have both protein kinase activity and SH2 domains. In addition, autophosphorylation of tyrosines on the receptor molecule itself, creates binding sites for additional SH2 domain containing proteins.


Transmembrane proteins may contain from one to many transmembrane domains. For example, receptor tyrosine kinases, certain cytokine receptors, receptor guanylyl cyclases and receptor serine/threonine protein kinases contain a single transmembrane domain. However, various other proteins including channels and adenylyl cyclases contain numerous transmembrane domains. Many important cell surface receptors such as G protein coupled receptors (GPCRs) are classified as “seven transmembrane domain” proteins, as they contain 7 membrane spanning regions. Characteristics of transmembrane domains include approximately 20 consecutive hydrophobic amino acids that are followed by charged amino acids. Therefore, upon analysis of the amino acid sequence of a particular protein, the localization and number of transmembrane domains within the protein are predicted (see, e.g. PSORT web site http://psort.nibb.ac.jp/). Important transmembrane protein receptors include, but are not limited to the insulin receptor, insulin-like growth factor receptor, human growth hormone receptor, glucose transporters, transferrin receptor, epidermal growth factor receptor, low density lipoprotein receptor, epidermal growth factor receptor, leptin receptor, interleukin receptors, e.g. IL-1 receptor, IL-2 receptor,


The extracellular domains of transmembrane proteins are diverse; however, conserved motifs are found repeatedly among various extracellular domains. Conserved structure and/or functions have been ascribed to different extracellular motifs. Many extracellular domains are involved in binding to other molecules. In one aspect, extracellular domains are found on receptors. Factors that bind the receptor domain include circulating ligands, which are peptides, proteins, or small molecules such as adenosine and the like. For example, growth factors such as EGF, FGF and PDGF are circulating growth factors that bind to their cognate receptors to initiate a variety of cellular responses. Other factors include cytokines, mitogenic factors, neurotrophic factors and the like. Extracellular domains also bind to cell-associated molecules. In this respect, they mediate cell-cell interactions. Cell-associated ligands are tethered to the cell, e.g., via a glycosylphosphatidylinositol (GPI) anchor, or may themselves be transmembrane proteins. Extracellular domains also associate with the extracellular matrix and contribute to the maintenance of the cell structure.


Ovarian cancer-associated proteins that are transmembrane are particularly preferred in the present invention as they are readily accessible targets for immunotherapeutics, as are described herein. In addition, as outlined below, transmembrane proteins are also useful in imaging modalities. Antibodies are used to label such readily accessible proteins in situ. Alternatively, antibodies can also label intracellular proteins, in which case samples are typically permeablized to provide access to intracellular proteins.


It will also be appreciated by those in the art that a transmembrane protein are made soluble by removing transmembrane sequences, e.g., through recombinant methods. Furthermore, transmembrane proteins that have been made soluble are made to be secreted through recombinant means by adding an appropriate signal sequence.


In another embodiment, the ovarian cancer-associated proteins are secreted proteins; the secretion of which are either constitutive or regulated. These proteins have a signal peptide or signal sequence that targets the molecule to the secretory pathway. Secreted proteins are involved in numerous physiological events; by virtue of their circulating nature, they serve to transmit signals to various other cell types. The secreted protein may function in an autocrine manner (acting on the cell that secreted the factor), a paracrine manner (acting on cells in close proximity to the cell that secreted the factor) or an endocrine manner (acting on cells at a distance). Thus secreted molecules find use in modulating or altering numerous aspects of physiology. Ovarian cancer-associated proteins that are secreted proteins are particularly preferred in the present invention as they serve as good targets for diagnostic markers, e.g., for blood, plasma, serum, or stool tests.


Mammalian Subjects


The present invention provides nucleic acid and protein sequences that are differentially expressed in ovarian cancer, herein termed “ovarian cancer sequences.” As outlined below, ovarian cancer sequences include those that are up-regulated (i.e., expressed at a higher level) in ovarian cancer, as well as those that are down-regulated (i.e., expressed at a lower level). In a preferred embodiment, the ovarian cancer sequences are from humans; however, as will be appreciated by those in the art, ovarian cancer sequences from other organisms are useful in animal models of disease and drug evaluation; thus, other ovarian cancer sequences are provided, from vertebrates, including mammals, including rodents (rats, mice, hamsters, guinea pigs, etc.), primates, farm animals (including sheep, goats, pigs, cows, horses, etc.) and pets, e.g., (dogs, cats, etc.).


Assay Control Samples


It will be apparent from the preceding discussion that many of the diagnostic methods provided by the present invention involve a degree of quantification to determine, on the one hand, the over-expression or reduced-expression of a diagnostic/prognostic marker in tissue that is suspected of comprising a cancer cell. Such quantification can be readily provided by the inclusion of appropriate control samples in the assays described below, derived from healthy or normal individuals. Alternatively, if internal controls are not included in each assay conducted, the control may be derived from an established data set that has been generated from healthy or normal individuals.


In the present context, the term “healthy individual” shall be taken to mean an individual who is known not to suffer from ovarian cancer, such knowledge being derived from clinical data on the individual, including, but not limited to, a different cancer assay to that described herein. As the present invention is particularly useful for the early detection of ovarian cancer, it is preferred that the healthy individual is asymptomatic with respect to the early symptoms associated with ovarian cancer. Although early detection using well-known procedures is difficult, reduced urinary frequency, rectal pressure, and abdominal bloating and swelling, are associated with the disease in its early stages, and, as a consequence, healthy individuals should not have any of these clinical symptoms. Clearly, subjects suffering from later symptoms associated with ovarian cancer, such as, for example, metastases in the omentum, abdominal fluid, lymph nodes, lung, liver, brain, or bone, and subjects suffering from spinal cord compression, elevated calcium level, chronic pain, or pleural effusion, should also be avoided from the “healthy individual” data set.


The term “normal individual” shall be taken to mean an individual having a normal level of expression of a cancer-associate gene or cancer-associated protein in a particular sample derived from said individual. As will be known to those skilled in the art, data obtained from a sufficiently large sample of the population will normalize, allowing the generation of a data set for determining the average level of a particular parameter. Accordingly, the level of expression of a cancer-associate gene or cancer-associated protein can be determined for any population of individuals, and for any sample derived from said individual, for subsequent comparison to levels determined for a sample being assayed. Where such normalized data sets are relied upon, internal controls are preferably included in each assay conducted to control for variation.


In one embodiment, the present invention provides a method for detecting a cancer cell in a subject, said method comprising:

  • (i) determining the level of mRNA encoding a cancer-associated protein expressed in a test sample from said subject; and
  • (ii) comparing the level of mRNA determined at (i) to the level of mRNA encoding a cancer-associated protein expressed in a comparable sample from a healthy or normal individual,


    wherein a level of mRNA at (i) that is modified in the test sample relative to the comparable sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.


Alternatively, or in addition, the control may comprise a cancer-associated sequence that is known to be expressed at a particular level in an ovarian cancer, eg., CA125, MUC-1 or E-Cadherin, amongast others.


Biological Samples


Preferred biological samples in which the assays of the invention are performed include bodily fluids, ovarian tissue and cells, and those tissues known to comprise cancer cells arising from a metastasis of an ovarian cancer, such as, for example, in carcinomas of the lung, prostate, breast, colon, pancreas, placenta, or omentum, and in cells of brain anaplastic oligodendrogliomas.


Bodily fluids shall be taken to include whole blood, serum, peripheral blood mononuclear cells (PBMC), or buffy coat fraction.


In the present context, the term “cancer cell” includes any biological specimen or sample comprising a cancer cell irrespective of its degree of isolation or purity, such as, for example, tissues, organs, cell lines, bodily fluids, or histology specimens that comprise a cell in the early stages of transformation or having been transformed.


As the present invention is particularly useful for the early detection and prognosis of cancer ofe rthe medium to long term, the definition of “cancer cell” is not to be limited by the stage of a cancer in the subject from which said cancer cell is derived (ie. whether or not the patient is in remission or undergoing disease recurrence or whether or not the cancer is a primary tumor or the consequence of metastases). Nor is the term “cancer cell” to be limited by the stage of the cell cycle of said cancer cell.


Preferably, the sample comprises ovarian tissue, prostate tissue, kidney tissue, uterine tissue, placenta, a cervical specimen, omentum, rectal tissue, brain tissue, bone tissue, lung tissue, lymphatic tissue, urine, semen, blood, abdominal fluid, or serum, or a cell preparation or nucleic acid preparation derived therefrom. More preferably, the sample comprises serum or abdominal fluid, or a tissue selected from the group consisting of: ovary, lymph, lung, liver, brain, placenta, brain, omentum, and prostate. Even more preferably, the sample comprises serum or abdominal fluid, ovary (eg. OSE), or lymph node tissue. The sample can be prepared on a solid matrix for histological analyses, or alternatively, in a suitable solution such as, for example, an extraction buffer or suspension buffer, and the present invention clearly extends to the testing of biological solutions thus prepared.


Polynucleotide Probes and Amplification Primers


Polynucleotide probes are derived from or comprise the nucleic acid sequences whose nucleotide sequences are provided by reference to the public database accession numbers given in Tables 1-3 (referred to herein as the nucleotide sequences shown in Tables 1-3), and sequences homologues thereto as well as variants, derivatives and fragments thereof.


Whilst the probes may comprise double-stranded or single-stranded nucleic acid, single-stranded probes are preferred because they do not require melting prior to use in hybridizations. On the other hand, longer probes are also preferred because they can be used at higher hybridization stringency than shorter probes and may produce lower background hybridization than shorter probes.


So far as shorter probes are concerned, single-stranded, chemically-synthesized oligonucleotide probes are particularly preferred by the present invention. To reduce the noise associated with the use of such probes during hybridization, the nucleotide sequence of the probe is carefully selected to maximize the Tm at which hybridizations can be performed, reduce non-specific hybridization, and to reduce self-hybridization. Such considerations may be particularly important for applications involving high throughput screening using microarray technology. In general, this means that the nucleotide sequence of an oligonucleotide probe is selected such that it is unique to the target RNA or protein-encoding sequence, has a low propensity to form secondary structure, low self-complementary, and is not highly A/T-rich.


The only requirement for the probes is that they cross-hybridize to nucleic acid encoding the target diagnostic protein or the complementary nucleotide sequence thereto and are sufficiently unique in sequence to generate high signal:noise ratios under specified hybridization conditions. As will be known to those skilled in the art, long nucleic acid probes are preferred because they tend to generate higher signal:noise ratios than shorter probes and/or the duplexes formed between longer molecules have higher melting temperatures (i.e. Tm values) than duplexes Involving short probes. Accordingly, full-length DNA or RNA probes are contemplated by the present invention, as are specific probes comprising the sequence of the 3′-untranslated region or complementary thereto.


In a particularly preferred embodiment, the nucleotide sequence of an oligonucleotide probe has no detectable nucleotide sequence identity to a nucleotide sequence in a BLAST search (Altschul et al., J. Mol. Biol. 215, 403-410, 1990) or other database search, other than a sequence selected from the group consisting of: (a) a sequence encoding a polypeptide listed in any one of Tables 1-3; (b) the 5′-untranslated region of a sequence encoding a polypeptide listed in any one of Tables 1-3; (c) a 3′-untranslated region of a sequence encoding a polypeptide listed in any one of Tables 1-3; and (d) an exon region of a sequence encoding a polypeptide listed in any one of Tables 1-3.


Additionally, the self-complementarity of a nucleotide sequence can be determined by aligning the sequence with its reverse complement, wherein detectable regions of identity are indicative of potential self-complementarity. As will be known to those skilled in the art, such sequences may not necessarily form secondary structures during hybridization reaction, and, as a consequence, successfully identify a target nucleotide sequence. It is also known to those skilled in the art that, even where a sequence does form secondary structures during hybridization reactions, reaction conditions can be modified to reduce the adverse consequences of such structure formation. Accordingly, a potential for self-complementarity should not necessarily exclude a particular candidate oligonucleotide from selection. In cases where it is difficult to determine nucleotide sequences having no potential self-complementarity, the uniqueness of the sequence should outweigh a consideration of its potential for secondary structure formation.


Recommended pre-requisites for selecting oligonucleotide probes, particularly with respect to probes suitable for microarray technology, are described in detail by Lockhart et al., “Expression monitoring by hybridization to high-density oligonucleotide arrays”, Nature Biotech. 14, 1675-1680, 1996.


The nucleic acid probe may comprise a nucleotide sequence that is within the coding strand of a gene listed in any one of Tables 1-3. Such “sense” probes are useful for detecting RNA by amplification procedures, such as, for example, polymerase chain reaction (PCR), and more preferably, quantitative PCR or reverse transcription polymerase chain reaction (RT-PCR). Alternatively, “sense” probes may be expressed to produce polypeptides or immunologically active derivatives thereof that are useful for detecting the expressed protein in samples.


The nucleotide sequences referred to in Tables 1-3 and homologues thereof, typically encode polypeptides. It will be understood by a skilled person that numerous different polynucleotides can encode the same polypeptide as a result of the degeneracy of the genetic code. In addition, it is to be understood that skilled persons may, using routine techniques, make nucleotide substitutions that do not affect the polypeptide sequence encoded by the polynucleotides of the invention to reflect the codon usage of any particular host organism in which the polypeptides of the invention are to be expressed.


Polynucleotides may comprise DNA or RNA. They are single-stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art. These include methylphosphonate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3′ and/or 5′ ends of the molecule. For the purposes of the present invention, it is to be understood that the polynucleotides described herein are modified by any method available in the art. Such modifications are carried out in order to enhance the in vivo activity or life span of the diagnostic/prognostic polynucleotides.


The terms “variant” or “derivative” in relation to the nucleotide sequences of the present invention include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid from or to the sequence provided that the resultant nucleotide sequence codes for a polypeptide having biological activity, preferably having substantially the same activity as the polypeptide sequences presented in the sequence listings.


With respect to sequence homology, preferably there is at least 75%, more preferably at least 85%, more preferably at least 90% homology to a sequence shown in Tables 1-3 herein over a region of at least 20, preferably at least 25 or 30, for instance at least 40, 60, 100, 500, 1000 or more contiguous nucleotides. More preferably there is at least 95%, more preferably at least 98%, homology. In one embodiment, homologues are naturally occurring sequences, such as orthologues, tissue-specific isoforms and allelic variants.


Homology comparisons are conducted by eye, or more usually, with the aid of readily available sequence comparison programs. These commercially available computer programs can calculate % homology between two or more sequences.


Percentage (%) homology are calculated over contiguous sequences, i.e. one sequence is aligned with the other sequence and each nucleotide in one sequence directly compared with the corresponding nucleotide in the other sequence, one base at a time. This is called an “ungapped” alignment. Typically, such ungapped alignments are performed only over a relatively short number of bases (for example less than 50 contiguous nucleotides).


Although this is a very simple and consistent method, it fails to take into consideration that, for example, in an otherwise identical pair of sequences, one insertion or deletion will cause the following nucleotides to be put out of alignment, thus potentially resulting in a large reduction in % homology when a global alignment is performed. Consequently, most sequence comparison methods are designed to produce optimal alignments that take into consideration possible insertions and deletions without penalising unduly the overall homology score. This is achieved by inserting “gaps” in the sequence alignment to try to maximise local homology.


However, these more complex methods assign “gap penalties” to each gap that occurs in the alignment so that, for the same number of identical amino acids, a sequence alignment with as few gaps as possible—reflecting higher relatedness between the two compared sequences—will achieve a higher score than one with many gaps. “Affine gap costs” are typically used that charge a relatively high cost for the existence of a gap and a smaller penalty for each subsequent residue in the gap. This is the most commonly used gap scoring system. High gap penalties will of course produce optimised alignments with fewer gaps. Most alignment programs allow the gap penalties to be modified. However, it is preferred to use the default values when using such software for sequence comparisons.


In determining whether or not two amino acid sequences fall within the stated defined percentage identity limits, those skilled in the art will be aware that it is necessary to conduct a side-by-side comparison of amino acid sequences. In such comparisons or alignments, differences will arise in the positioning of non-identical amino acid residues depending upon the algorithm used to perform the alignment. In the present context, references to percentage identities and similarities between two or more amino acid sequences shall be taken to refer to the number of identical and similar residues respectively, between said sequences as determined using any standard algorithm known to those skilled in the art. In particular, amino acid identities and similarities are calculated using the GAP program of the Computer Genetics Group, Inc., University Research Park, Madison, Wis., United States of America (Devereaux et al, Nucl. Acids Res. 12, 387-395, 1984), which utilizes the algorithm of Needleman and Wunsch J. Mol. Biol. 48, 443-453, 1970, or alternatively, the CLUSTAL W algorithm of Thompson et al., Nucl. Acids Res. 22, 4673-4680, 1994, for multiple alignments, to maximize the number of identical/similar amino acids and to minimize the number and/or length of sequence gaps in the alignment.


A suitable computer program for carrying out such an alignment is the GCG Wisconsin Bestfit package (University of Wisconsin, U.S.A.; Devereux et al., 1984, Nucleic Acids Research 12:387). The default scoring matrix has a match value of 10 for each identical nucleotide and −9 for each mismatch. The default gap creation penalty is −50 and the default gap extension penalty is −3 for each nucleotide.


Examples of other software than can perform sequence comparisons include, but are not limited to, the BLAST package (see Ausubel et al., 1999 ibid—Chapter 18), FASTA (Atschul et al, 1990, J. Mol. Biol., 403-410) and the GENEWORKS suite of comparison tools. Both BLAST and FASTA are available for offline and online searching (see Ausubel et al., 1999 ibid, pages 7-58 to 7-60). However it is preferred to use the GCG Bestfit program.


Once the software has produced an optimal alignment, it is possible to calculate % homology, preferably % sequence identity. The software typically does this as part of the sequence comparison and generates a numerical result.


A preferred sequence comparison program is the GCG Wisconsin Bestfit program described above.


The present invention also encompasses the use of nucleotide sequences that are capable of hybridizing selectively to the sequences presented herein, or any variant, fragment or derivative thereof, or to the complement of any of the above. Nucleotide sequences are preferably at least 15 nucleotides in length, more preferably at least 20, 30, 40 or 50 nucleotides in length.


The term “hybridization” as used herein shall include “the process by which a strand of nucleic acid joins with a complementary strand through base pairing” as well as the process of amplification as carried out in polymerase chain reaction technologies.


Polynucleotides capable of selectively hybridizing to the nucleotide sequences presented herein, or to their complement, will be generally at least 70%, preferably at least 80 or 90% and more preferably at least 95% or 98% homologous to the corresponding nucleotide sequences referred to in Tables 1-3 over a region of at least 20, preferably at least 25 or 30, for instance at least 40, 60, 100, 500, 1000 or more contiguous nucleotides.


The term “selectively hybridizable” means that the polynucleotide used as a probe is used under conditions where a target polynucleotide is found to hybridize to the probe at a level significantly above background. The background hybridization may occur because of other polynucleotides present, for example, in the cDNA or genomic DNA library being screening. In this event, background implies a level of signal generated by interaction between the probe and a non-specific DNA member of the library which is less than 10 fold, preferably less than 100 fold as intense as the specific interaction observed with the target DNA. The intensity of interaction are measured, for example, by radiolabelling the probe, e.g. with 32P.


Hybridization conditions are based on the melting temperature (Tm) of the nucleic acid binding complex, as taught in Berger and Kimmel (1987, Guide to Molecular Cloning Techniques, Methods in Enzymology, Vol 152, Academic Press, San Diego Calif.), and confer a defined “stringency” as explained below.


For the purposes of defining the level of stringency, a high stringency hybridization is achieved using a hybridization buffer and/or a wash solution comprising the following:

  • (i) a salt concentration that is equivalent to 0.1×SSC-0.2×SSC buffer or lower salt concentration;
  • (ii) a detergent concentration equivalent to 0.1% (w/v) SDS or higher; and
  • (iii) an incubation temperature of 55° C. or higher.


Conditions for specifically hybridizing nucleic acid, and conditions for washing to remove non-specific hybridizing nucleic acid, are well understood by those skilled in the art. For the purposes of further clarification only, reference to the parameters affecting hybridization between nucleic acid molecules is found in Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, ISBN 047150338, 1992), which is herein incorporated by reference.


Maximum stringency typically occurs at about Tm-5° C. (5° C. below the Tm of the probe); high stringency at about 5° C. to 10° C. below Tm; intermediate stringency at about 10° C. to 20° C. below Tm; and low stringency at about 20° C. to 25° C. below Tm. As will be understood by those of skill in the art, a maximum stringency hybridization are used to identify or detect identical polynucleotide sequences while an intermediate (or low) stringency hybridization are used to identify or detect similar or related polynucleotide sequences.


In a preferred aspect, the present invention covers nucleotide sequences that can hybridize to the nucleotide sequence of the present invention under stringent conditions (e.g. 65° C. and 0.1×SSC {1×SSC=0.15 M NaCl, 0.015 M Na3Citrate pH 7.0}).


Where the diagnostic/prognostic polynucleotide is double-stranded, both strands of the duplex, either individually or in combination, are encompassed by the present invention. Where the polynucleotide is single-stranded, it is to be understood that the complementary sequence of that polynucleotide is also included within the scope of the present invention.


Polynucleotides which are not 100% homologous to the sequences of the present invention but are useful in perfoming the diagnostic and/or prognostic assays of the invention by virtue of their ability to selectively hybridize to the target gene transcript, or to encode an immunologically cross-reactive protein to the target protein, are obtained in a number of ways, such as, for example by probing DNA libraries made from a range of individuals, for example individuals from different populations. In particular, given that that changes in the expression of diagnostic/prognostic cancer-associated genes correlate with ovarian cancer, characterisation of variant sequences in individuals suffering from ovarian cancer is used to identify variations in the sequences of ovarian-cancer associated genes (and proteins) that are predictive of and/or causative of ovarian cancer.


Accordingly the present invention provides methods of identifying sequence variants that are associated with ovarian cancer which methods comprise determining all or part of the nucleotide sequence of a gene referred to in Tables 1-3, derived from an individual suffering from ovarian cancer and comparing the sequence to that of the corresponding wild-type gene.


In addition, other viral/bacterial, or cellular homologues particularly cellular homologues found in mammalian cells (e.g. rat, mouse, bovine and primate cells), are obtained and such homologues and fragments thereof in general will be capable of selectively hybridizing to the sequences shown in the sequence listing herein. Such sequences are obtained by probing cDNA libraries made from or genomic DNA libraries from other animal species, and probing such libraries with probes comprising all or part of the sequences referred to in Tables 1-3 under conditions of medium to high stringency. Similar considerations apply to obtaining species homologues and allelic variants of the nucleotide sequences referred to in Tables 1-3.


Variants and strain/species homologues may also be obtained using degenerate PCR which will use primers designed to target sequences within the variants and homologues encoding conserved amino acid sequences within the sequences of the present invention. Conserved sequences are predicted, for example, by aligning the amino acid sequences from several variants/homologues. Sequence alignments are performed using computer software known in the art. For example the GCG Wisconsin PileUp program is widely used.


The primers used in degenerate PCR will contain one or more degenerate positions and will be used at stringency conditions lower than those used for cloning sequences with single sequence primers against known sequences.


Alternatively, such polynucleotides are obtained by site-directed mutagenesis of characterised sequences, such as the sequences referred to in Tables 1-3. This are useful where for example silent codon changes are required to sequences to optimise codon preferences for a particular host cell in which the polynucleotide sequences are being expressed. Other sequence changes are desired in order to introduce restriction enzyme recognition sites, or to alter the property or function of the polypeptides encoded by the polynucleotides.


Polynucleotides comprising a diagnostic/prognostic cancer-associated gene are used to produce a primer by standard derivatization means, e.g. a PCR primer, a primer for an alternative amplification reaction, a probe e.g. labelled with a detectable label by conventional means using radioactive or nonradioactive labels, or the polynucleotides are cloned into vectors. Such primers, probes and other fragments will be at least 15, preferably at least 20, for example at least 25, 30 or 40 nucleotides in length. Preferred fragments are less than 5000, 2000, 1000, 500 or 200 nucleotides in length.


Polynucleotides such as a DNA polynucleotides and probes according to the invention are produced by recombinant or synthetic means, including cloning by standard techniques.


In general, primers will be produced by synthetic means, involving a step wise manufacture of the desired nucleic acid sequence one nucleotide at a time. Techniques for accomplishing this using automated techniques are readily available in the art.


Longer polynucleotides will generally be produced using recombinant means, for example using PCR (polymerase chain reaction) cloning techniques. This will involve making a pair of primers (e.g. of about 15 to 30 nucleotides) flanking a region of the sequence which it is desired to clone, bringing the primers into contact with mRNA or cDNA obtained from an animal or human cell, performing a polymerase chain reaction under conditions which bring about amplification of the desired region, isolating the amplified fragment (e.g. by purifying the reaction mixture on an agarose gel) and recovering the amplified DNA. The primers are designed to contain suitable restriction enzyme recognition sites so that the amplified DNA are cloned into a suitable cloning vector


Polynucleotide probes or primers preferably carry a detectable label. Suitable labels include radioisotopes such as 32P or 35S, enzyme labels, or other protein labels such as biotin. Such labels are added to polynucleotides or primers and are detected using by techniques known in the art.


Polynucleotide probes or primers labeled or unlabeled are also used by a person skilled in the art in nucleic acid-based tests for detecting or sequencing diagnostic/prognostic cancer-associated gene.


Such tests for detecting generally comprise bringing a biological sample containing DNA or RNA into contact with a probe comprising a polynucleotide probe or primer under at least low stringency hybridization conditions and detecting any duplex formed between the probe/primer and nucleic acid in the sample. Such detection are achieved using techniques such as PCR or by immobilising the probe on a solid support, removing nucleic acid in the sample which is not hybridized to the probe, and then detecting nucleic acid which has hybridized to the probe. Alternatively, the sample nucleic acid are immobilised on a solid support, and the amount of probe bound to such a support are detected. Suitable assay methods of this and other formats are found in for example WO89/03891 and WO90/13667.


Tests for sequencing nucleotides include bringing a biological sample containing target DNA or RNA into contact with a probe comprising a polynucleotide probe or primer under at least low stringency hybridization conditions and determining the sequence by, for example the Sanger dideoxy chain termination method (see Sambrook et al.).


Such a method generally comprises elongating, in the presence of suitable reagents, the primer by synthesis of a strand complementary to the target DNA or RNA and selectively terminating the elongation reaction at one or more of an A, C, G or T/U residue; allowing strand elongation and termination reaction to occur; separating out according to size the elongated products to determine the sequence of the nucleotides at which selective termination has occurred. Suitable reagents include a DNA polymerase enzyme, the deoxynucleotides dATP, dCTP, dGTP and dTTP, a buffer and ATP. Dideoxynucleotides are used for selective termination.


Tests for detecting or sequencing nucleotides in a biological sample are used as part of the methods of the invention for detecting ovarian cancer-associated transcripts and monitoring the efficacy of treatment of patients suffering from ovarian cancer as described in more detail herein.


The probes/primers may conveniently be packaged in the form of a test kit in a suitable container. In such kits the probe are bound to a solid support where the assay format for which the kit is designed requires such binding. The kit may also contain suitable reagents for treating the sample to be probed, hybridizing the probe to nucleic acid in the sample, control reagents, instructions, and the like.


Preferably, a kit of the invention comprises primers/probes suitable for selectively detecting a plurality of sequences, more preferably for selectively detecting a plurality of sequences that are listed in one or more of Tables 1-3 as having a P value of less than 0.05, more preferably a P value of less than 0.01. Similarly, a kit of the invention preferably comprises primers suitable for selectively detecting a plurality of sequences referred to in Table 1 or 2 or 3.


Nucleic Acid-Based Assay Formats


As discussed in detail below, the status of expression of a cancer-associated gene in patient samples may be analyzed by a variety protocols that are well known in the art including in situ hybridization, northern blotting techniques, RT-PCR analysis (such as, for example, performed on laser capture microdissected samples), and microarray technology, such as, for example, using tissue microarrays probed with nucleic acid probes, or nucleic acid microarrays (ie. RNA microarrays or amplified DNA microarrays) microarrays probed with nucleic acid probes. All such assay formats are encompassed by the present invention.


For high throughput screening of large numbers of samples, such as, for example, public health screening of subjects, particularly human subjects, having a higher risk of developing cancer, microarray technology is a preferred assay format.


In accordance with such high throughput formats, techniques for producing immobilised arrays of DNA molecules have been described in the art. Generally, most prior art methods describe how to synthesise single-stranded nucleic acid molecule arrays, using for example masking techniques to build up various permutations of sequences at the various discrete positions on the solid substrate. U.S. Pat. No. 5,837,832, the contents of which are incorporated herein by reference, describes an improved method for producing DNA arrays immobilised to silicon substrates based on very large scale integration technology. In particular, U.S. Pat. No. 5,837,832 describes a strategy called “tiling” to synthesize specific sets of probes at spatially-defined locations on a substrate which are used to produced the immobilised DNA arrays. U.S. Pat. No. 5,837,832 also provides references for earlier techniques that may also be used.


Thus DNA are synthesised in situ on the surface of the substrate. However, DNA may also be printed directly onto the substrate using for example robotic devices equipped with either pins or piezo electric devices.


The plurality of polynucleotide sequences are typically immobilised onto or in discrete regions of a solid substrate. The substrate are porous to allow immobilisation within the substrate or substantially non-porous, in which case the library sequences are typically immobilised on the surface of the substrate. The solid substrate are made of any material to which polypeptides can bind, either directly or indirectly. Examples of suitable solid substrates include flat glass, silicon wafers, mica, ceramics and organic polymers such as plastics, including polystyrene and polymethacrylate. It may also be possible to use semi-permeable membranes such as nitrocellulose or nylon membranes, which are widely available. The semi-permeable membranes are mounted on a more robust solid surface such as glass. The surfaces may optionally be coated with a layer of metal, such as gold, platinum or other transition metal. A particular example of a suitable solid substrate is the commercially available BIACore™ chip (Pharmacia Biosensors).


Preferably, the solid substrate is generally a material having a rigid or semi-rigid surface. In preferred embodiments, at least one surface of the substrate will be substantially flat, although in some embodiments it are desirable to physically separate synthesis regions for different polymers with, for example, raised regions or etched trenches. It is also preferred that the solid substrate is suitable for the high density application of DNA sequences in discrete areas of typically from 50 to 100 μm, giving a density of 10000 to 40000 cm−2.


The solid substrate is conveniently divided up into sections. This are achieved by techniques such as photoetching, or by the application of hydrophobic inks, for example teflon-based inks (Cel-line, USA).


Discrete positions, in which each different member of the array is located may have any convenient shape, e.g., circular, rectangular, elliptical, wedge-shaped, etc.


Attachment of the polynucleotide sequences to the substrate are by covalent or non-covalent means. The plurality of polynucleotide sequences are attached to the substrate via a layer of molecules to which the sequences bind. For example, the sequences are labelled with biotin and the substrate coated with avidin and/or streptavidin. A convenient feature of using biotinylated sequences is that the efficiency of coupling to the solid substrate are determined easily. Since the library sequences may bind only poorly to some solid substrates, it is often necessary to provide a chemical interface between the solid substrate (such as in the case of glass) and the sequences. Examples of suitable chemical interfaces include hexaethylene glycol. Another example is the use of polylysine coated glass, the polylysine then being chemically modified using standard procedures to introduce an affinity ligand. Other methods for attaching molecules to the surfaces of solid substrate by the use of coupling agents are known in the art, see for example WO98/49557.


The complete DNA array is typically read at the same time by charged coupled device (CCD) camera or confocal imaging system. Alternatively, the DNA array are placed for detection in a suitable apparatus that can move in an x-y direction, such as a plate reader. In this way, the change in characteristics for each discrete position are measured automatically by computer controlled movement of the array to place each discrete element in turn in line with the detection means.


The detection means are capable of interrogating each position in the library array optically or electrically. Examples of suitable detection means include CCD cameras or confocal imaging systems.


In a preferred embodiment, the level of expression of the cancer-associated gene in the test sample is determined by hybridizing a probe/primer to RNA in the test sample under at least low stringency hybridization conditions and detecting the hybridization using a detection means.


Similarly, the level of mRNA in the comparable sample from the healthy or normal individual is preferably determined by hybridizing a probe/primer to RNA in said comparable sample under at least low stringency hybridization conditions and detecting the hybridization using a detection means.


For the purposes of defining the level of stringency to be used in these diagnostic assays, a low stringency is defined herein as being a hybridization and/or a wash carried out in 6×SSC buffer, 0.1% (w/v) SDS at 28° C., or equivalent conditions. A moderate stringency is defined herein as being a hybridization and/or washing carried out in 2×SSC buffer, 0.1% (w/v) SDS at a temperature in the range 45° C. to 65° C., or equivalent conditions. A high stringency is defined herein as being a hybridization and/or wash carried out in 0.1×SSC buffer, 0.1% (w/v) SDS, or lower salt concentration, and at a temperature of at least 65° C., or equivalent conditions. Reference herein to a particular level of stringency encompasses equivalent conditions using wash/hybridization solutions other than SSC known to those skilled in the art.


Generally, the stringency is increased by reducing the concentration of SSC buffer, and/or increasing the concentration of SDS and/or increasing the temperature of the hybridization and/or wash. Those skilled in the art will be aware that the conditions for hybridization and/or wash may vary depending upon the nature of the hybridization matrix used to support the sample RNA, or the type of hybridization probe used.


In general, the sample or the probe is immobilized on a solid matrix or surface (e.g., nitrocellulose). For high throughput screening, the sample or probe will generally comprise an array of nucleic acids on glass or other solid matrix, such as, for example, as described in WO 96/17958. Techniques for producing high density arrays are described, for example, by Fodor et al., Science 767-773, 1991, and in U.S. Pat. No. 5,143,854. Typical protocols for other assay formats can be found, for example in Current Protocols In Molecular Biology, Unit 2 (Northern Blotting), Unit 4 (Southern Blotting), and Unit 18 (PCR Analysis), Frederick M. Ausubul et al. (ed)., 1995.


The detection means according to this aspect of the invention may be any nucleic acid-based detection means such as, for example, nucleic acid hybridization or amplification reaction (eg. PCR), a nucleic acid sequence-based amplification (NASBA) system, inverse polymerase chain reaction (iPCR), in situ polymerase chain reaction, or reverse transcription polymerase chain reaction (RT-PCR), amongst others.


The probe can be labelled with a reporter molecule capable of producing an identifiable signal (e.g., a radioisotope such as 32P or 35S, or a fluorescent or biotinylated molecule). According to this embodiment, those skilled in the art will be aware that the detection of said reporter molecule provides for identification of the probe and that, following the hybridization reaction, the detection of the corresponding nucleotide sequences in the sample is facilitated. Additional probes can be used to confirm the assay results obtained using a single probe.


Wherein the detection means is an amplification reaction such as, for example, a polymerase chain reaction or a nucleic acid sequence-based amplification (NASBA) system or a variant thereof, one or more nucleic acid probes molecules of at least about contiguous nucleotides in length is hybridized to mRNA encoding a cancer-associated protein, or alternatively, hybridized to cDNA or cRNA produced from said mRNA, and nucleic acid copies of the template are enzymically-amplified.


Those skilled in the art will be aware that there must be a sufficiently high percentage of nucleotide sequence identity between the probes and the RNA sequences in the sample template molecule for hybridization to occur. As stated previously, the stringency conditions can be selected to promote hybridization.


In one format, PCR provides for the hybridization of non-complementary probes to different strands of a double-stranded nucleic acid template molecule (ie. a DNA/RNA, RNA/RNA or DNA/DNA template), such that the hybridized probes are positioned to facilitate the 5′- to 3′ synthesis of nucleic acid in the intervening region, under the control of a thermostable DNA polymerase enzyme. In accordance with this embodiment, one sense probe and one antisense probe as described herein would be used to amplify DNA from the hybrid RNA/DNA template or cDNA.


In the present context, the cDNA would generally be produced by reverse transcription of mRNA present in the sample being tested (ie. RT-PCR). RT-PCR is particularly useful when it is desirable to determine expression of a cancer-associated gene. It is also known to those skilled in the art to use mRNA/DNA hybrid molecules as a template for such amplification reactions, and, as a consequence, first strand cDNA synthesis is all that is required to be performed prior to the amplification reaction.


Variations of the embodiments described herein are described in detail by McPherson et al., PCR: A Practical Approach. (series eds, D. Rickwood and B. D. Hames), IRL Press Limited, Oxford. pp 1-253, 1991.


The amplification reaction detection means described supra can be further coupled to a classical hybridization reaction detection means to further enhance sensitivity and specificity of the inventive method, such as by hybridizing the amplified DNA with a probe which is different from any of the probes used in the amplification reaction.


Similarly, the hybridization reaction detection means described supra can be further coupled to a second hybridization step employing a probe which is different from the probe used in the first hybridization reaction.


The comparison to be performed in accordance with the present invention may be a visual comparison of the signal generated by the probe, or alternatively, a comparison of data integrated from the signal, such as, for example, data that have been corrected or normalized to allow for variation between samples. Such comparisons can be readily performed by those skilled in the art.


Polypeptides


Cancer-associated polypeptides are encoded by cancer-associated genes. It will be understood that such polypeptides include those polypeptide and fragments thereof that are homologous to the polypeptides encoded by the nucleotide sequences referred to in Tables 1-3, which are obtained from any source, for example related viral/bacterial proteins, cellular homologues and synthetic peptides, as well as variants or derivatives thereof.


Thus, the present invention encompasses the use of variants, homologues or derivatives of the cancer-associated proteins described in the accompanying Tables. In one embodiment, homologues are naturally occurring sequences, such as orthologues, tissue-specific isoforms and allelic variants.


In the context of the present invention, a homologous sequence is taken to include an amino acid sequence which is at least 60, 70, 80 or 90% identical, preferably at least 95 or 98% identical at the amino acid level over at least 20, 40, 60 or 80 amino acids with a sequence encoded by a nucleotide sequence referred to in any one of Tables 1-3. In particular, homology should typically be considered with respect to those regions of the sequence known to be essential for specific biological functions rather than non-essential neighbouring sequences.


Although amino acid homology can also be considered in terms of similarity (i.e. amino acid residues having similar chemical properties/functions), in the context of the present invention it is preferred to express homology in terms of sequence identity.


Homology comparisons are carried out as described above for nucleotide sequences with the appropriate modifications for amino acid sequences. For example when using the GCG Wisconsin Bestfit package (see below) the default gap penalty for amino acid sequences is −12 for a gap and −4 for each extension.


It should also be noted that where computer algorithms are used to align amino acid sequences, although the final % homology are measured in terms of identity, the alignment process itself is typically not based on an all-or-nothing pair comparison. Instead, a scaled similarity score matrix is generally used that assigns scores to each pairwise comparison based on chemical similarity or evolutionary distance. An example of such a matrix commonly used is the BLOSUM62 matrix—the default matrix for the BLAST suite of programs. GCG Wisconsin programs generally use either the public default values or a custom symbol comparison table if supplied (see user manual for further details). It is preferred to use the public default values for the GCG package, or in the case of other software, the default matrix, such as BLOSUM62.


The terms “variant” or “derivative” in relation to the amino acid sequences of the present invention includes any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) amino acids from or to the sequence providing the resultant amino acid sequence preferably has biological activity, preferably having at least 25 to 50% of the activity as the polypeptides referred to in the sequence listings, more preferably at least substantially the same activity. Particular details of biological activity for each polypeptide are given in Tables 1-3.


Thus, the polypeptides referred to in Tables 1-3 and homologues thereof, are modified for use in the present invention. Typically, modifications are made that maintain the activity of the sequence. Thus, in one embodiment, amino acid substitutions are made, for example from 1, 2 or 3 to 10, 20 or 30 substitutions provided that the modified sequence retains at least about 25 to 50% of, or substantially the same activity. However, in an alternative preferred embodiment, modifications to the amino acid sequences of a cancer-associated protein are made intentionally to reduce the biological activity of the polypeptide. For example truncated polypeptides that remain capable of binding to target molecules but lack functional effector domains are useful as inhibitors of the biological activity of the full length molecule.


In general, preferably less than 20%, 10% or 5% of the amino acid residues of a variant or derivative are altered as compared with the corresponding region of the polypeptides referred to in Tables 1-3.


Amino acid substitutions may include the use of non-naturally occurring analogues, for example to Increase blood plasma half-life of a therapeutically administered polypeptide (see below for further details on the production of peptide derivatives for use in therapy).


Conservative substitutions are made, for example according to the Table below. Amino acids In the same block in the second column and preferably In the same line in the third column are substituted for each other:

ALIPHATICNon-polarG A PI L VPolar - unchargedC S T MN QPolar - chargedD EK RAROMATICH F W Y


Cancer-associated proteins also include fragments of the above mentioned full length polypeptides and variants thereof, including fragments of the sequences referred to in Tables 1-3 and homologues thereof. Preferred fragments include those which include an epitope. Suitable fragments will be at least about 6 or 8, e.g. at least 10, 12, 15 or 20 amino acids in length. They may also be less than 200, 100 or 50 amino acids in length. Polypeptide fragments may contain one or more (e.g. 2, 3, 5, or 10) substitutions, deletions or insertions, including conserved substitutions. Where substitutions, deletion and/or insertions have been made, for example by means of recombinant technology, preferably less than 20%, 10% or 5% of the amino acid residues depicted in the sequence listings are altered.


Cancer-associated proteins are preferably in a substantially isolated form. It will be understood that the protein are mixed with carriers or diluents which will not interfere with the intended purpose of the protein and still be regarded as substantially isolated. A cancer-associated protein of the invention may also be in a substantially purified form, in which case it will generally comprise the protein in a preparation in which more than 90%, e.g. 95%, 98% or 99% pure as determined by SDS/PAGE or other art-recognized means for assessing protein purity.


Protein Production


For producing full-length polypeptides or immunologically active derivatives thereof by recombinant means, a protein-encoding region comprising at least about 15 contiguous nucleotides of the protein-encoding region of a nucleic acid referred to in any one of Tables 1-3 is placed in operable connection with a promoter or other regulatory sequence capable of regulating expression in a cell-free system or cellular system.


Reference herein to a “promoter” is to be taken in its broadest context and includes the transcriptional regulatory sequences of a classical genomic gene, including the TATA box which is required for accurate transcription initiation, with or without a CCAAT box sequence and additional regulatory elements (i.e., upstream activating sequences, enhancers and silencers) which alter gene expression in response to developmental and/or external stimuli, or in a tissue-specific manner. In the present context, the term “promoter” is also used to describe a recombinant, synthetic or fusion molecule, or derivative which confers, activates or enhances the expression of a nucleic acid molecule to which it is operably connected, and which encodes the polypeptide or peptide fragment. Preferred promoters can contain additional copies of one or more specific regulatory elements to further enhance expression and/or to alter the spatial expression and/or temporal expression of the said nucleic acid molecule.


Placing a nucleic acid molecule under the regulatory control of, i.e., “in operable connection with”, a promoter sequence means positioning said molecule such that expression is controlled by the promoter sequence. Promoters are generally positioned 5′ (upstream) to the coding sequence that they control. To construct heterologous promoter/structural gene combinations, it is generally preferred to position the promoter at a distance from the gene transcription start site that is approximately the same as the distance between that promoter and the gene it controls in its natural setting, i.e., the gene from which the promoter is derived. Furthermore, the regulatory elements comprising a promoter are usually positioned within 2 kb of the start site of transcription of the gene. As is known in the art, some variation in this distance can be accommodated without loss of promoter function. Similarly, the preferred positioning of a regulatory sequence element with respect to a heterologous gene to be placed under its control is defined by the positioning of the element in its natural setting, i.e., the genes from which it is derived. Again, as is known in the art, some variation in this distance can also occur.


The prerequisite for producing intact polypeptides and peptides in bacteria such as E. coli is the use of a strong promoter with an effective ribosome binding site. Typical promoters suitable for expression in bacterial cells such as E. coli include, but are not limited to, the lacz promoter, temperature-sensitive λL or λR promoters, T7 promoter or the IPTG-inducible tac promoter. A number of other vector systems for expressing the nucleic acid molecule of the invention in E. coli are well-known in the art and are described, for example, in Ausubel et al (In: Current Protocols in Molecular Biology. Wiley Interscience, ISBN 047150338, 1987) or Sambrook et al (In: Molecular cloning. A laboratory manual, second edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989). Numerous plasmids with suitable promoter sequences for expression in bacteria and efficient ribosome binding sites have been described, such as for example, pKC30 (λL: Shimatake and Rosenberg, Nature 292, 128, 1981); pKK173-3 (tac: Amann and Brosius, Gene 40, 183, 1985), pET-3 (T7: Studier and Moffat, J. Mol. Biol. 189, 113, 1986); the pBAD/TOPO or pBAD/Thio-TOPO series of vectors containing an arabinose-inducible promoter (Invitrogen, Carlsbad, Calif.), the latter of which is designed to also produce fusion proteins with thioredoxin to enhance solubility of the expressed protein; the pFLEX series of expression vectors (Pfizer Inc., CT, USA); or the pQE series of expression vectors (Qiagen, Calif.), amongst others.


Typical promoters suitable for expression in viruses of eukaryotic cells and eukaryotic cells include the SV40 late promoter, SV40 early promoter and cytomegalovirus (CMV) promoter, CMV IE (cytomegalovirus immediate early) promoter amongst others. Preferred vectors for expression in mammalian cells (eg. 293, COS, CHO, 293T cells) include, but are not limited to, the pcDNA Vector suite supplied by Invitrogen, in particular pcDNA 3.1 myc-His-tag comprising the CMV promoter and encoding a C-terminal 6xHis and MYC tag; and the retrovirus vector pSRαtkneo (Muller et al., Mol. Cell. Biol., 11, 1785, 1991). The vector pcDNA 3.1 myc-His (Invitrogen) is particularly preferred for expressing a secreted form of a protein in 293T cells, wherein the expressed peptide or protein can be purified free of conspecific proteins, using standard affinity techniques that employ a Nickel column to bind the protein via the His tag.


A wide range of additional host/vector systems suitable for expressing polypeptides or immunological derivatives thereof are available publicly, and described, for example, in Sambrook et al (In: Molecular cloning. A laboratory manual, second edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989).


Means for introducing the isolated nucleic acid molecule or a gene construct comprising same into a cell for expression are well-known to those skilled in the art. The technique used for a given organism depends on the known successful techniques. Means for introducing recombinant DNA into animal cells include microinjection, transfection mediated by DEAE-dextran, transfection mediated by liposomes such as by using lipofectamine (Gibco, Md., USA) and/or cellfectin (Gibco, Md., USA), PEG-mediated DNA uptake, electroporation and microparticle bombardment such as by using DNA-coated tungsten or gold particles (Agracetus Inc., WI, USA) amongst others.


For producing mutants, nucleotide insertion derivatives of the protein-encoding region are produced by making 5′ and 3′ terminal fusions, or by making intra-sequence insertions of single or multiple nucleotides or nucleotide analogues. Insertion nucleotide sequence variants are produced by introducing one or more nucleotides or nucleotide analogues into a predetermined site in the nucleotide sequence of said sequence, although random insertion is also possible with suitable screening of the resulting product being performed. Deletion variants are produced by removing one or more nucleotides from the nucleotide sequence. Substitutional nucleotide variants are produced by substituting at least one nucleotide in the sequence with a different nucleotide or a nucleotide analogue in its place, with the immunologically active derivative encoded therefor having an identical amino acid sequence, or only a limited number of amino acid modifications that do not alter its antigenicity compared to the base peptide or its ability to bind antibodies prepared against the base peptide. Such mutant derivatives will preferably have at least 80% identity With the base amino acid sequence from which they are derived.


Preferred immunologically active derivatives of a full-length polypeptide encoded by a gene referred to in any one of Tables 1-3 will comprise at least about 5-10 contiguous amino acids of the full-length amino acid sequence, more preferably at least about 10-20 contiguous amino acids in length, and even more preferably 20-30 contiguous amino acids in length.


For the purposes of producing derivatives using standard peptide synthesis techniques, such as, for example, Fmoc chemistry, a length not exceeding about 30-50 amino acids in length is preferred, as longer peptides are difficult to produce at high efficiency. Longer peptide fragments are readily achieved using recombinant DNA techniques wherein the peptide is expressed in a cell-free or cellular expression system comprising nucleic acid encoding the desired peptide fragment.


It will be apparent to the skilled artisan that any sufficiently antigenic region of at least about 5-10 amino acid residues can be used to prepare antibodies that bind generally to the polypeptides listed in Tables 1-3.


An expressed protein or synthetic peptide is preferably produced as a recombinant fusion protein, such as for example, to aid in extraction and purification. To produce a fusion polypeptide, the open reading frames are covalently linked in the same reading frame, such as, for example, using standard cloning procedures as described by Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, ISBN 047150338, 1992), and expressed under control of a promoter. Examples of fusion protein partners include glutathione-5-transferase (GST), FLAG, hexahistidine, GAL4 (DNA binding and/or transcriptional activation domains) and β-galactosidase. It may also be convenient to include a proteolytic cleavage site between the fusion protein partner and the protein sequence of interest to allow removal of fusion protein sequences. Preferably the fusion protein will not hinder the immune function of the target protein.


In a particularly preferred embodiment, polypeptides are produced substantially free of conspecific proteins. Such purity can be assessed by standard procedures, such as, for example, SDS/polyacrylamide gel electrophoresis, 2-dimensional gene electrophoresis, chromatography, amino acid composition analysis, or amino acid sequence analysis.


To produce isolated polypeptides or fragments, eg., for antibody production, standard protein purification techniques may be employed. For example, gel filtration, ion exchange chromatography, reverse phase chromatography, or affinity chromatography, or a combination of any one or more said procedures, may be used. High pressure and low pressure procedures can also be employed, such as, for example, FPLC, or HPLC. To isolate the full-length proteins or peptide fragments comprising more than about 50-100 amino acids in length, it is particularly preferred to express the polypeptide in a suitable cellular expression system in combination with a suitable affinity tag, such as a 6xHis tag, and to purify the polypeptide using an affinity step that bonds it via the tag (supra). Optionally, the tag may then be cleaved from the expressed polypeptide.


Alternatively, for short immunologically active derivatives of a full-length polypeptide, preferably those peptide fragments comprising less than about 50 amino acids in length, chemical synthesis techniques are conveniently used. As will be known to those skilled in the art, such techniques may also produce contaminating peptides that are shorter than the desired peptide, in which case the desired peptide is conveniently purified using reverse phase and/or ion exchange chromatography procedures at high pressure (ie. HPLC or FPLC).


Antibodies


The invention also provides monoclonal or polyclonal antibodies that bind specifically to polypeptides of the invention or fragments thereof. Thus, the present invention further provides a process for the production of monoclonal or polyclonal antibodies to polypeptides of the invention.


The phrase “binds specifically” to a polypeptide means that the binding of the antibody to the protein or peptide 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 the background and more typically more than 10 to 100 times background. Typically, antibodies of the invention bind to a protein of interest with a Kd of at least about 0.1 mM, more usually at least about 1 μM, preferably at least about 0.1 μM, and most preferably at least, 0.01 μM.


Reference herein to antibody or antibodies includes whole polyclonal and monoclonal antibodies, and parts thereof, either alone or conjugated with other moieties. Antibody parts include Fab and F(ab)2 fragments and single chain antibodies. The antibodies may be made in vivo in suitable laboratory animals, or, in the case of engineered antibodies (Single Chain Antibodies or SCABS, etc) using recombinant DNA techniques in vitro.


In accordance with this aspect of the invention, the antibodies may be produced for the purposes of immunizing the subject, in which case high titer or neutralizing antibodies that bind to a B cell epitope will be especially preferred. Suitable subjects for immunization will, of course, depend upon the immunizing antigen or antigenic B cell epitope. It is contemplated that the present invention will be broadly applicable to the immunization of a wide range of animals, such as, for example, farm animals (e.g. horses, cattle, sheep, pigs, goats, chickens, ducks, turkeys, and the like), laboratory animals (e.g. rats, mice, guinea pigs, rabbits), domestic animals (cats, dogs, birds and the like), feral or wild exotic animals (e.g. possums, cats, pigs, buffalo, wild dogs and the like) and humans.


Alternatively, the antibodies may be for commercial or diagnostic purposes, in which case the subject to whom the diagnostic/prognostic protein or immunogenic fragment or epitope thereof is administered will most likely be a laboratory or farm animal. A wide range of animal species are used for the production of antisera. Typically the animal used for production of antisera is a rabbit, a mouse, rat, hamster, guinea pig, goat, sheep, pig, dog, horse, or chicken. Because of the relatively large blood volume of rabbits, a rabbit is a preferred choice for production of polyclonal antibodies. However, as will be known to those skilled tin the art, larger amounts of immunogen are required to obtain high antibodies from large animals as opposed to smaller animals such as mice. In such cases, it will be desirable to isolate the antibody from the immunized animal.


Preferably, the antibody is a high titer antibody. By “high titer” means a sufficiently high titer to be suitable for use in diagnostic or therapeutic applications. As will be known in the art, there is some variation in what might be considered “high titer”. For most applications a titer of at least about 103-104 is preferred. More preferably, the antibody titer will be in the range from about 1 to about 105, even more preferably in the range from about 105 to about 106.


More preferably, in the case of B cell epitopes from pathogens, viruses or bacteria, the antibody is a neutralizing antibody (i.e. it is capable of neutralizing the infectivity of the organism fro which the B cell epitope is derived).


To generate antibodies, the diagnostic/prognostic protein or immunogenic fragment or epitope thereof, optionally formulated with any suitable or desired carrier, adjuvant, BRM, or pharmaceutically acceptable excipient, is conveniently administered in the form of an injectable composition. Injection may be intranasal, intramuscular, sub-cutaneous, intravenous, intradermal, intraperitoneal, or by other known route. For intravenous injection, it is desirable to include one or more fluid and nutrient replenishers. Means for preparing and characterizing antibodies are well known in the art. (See, e.g., ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor Laboratory, 1988, incorporated herein by reference).


The efficacy of the diagnostic/prognostic protein or immunogenic fragment or epitope thereof in producing an antibody is established by injecting an animal, for example, a mouse, rat, rabbit, guinea pig, dog, horse, cow, goat or pig, with a formulation comprising the diagnostic/prognostic protein or immunogenic fragment or epitope thereof, and then monitoring the immune response to the B cell epitope, as described in the Examples. Both primary and secondary immune responses are monitored. The antibody titer is determined using any conventional immunoassay, such as, for example, ELISA, or radio immunoassay.


The production of polyclonal antibodies may be monitored by sampling blood of the immunized animal at various points following immunization. A second, booster injection, may be given, if required to achieve a desired antibody titer. The process of boosting and titering is repeated until a suitable titer is achieved. When a desired level of immunogenicity is obtained, the immunized animal is bled and the serum isolated and stored, and/or the animal is used to generate monoclonal antibodies (Mabs).


For the production of monoclonal antibodies (Mabs) any one of a number of well-known techniques may be used, such as, for example, the procedure exemplified in U.S. Pat. No. 4,196,265, incorporated herein by reference.


For example, a suitable animal will be immunized with an effective amount of the diagnostic/prognostic protein or immunogenic fragment or epitope thereof under conditions sufficient to stimulate antibody producing cells. Rodents such as mice and rats are preferred animals, however, the use of rabbit, sheep, or frog cells is also possible. The use of rats may provide certain advantages, but mice are preferred, with the BALB/c mouse being most preferred as the most routinely used animal and one that generally gives a higher percentage of stable fusions.


Following immunization, somatic cells with the potential for producing antibodies, specifically B lymphocytes (B cells), are selected for use in the MAb generating protocol. These cells may be obtained from biopsied spleens, tonsils or lymph nodes, or from a peripheral blood sample. Spleen cells and peripheral blood cells are preferred, the former because they are a rich source of antibody-producing cells that are in the dividing plasmablast stage, and the latter because peripheral blood is easily accessible. Often, a panel of animals will have been immunized and the spleen of animal with the highest antibody titer removed. Spleen lymphocytes are obtained by homogenizing the spleen with a syringe. Typically, a spleen from an immunized mouse contains approximately 5×107 to 2×108 lymphocytes.


The B cells from the immunized animal are then fused with cells of an immortal myeloma cell, generally derived from the same species as the animal that was immunized with the diagnostic/prognostic protein or immunogenic fragment or epitope thereof. Myeloma cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency and enzyme deficiencies that render them incapable of growing in certain selective media which support the growth of only the desired fused cells, or hybridomas. Any one of a number of myeloma cells may be used and these are known to those of skill in the art (e.g. murine P3-X63/Ag8, X63-Ag8.653, NS1/1.Ag 41, Sp210-Ag14, FO, NSO/U, MPC11, MPC11-X45-GTG 1.7 and S194/5XX0; or rat R210.RCY3, Y3-Ag 1.2.3, IR983F and 4B210; and U-266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6). A preferred murine myeloma cell is the NS-1 myeloma cell line (also termed P3-NS-1-Ag4-1), which is readily available from the NIGMS Human Genetic Mutant Cell Repository under Accession No. GM3573. Alternatively, a murine myeloma SP2/0 non-producer cell line that is 8-azaguanine-resistant is used.


To generate hybrids of antibody-producing spleen or lymph node cells and myeloma cells, somatic cells are mixed with myeloma cells in a proportion between about 20:1 to about 1:1, respectively, in the presence of an agent or agents (chemical or electrical) that promote the fusion of cell membranes. Fusion methods using Sendai virus have been described by Kohler and Milstein, Nature 256, 495-497, 1975; and Kohler and Milstein, Eur. J. Immunol. 6, 511-519, 1976. Methods using polyethylene glycol (PEG), such as 37% (v/v) PEG, are described in detail by Gefter et al., Somatic Cell Genet 3, 231-236, 1977. The use of electrically induced fusion methods is also appropriate.


Hybrids are amplified by culture in a selective medium comprising an agent that blocks the de novo synthesis of nucleotides in the tissue culture media. Exemplary and preferred agents are aminopterin, methotrexate and azaserine. Aminopterin and methotrexate block de novo synthesis of both purines and pyrimidines, whereas azaserine blocks only purine synthesis. Where aminopterin or methotrexate is used, the media is supplemented with hypoxanthine and thymidine as a source of nucleotides (HAT medium). Where azaserine is used, the media is supplemented with hypoxanthine.


The preferred selection medium is HAT, because only those hybridomas capable of operating nucleotide salvage pathways are able to survive in HAT medium, whereas myeloma cells are defective in key enzymes of the salvage pathway, (e.g., hypoxanthine phosphoribosyl transferase or HPRT), and they cannot survive. B cells can operate this salvage pathway, but they have a limited life span in culture and generally die within about two weeks. Accordingly, the only cells that can survive in the selective media are those hybrids formed from myeloma and B cells.


The amplified hybridomas are subjected to a functional selection for antibody specificity and/or titer, such as, for example, by immunoassay (e.g. radioimmunoassay, enzyme immunoassay, cytotoxicity assay, plaque assay, dot immunobinding assay, and the like).


The selected hybridomas are serially diluted and cloned into individual antibody-producing cell lines, which clones can then be propagated indefinitely to provide MAbs. The cell lines may be exploited for MAb production in two basic ways. A sample of the hybridoma is injected, usually in the peritoneal cavity, into a histocompatible animal of the type that was used to provide the somatic and myeloma cells for the original fusion. The injected animal develops tumors secreting the specific monoclonal antibody produced by the fused cell hybrid. The body fluids of the animal, such as serum or ascites fluid, can then be tapped to provide MAbs in high concentration. The individual cell lines could also be cultured in vitro, where the MAbs are naturally secreted into the culture medium from which they are readily obtained in high concentrations. MAbs produced by either means may be further purified, if desired, using filtration, centrifugation and various chromatographic methods such as HPLC or affinity chromatography.


Monoclonal antibodies of the present invention also include anti-idiotypic antibodies produced by methods well-known in the art. Monoclonal antibodies according to the present invention also may be monoclonal heteroconjugates, (i.e., hybrids of two or more antibody molecules). In another embodiment, monoclonal antibodies according to the invention are chimeric monoclonal antibodies. In one approach, the chimeric monoclonal antibody is engineered by cloning recombinant DNA containing the promoter, leader, and variable-region sequences from a mouse anti-PSA producing cell and the constant-region exons from a human antibody gene. The antibody encoded by such a recombinant gene is a mouse-human chimera. Its antibody specificity is determined by the variable region derived from mouse sequences. Its isotype, which is determined by the constant region, is derived from human DNA.


In another embodiment, the monoclonal antibody according to the present invention is a “humanized” monoclonal antibody, produced by any one of a number of techniques well-known in the art. That is, mouse complementary determining regions (“CDRs”) are transferred from heavy and light V-chains of the mouse Ig into a human V-domain, followed by the replacement of some human residues in the framework regions of their murine counterparts. “Humanized” monoclonal antibodies in accordance with this invention are especially suitable for use in vivo in diagnostic and therapeutic methods.


As stated above, the monoclonal antibodies and fragments thereof according to this invention are multiplied according to in vitro and in vivo methods well-known in the art. Multiplication in vitro is carried out in suitable culture media such as Dulbecco's modified Eagle medium or RPMI 1640 medium, optionally replenished by a mammalian serum such as fetal calf serum or trace elements and growth-sustaining supplements, e.g., feeder cells, such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages or the like. In vitro production provides relatively pure antibody preparations and allows scale-up to give large amounts of the desired antibodies. Techniques for large scale hybridoma cultivation under tissue culture conditions are known in the art and include homogenous suspension culture, (e.g., in an airlift reactor or in a continuous stirrer reactor or immobilized or entrapped cell culture).


Large amounts of the monoclonal antibody of the present invention also may be obtained by multiplying hybridoma cells in vivo. Cell clones are injected into mammals which are histocompatible with the parent cells, (e.g., syngeneic mice, to cause growth of antibody-producing tumors. Optionally, the animals are primed with a hydrocarbon, especially oils such as Pristane (tetramethylpentadecane) prior to injection.


In accordance with the present invention, fragments of the monoclonal antibody of the invention are obtained from monoclonal antibodies produced as described above, by methods which include digestion with enzymes such as pepsin or papain and/or cleavage of disulfide bonds by chemical reduction. Alternatively, monoclonal antibody fragments encompassed by the present invention are synthesized using an automated peptide synthesizer, or they may be produced manually using techniques well known in the art.


The monoclonal conjugates of the present invention are prepared by methods known in the art, e.g., by reacting a monoclonal antibody prepared as described above with, for instance, an enzyme in the presence of a coupling agent such as glutaraldehyde or periodate. Conjugates with fluorescein markers are prepared in the presence of these coupling agents, or by reaction with an Isothiocyanate. Conjugates with metal chelates are similarly produced. Other moieties to which antibodies may be conjugated include radionuclides such as, for example, 3H, 125I, 32P, 35S, 14C, 51Cr, 36Cl, 57Co, 58Co, 59Fe, 75Se, and 152Eu.


Radioactively labeled monoclonal antibodies of the present invention are produced according to well-known methods in the art. For instance, monoclonal antibodies are iodinated by contact with sodium or potassium iodide and a chemical oxidizing agent such as sodium hypochlorite, or an enzymatic oxidizing agent, such as lactoperoxidase. Monoclonal antibodies according to the invention may be labeled with technetium99 by ligand exchange process, for example, by reducing pertechnetate with stannous solution, chelating the reduced technetium onto a Sephadex column and applying the antibody to this column or by direct labeling techniques, (e.g., by incubating pertechnate, a reducing agent such as SNCl2, a buffer solution such as sodium-potassium phthalate solution, and the antibody).


Any immunoassay may be used to monitor antibody production by the diagnostic/prognostic protein or immunogenic fragment or epitope thereof. Immunoassays, in their most, simple and direct sense, are binding assays. Certain preferred immunoassays are the various types of enzyme linked immunosorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful. However, it will be readily appreciated that detection is not limited to such techniques, and Western blotting, dot blotting, FACS analyses, and the like may also be used.


Most preferably, the assay will be capable of generating quantitative results.


For example, antibodies are tested in simple competition assays. A known antibody preparation that binds to the B cell epitope and the test antibody are incubated with an antigen composition comprising the B cell epitope, preferably in the context of the native antigen. “Antigen composition” as used herein means any composition that contains some version of the B cell epitope in an accessible form. Antigen-coated wells of an ELISA plate are particularly preferred. In one embodiment, one would pre-mix the known antibodies with varying amounts of the test antibodies (e.g., 1:1, 1:10 and 1:100) for a period of time prior to applying to the antigen composition. If one of the known antibodies is labeled, direct detection of the label bound to the antigen is possible; comparison to an unmixed sample assay will determine competition by the test antibody and, hence, cross-reactivity.


Alternatively, using secondary antibodies specific for either the known or test antibody, one will be able to determine competition.


An antibody that binds to the antigen composition will be able to effectively compete for binding of the known antibody and thus will significantly reduce binding of the latter. The reactivity of the known antibodies in the absence of any test antibody is the control. A significant reduction in reactivity in the presence of a test antibody is indicative of a test antibody that binds to the B cell epitope (i.e., it cross-reacts with the known antibody).


In one exemplary ELISA, the antibodies against the diagnostic/prognostic protein or immunogenic fragment or B cell epitope are immobilized onto a selected surface exhibiting protein affinity, such as a well in a polystyrene microtiter plate. Then, a composition containing a peptide comprising the B cell epitope is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound epitope may be detected. Detection is generally achieved by the addition of a second antibody that is known to bind to the B cell epitope and is linked to a detectable label. This type of ELISA is a simple “sandwich ELISA”. Detection may also be achieved by the addition of said second antibody, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label.


Antibodies of the invention may be bound to a solid support and/or packaged into kits in a suitable container along with suitable reagents, controls, instructions and the like.


Immunoassay Formats


In one embodiment, a cancer-associated protein or an immunogenic fragment or epitope thereof is detected in a patient sample, wherein the level of the protein or immunogenic fragment or epitope in the sample is indicative of ovarian cancer or disease recurrence or an indicator of poor survival. Preferably, the method comprises contacting a biological sample derived from the subject with an antibody capable of binding to a cancer-associated protein or an immunogenic fragment or epitope thereof, and detecting the formation of an antigen-antibody complex.


In another embodiment, an antibody against a cancer-associated protein or epitope thereof is detected in a patient sample, wherein the level of the antibody in the sample is indicative of ovarian cancer or disease recurrence or an indicator of poor survival.


Preferably, the method comprises contacting a biological sample derived from the subject with a cancer-associated protein or an antigenic fragment eg., a B cell epitope or other immunogenic fragment thereof, and detecting the formation of an antigen-antibody complex.


The diagnostic assays of the invention are useful for determining the progression of ovarian cancer or a metastasis thereof in a subject. In accordance with these prognostic applications of the invention, the level of a cancer-associated protein or an immunogenic fragment or epitope thereof in a biological sample is correlated with the disease state eg., as determined by clinical symptoms or biochemical tests (eg., CA125 levels).


Accordingly, a further embodiment of the invention provides a method for detecting a cancer cell in a subject, said method comprising:

  • (i) determining the level of a cancer-associate protein in a test sample from said subject; and
  • (ii) comparing the level determined at (i) to the level of said cancer-associated protein in a comparable sample from a healthy or normal individual,


    wherein a level of said cancer-associate protein at (i) that is modified in the test sample relative to the comparable sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.


In one embodiment of the diagnostic/prognostic methods described herein, the biological sample is obtained previously from the subject. In accordance with such an embodiment, the prognostic or diagnostic method is performed ex vivo.


In yet another embodiment, the subject diagnostic/prognostic methods further comprise processing the sample from the subject to produce a derivative or extract that comprises the analyte.


Preferred detection systems contemplated herein include any known assay for detecting proteins or antibodies in a biological sample isolated from a human subject, such as, for example, SDS/PAGE, isoelectric focussing, 2-dimensional gel electrophoresis comprising SDS/PAGE and isoelectric focussing, an immunoassay, a detection based system using an antibody or non-antibody ligand of the protein, such as, for example, a small molecule (e.g. a chemical compound, agonist, antagonist, allosteric modulator, competitive inhibitor, or non-competitive inhibitor, of the protein). In accordance with these embodiments, the antibody or small molecule may be used in any standard solid phase or solution phase assay format amenable to the detection of proteins. Optical or fluorescent detection, such as, for example, using mass spectrometry, MALDI-TOF, biosensor technology, evanescent fiber optics, or fluorescence resonance energy transfer, is clearly encompassed by the present invention. Assay systems suitable for use in high throughput screening of mass samples, particularly a high throughput spectroscopy resonance method (e.g. MALDI-TOF, electrospray MS or nano-electrospray MS), are particularly contemplated.


Immunoassay formats are particularly preferred, eg., selected from the group consisting of, an immunoblot, a Western blot, a dot blot, an enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), enzyme immunoassay. Modified immunoassays utilizing fluorescence resonance energy transfer (FRET), isotope-coded affinity tags (ICAT), matrix-assisted laser desorption/ionization time of flight (MALDI-TOF), electrospray ionization (ESI), biosensor technology, evanescent fiber-optics technology or protein chip technology are also useful.


Preferably, the assay is a semi-quantitative assay or quantitative assay.


Standard solid phase ELISA formats are particularly useful in determining the concentration of a protein or antibody from a variety of patient samples.


In one form such as an assay involves immobilising a biological sample comprising antibodies against the cancer-associated protein or epitope, or alternatively an ovarian cancer-associated protein or an immunogenic fragment thereof, onto a solid matrix, such as, for example a polystyrene or polycarbonate microwell or dipstick, a membrane, or a glass support (e.g. a glass slide).


In the case of an antigen-based assay, an antibody that specifically binds an ovarian cancer-associated protein is brought into direct contact with the immobilised biological sample, and forms a direct bond with any of its target protein present in said sample. For an antibody-based assay, an immobilized ovarian cancer-associated protein or an immunogenic fragment or epitope thereof is contacted with the sample. The added antibody or protein in solution is generally labelled with a detectable reporter molecule, such as for example, a fluorescent label (e.g. FITC or Texas Red) or an enzyme (e.g. horseradish peroxidase (HRP)), alkaline phosphatase (AP) or β-galactosidase.


Alternatively, or in addition, a second labelled antibody can be used that binds to the first antibody or to the isolated/recombinant antigen. Following washing to remove any unbound antibody or antigen, as appropriate, the label is detected either directly, in the case of a fluorescent label, or through the addition of a substrate, such as for example hydrogen peroxide, TMB, or toluidine, or 5-bromo-4-chloro-3-indol-beta-D-galaotopyranoside (x-gal).


Such ELISA based systems are particularly suitable for quantification of the amount of a protein or antibody in a sample, such as, for example, by calibrating the detection system against known amounts of a standard.


In another form, an ELISA consists of immobilizing an antibody that specifically binds an ovarian cancer-associated protein on a solid matrix, such as, for example, a membrane, a polystyrene or polycarbonate microwell, a polystyrene or polycarbonate dipstick or a glass support. A patient sample is then brought into physical relation with said antibody, and the antigen in the sample is bound or ‘captured’. The bound protein can then be detected using a labelled antibody. For example if the protein is captured from a human sample, an anti-human antibody is used to detect the captured protein. Alternatively, a third labelled antibody can be used that binds the second (detecting) antibody.


It will be apparent to the skilled person that the assay formats described herein are amenable to high throughput formats, such as, for example automation of screening processes, or a microarray format as described in Mendoza et al, Biotechniques 27(4): 778-788, 1999. Furthermore, variations of the above described assay will be apparent to those skilled in the art, such as, for example, a competitive ELISA.


Alternatively, the presence of antibodies against the cancer-associate protein, or alternatively an oarian cancer-associated protein or an immunogenic fragment thereof, is detected using a radioimmunoassay (RIA). The basic principle of the assay is the use of a radiolabelled antibody or antigen to detect antibody antigen interactions. For example, an antibody that specifically binds to an ovarian cancer-associated protein can be bound to a solid support and a biological sample brought into direct contact with said antibody. To detect the bound antigen, an isolated and/or recombinant form of the antigen is radiolabelled is brought into contact with the same antibody. Following washing the amount of bound radioactivity is detected. As any antigen in the biological sample inhibits binding of the radiolabelled antigen the amount of radioactivity detected is inversely proportional to the amount of antigen in the sample. Such an assay may be quantitated by using a standard curve using increasing known concentrations of the isolated antigen.


As will be apparent to the skilled artisan, such an assay may be modified to use any reporter molecule, such as, for example, an enzyme or a fluorescent molecule, in place of a radioactive label.


Western blotting is also useful for detecting an ovarian cancer-associated protein or an immunogenic fragment thereof. In such an assay protein from a biological sample is separated using sodium dodecyl sulphate (SDS) polyacrylamide gel electrophoresis (SDS-PAGE) using techniques well known in the art and described in, for example, Scopes (In: Protein Purification: Principles and Practice, Third Edition, Springer Verlag, 1994). Separated proteins are then transferred to a solid support, such as, for example, a membrane or more specifically PVDF membrane, using methods well known in the art, for example, electrotransfer. This membrane may then be blocked and probed with a labelled antibody or ligand that specifically binds an ovarian cancer-associated protein. Alternatively, a labelled secondary, or even tertiary, antibody or ligand can be used to detect the binding of a specific primary antibody.


High-throughput methods for detecting the presence or absence of antibodies, or alternatively ovarian cancer-associated protein or an immunogenic fragment thereof are particularly preferred.


In one embodiment, MALDI-TOF is used for the rapid identification of a protein. Accordingly, there is no need to detect the proteins of interest using an antibody or ligand that specifically binds to the protein of interest. Rather, proteins from a biological sample are separated using gel electrophoresis using methods well known in the art and those proteins at approximately the correct molecular weight and/or isoelectric point are analysed using MALDI-TOF to determine the presence or absence of a protein of interest.


Alternatively, MALDI or ESI or a combination of approaches is used to determine the concentration of a particular protein in a biological sample, such as, for example sputum. Such proteins are preferably well characterised previously with regard to parameters such as molecular weight and isoelectric point.


Biosensor devices generally employ an electrode surface in combination with current or impedance measuring elements to be integrated into a device in combination with the assay substrate (such as that described in U.S. Pat. No. 5,567,301). An antibody or ligand that specifically binds to a protein of interest is preferably incorporated onto the surface of a biosensor device and a biological sample isolated from a patient (for example sputum that has been solubilised using the methods described herein) contacted to said device. A change in the detected current or impedance by the biosensor device indicates protein binding to said antibody or ligand. Some forms of biosensors known in the art also rely on surface plasmon resonance to detect protein interactions, whereby a change in the surface plasmon resonance surface of reflection is indicative of a protein binding to a ligand or antibody (U.S. Pat. Nos. 5,485,277 and 5,492,840).


Biosensors are of particular use in high throughput analysis due to the ease of adapting such systems to micro- or nano-scales. Furthermore, such systems are conveniently adapted to incorporate several detection reagents, allowing for multiplexing of diagnostic reagents in a single biosensor unit. This permits the simultaneous detection of several epitopes in a small amount of body fluids.


Evanescent biosensors are also preferred as they do not require the pretreatment of a biological sample prior to detection of a protein of interest. An evanescent biosensor generally relies upon light of a predetermined wavelength interacting with a fluorescent molecule, such as for example, a fluorescent antibody attached near the probe's surface, to emit fluorescence at a different wavelength upon binding of the diagnostic protein to the antibody or ligand.


To produce protein chips, the proteins, peptides, polypeptides, antibodies or ligands that are able to bind specific antibodies or proteins of interest are bound to a solid support such as for example glass, polycarbonate, polytetrafluoroethylene, polystyrene, silicon oxide, metal or silicon nitride. This immobilization is either direct (e.g. by covalent linkage, such as, for example, Schiff's base formation, disulfide linkage, or amide or urea bond formation) or indirect. Methods of generating a protein chip are known in the art and are described in for example U.S. Patent Application No. 20020136821, 20020192654, 20020102617 and U.S. Pat. No. 6,391,625. In order to bind a protein to a solid support it is often necessary to treat the solid support so as to create chemically reactive groups on the surface, such as, for example, with an aldehyde-containing silane reagent.


Alternatively, an antibody or ligand may be captured on a microfabricated polyacrylamide gel pad and accelerated into the gel using microelectrophoresis as described in, Arenkov et al. Anal. Biochem. 278:123-131, 2000.


A protein chip is preferably generated such that several proteins, ligands or antibodies are arrayed on said chip. This format permits the simultaneous screening for the presence of several proteins in a sample.


Alternatively, a protein chip may comprise only one protein, ligand or antibody, and be used to screen one or more patient samples for the presence of one polypeptide of interest. Such a chip may also be used to simultaneously screen an array of patient samples for a polypeptide of interest.


Preferably, a sample to be analysed using a protein chip is attached to a reporter molecule, such as, for example, a fluorescent molecule, a radioactive molecule, an enzyme, or an antibody that is detectable using methods well known in the art. Accordingly, by contacting a protein chip with a labelled sample and subsequent washing to remove any unbound proteins the presence of a bound protein is detected using methods well known in the art, such as, for example using a DNA microarray reader.


Alternatively, biomolecular interaction analysis-mass spectrometry (BIA-MS) is used to rapidly detect and characterise a protein present in complex biological samples at the low- to sub-fmole level (Nelson et al. Electrophoresis 21: 1155-1163, 2000). One technique useful in the analysis of a protein chip is surface enhanced laser desorption/ionization-time of flight-mass spectrometry (SELDI-TOF-MS) technology to characterise a protein bound to the protein chip. Alternatively, the protein chip is analysed using ESI as described in U.S. Patent Application 20020139751.


As will be apparent to the skilled artisan, protein chips are particularly amenable to multiplexing of detection reagents. Accordingly, several antibodies or ligands each able to specifically bind a different peptide or protein may be bound to different regions of said protein chip. Analysis of a biological sample using said chip then permits the detecting of multiple proteins of interest, or multiple B cell epitopes of the ovarian cancer-associated protein. Multiplexing of diagnostic and prognostic markers is particularly contemplated in the present invention.


In a further embodiment, the samples are analysed using ICAT, essentially as described in US Patent Application No. 20020076739. This system relies upon the labelling of a protein sample from one source (i.e. a healthy individual) with a reagent and the labelling of a protein sample from another source (i.e. a tuberculosis patient) with a second reagent that is chemically identical to the first reagent, but differs in mass due to isotope composition. It is preferable that the first and second reagents also comprise a biotin molecule. Equal concentrations of the two samples are then mixed, and peptides recovered by avidin affinity chromatography. Samples are then analysed using mass spectrometry. Any difference in peak heights between the heavy and light peptide ions directly correlates with a difference in protein abundance in a biological sample. The identity of such proteins may then be determined using a method well known in the art, such as, for example MALDI-TOF, or ESI.


As will be apparent to those skilled in the art a diagnostic or prognostic assay described herein may be a multiplexed assay. As used herein the term “multiplex”, shall be understood not only to mean the detection of two or more diagnostic or prognostic markers in a single sample simultaneously, but also to encompass consecutive detection of two or more diagnostic or prognostic markers in a single sample, simultaneous detection of two or more diagnostic or prognostic markers in distinct but matched samples, and consecutive detection of two or more diagnostic or prognostic markers in distinct but matched samples. As used herein the term “matched samples” shall be understood to mean two or more samples derived from the same initial biological sample, or two or more biological samples isolated at the same point in time.


Accordingly, a multiplexed assay may comprise an assay that detects several antibodies and/or epitopes in the same reaction and simultaneously, or alternatively, it may detect other one or more antigens/antibodies in addition to one or more antibodies and/or epitopes. As will be apparent to the skilled artisan, if such an assay is antibody or ligand based, both of these antibodies must function under the same conditions.


Diagnostic Assay Kits


A further aspect of the present invention provides a kit for detecting M. tuberculosis infection in a biological sample. In one embodiment, the kit comprises:

  • (i) one or more isolated antibodies that bind to an ovarian cancer-associated protein or an immunogenic fragment or epitope thereof; and
  • (ii) means for detecting the formation of an antigen-antibody complex.


In an alternative embodiment, the kit comprises:

  • (i) an isolated or recombinant ovarian cancer-associated protein or an immunogenic fragment or epitope thereof; and
  • (ii) means for detecting the formation of an antigen-antibody complex.


Optionally, the kit further comprises means for the detection of the binding of an antibody, fragment thereof or a ligand to an ovarian cancer-associated protein. Such means include a reporter molecule such as, for example, an enzyme (such as horseradish peroxidase or alkaline phosphatase), a substrate, a cofactor, an inhibitor, a dye, a radionucleotide, a luminescent group, a fluorescent group, biotin or a colloidal particle, such as colloidal gold or selenium. Preferably such a reporter molecule is directly linked to the antibody or ligand.


In yet another embodiment, a kit may additionally comprise a reference sample. Such a reference sample.


In another embodiment, a reference sample comprises a peptide that is detected by an antibody or a ligand. Preferably, the peptide is of known concentration. Such a peptide is of particular use as a standard. Accordingly various known concentrations of such a peptide may be detected using a prognostic or diagnostic assay described herein.


In yet another embodiment, a kit comprises means for protein isolation (Scopes (In: Protein Purification: Principles and Practice, Third Edition, Springer Verlag, 1994).


Bioinformatics


The ability to identify genes that are over or under expressed in ovarian cancer can additionally provide high-resolution, high-sensitivity datasets which are used in the areas of diagnostics, therapeutics, drug development, pharmacogenetics, protein structure, biosensor development, and other related areas. For example, the expression profiles are used in diagnostic or prognostic evaluation of patients with ovarian cancer. Or as another example, subcellular toxicological information are generated to better direct drug structure and activity correlation (see Anderson, Pharmaceutical Proteomics: Targets, Mechanism, and Function, paper presented at the IBC Proteomics conference, Coronado, Calif. (Jun. 11-12, 1998)). Subcellular toxicological information can also be utilized in a biological sensor device to predict the likely toxicological effect of chemical exposures and likely tolerable exposure thresholds (see U.S. Pat. No. 5,811,231). Similar advantages accrue from datasets relevant to other biomolecules and bioactive agents (e.g., nucleic acids, saccharides, lipids, drugs, and the like).


Thus, in another embodiment, the present invention provides a database that includes at least one set of assay data. The data contained in the database is acquired, e.g., using array analysis either singly or in a library format. The database are in substantially any form in which data are maintained and transmitted, but is preferably an electronic database. The electronic database of the invention are maintained on any electronic device allowing for the storage of and access to the database, such as a personal computer, but is preferably distributed on a wide area network, such as the World Wide Web.


The focus of the present section on databases that include peptide sequence data is for clarity of illustration only. It, will be apparent to those of skill in the art that similar databases are assembled for any assay data acquired using an assay of the invention.


The compositions and methods for identifying and/or quantitating the relative and/or absolute abundance of a variety of molecular and macromolecular species from a biological sample undergoing ovarian cancer, i.e., the identification of ovarian cancer-associated sequences described herein, provide an abundance of information, which are correlated with pathological conditions, predisposition to disease, drug testing, therapeutic monitoring, gene-disease causal linkages, identification of correlates of immunity and physiological status, among others. Although the data generated from the assays of the invention is suited for manual review and analysis, in a preferred embodiment, prior data processing using high-speed computers is utilized.


An array of methods for indexing and retrieving biomolecular information is known in the art. For example, U.S. Pat. Nos. 6,023,659 and 5,966,712 disclose a relational database system for storing biomolecular sequence information in a manner that allows sequences to be catalogued and searched according to one or more protein function hierarchies. U.S. Pat. No. 5,953,727 discloses a relational database having sequence records containing information in a format that allows a collection of partial-length DNA sequences to be catalogued and searched according to association with one or more sequencing projects for obtaining full-length sequences from the collection of partial length sequences. U.S. Pat. No. 5,706,498 discloses a gene database retrieval system for making a retrieval of a gene sequence similar to a sequence data item in a gene database based on the degree of similarity between a key sequence and a target sequence. U.S. Pat. No. 5,538,897 discloses a method using mass spectroscopy fragmentation patterns of peptides to identify amino acid sequences in computer databases by comparison of predicted mass spectra with experimentally-derived mass spectra using a closeness-of-fit measure. U.S. Pat. No. 5,926,818 discloses a multi-dimensional database comprising a functionality for multi-dimensional data analysis described as on-line analytical processing (OLAP), which entails the consolidation of projected and actual data according to more than one consolidation path or dimension. U.S. Pat. No. 5,295,261 reports a hybrid database structure in which the fields of each database record are divided into two classes, navigational and informational data, with navigational fields stored in a hierarchical topological map which are viewed as a tree structure or as the merger of two or more such tree structures.


See also Mount et al., Bioinformatics (2001); Biological Sequence Analysis: Probabilistic Models of Proteins and Nucleic Acids (Durbin et al., eds., 1999); Bioiraformatics: A Practical Guide to the Analysis of Genes and Proteins (Baxevanis & Oeullette eds., 1998)); Rashidi & Buehler, Bioinformatics: Basic Applications in Biological Science and Medicine (1999); Introduction to Computational Molecular Biology (Setubal et al., eds 1997); Bioinformatics: Methods, and Protocols (Misener & Krawetz, eds, 2000); Bioinformatics: Sequence, Structure, and Databanks: A Practical Approach (Higgins & Taylor, eds., 2000); Brown, Bioinfor7natics: A Biologist's Guide to Biocomputing and the Internet (2001); Han & Kamber, Data Mining: Concepts and Techniques (2000); and Waterman, Introduction to Computational Biology: Maps, Sequences, and Genomes (1995).


The present invention provides a computer database comprising a computer and software for storing in computer-retrievable form assay data records cross-tabulated, e.g., with data specifying the source of the target-containing sample from which each sequence specificity record was obtained.


In an exemplary embodiment, at least one of the sources of target-containing sample is from a control tissue sample known to be free of pathological disorders. In a variation, at least one of the sources is a known pathological tissue specimen, e.g., a neoplastic lesion or another tissue specimen to be analyzed for prostate cancer. In another variation, the assay records cross-tabulate one or more of the following parameters for each target species in a sample: (1) a unique identification code, which can include, e.g., a target molecular structure and/or characteristic separation coordinate (e.g., electrophoretic coordinates); (2) sample source; and (3) absolute and/or relative quantity of the target species present in the sample.


The invention also provides for the storage and retrieval of a collection of target data in a computer data storage apparatus, which can include magnetic disks, optical disks, magneto-optical disks, DRAM, SRAM, SGRAM, SDRAM, RDRAM, DDR RAM, magnetic bubble memory devices, and other data storage devices, including CPU registers and on-CPU data storage arrays. Typically, the target data records are stored as a bit pattern in an array of magnetic domains on a magnetizable medium or as an array of charge states or transistor gate states, such as an array of cells in a DRAM device (e.g., each cell comprised of a transistor and a charge storage area, which are on the transistor). In one embodiment, the invention provides such storage devices, and computer systems built therewith, comprising a bit pattern encoding a protein expression fingerprint record comprising unique identifiers for at least 10 target data records cross-tabulated with target source.


When the target is a peptide or nucleic acid, the invention preferably provides a method for identifying related peptide or nucleic acid sequences, comprising performing a computerised comparison between a peptide or nucleic acid sequence assay record stored in or retrieved from a computer storage device or database and at least one other sequence. The comparison can include a sequence analysis or comparison algorithm or computer program embodiment thereof (e.g., BLAST, FASTA, TFASTA, GAP, BESTFIT see above) and/or the comparison are of the relative amount of a peptide or nucleic acid sequence in a pool of sequences determined from a polypeptide or nucleic acid sample of a specimen.


The invention also preferably provides a magnetic disk, such as an IBM-compatible (DOS, Windows, Windows95/98/2000, Windows NT, OS/2) or other format (e.g., Linux, SunOS, Solaris, AIX, SCO Unix, VMS, MV, Macintosh, etc.) floppy diskette or hard (fixed, Winchester) disk drive, comprising a bit pattern encoding data from an assay of the invention in a file format suitable for retrieval and processing in a computerized sequence analysis, comparison, or relative quantitation method.


The invention also provides a network, comprising a plurality of computing devices linked via a data link, such as an Ethernet cable (coax or 10BaseT), telephone line, ISDN line, wireless network, optical fiber, or other suitable signal transmission medium, whereby at least one network device (e.g., computer, disk array, etc.) comprises a pattern of magnetic domains (e.g., magnetic disk) and/or charge domains (e.g., an array of DRAM cells) composing a bit pattern encoding data acquired from an assay of the invention.


The invention also provides a method for transmitting assay data that includes generating an electronic signal on an electronic communications device, such as a modem, ISDN terminal adapter, DSL, cable modem, ATM switch, or the like, wherein the signal includes (in native or encrypted format) a bit pattern encoding data from an assay or a database comprising a plurality of assay results obtained by the method of the invention.


In a preferred embodiment, the invention provides a computer system for comparing a query target to a database containing an array of data structures, such as an assay result obtained by the method of the invention, and ranking database targets based on the degree of identity and gap weight to the target data. A central processor is preferably initialized to load and execute the computer program for alignment and/or comparison of the assay results. Data for a query target is entered into the central processor via an I/O device. Execution of the computer program results in the central processor retrieving the assay data from the data file, which comprises a binary description of an assay result.


The target data or record and the computer program are transferred to secondary memory, which is typically random access memory (e.g., DRAM, SRAM, SGRAM, or SDRAM). Targets are ranked according to the degree of correspondence between a selected assay characteristic (e.g., binding to a selected affinity moiety) and the same characteristic of the query target and results are output via an I/O device. For example, a central processor are a conventional computer (e.g., Intel Pentium, PowerPC, Alpha, PA-8000, SPARC, MIPS 4400, MIPS 10000, VAX, etc.); a program are a commercial or public domain molecular biology software package (e.g., UWGCG Sequence Analysis Software, Darwin); a data file are an optical or magnetic disk, a data server, a memory device (e.g., DRAM, SRAM, SGRAM, SDRAM, EPROM, bubble memory, flash memory, etc.); an I/O device are a terminal comprising a video display and a keyboard, a modem, an ISDN terminal adapter, an Ethernet port, a punched card reader, a magnetic strip reader, or other suitable I/O device.


The invention also preferably provides the use of a computer system, such as that described above, which comprises: (1) a computer; (2) a stored bit pattern encoding a collection of peptide sequence specificity records obtained by the methods of the invention, which are stored in the computer; (3) a comparison target, such as' a query target; and (4) a program for alignment and comparison, typically with rank-ordering of comparison results on the basis of computed similarity values.


Transgenic Animals Expressing Ovarian Cancer-Associated Proteins and “Knock-Out” Animals


The present invention also contemplates transgenic animals which are transgenic by virtue of comprising a polynucleotide of the invention, i.e. animals transformed with a cancer-associated gene of the invention. Suitable animals are generally from the phylum chordata. Chordates includes vertebrate groups such as mammals, birds, reptiles and amphibians. Particular examples of mammals include non-human primates, cats, dogs, ungulates such as cows, goats, pigs, sheep and horses and rodents such as mice, rats, gerbils and hamsters. Transgenic animals within the meaning of the present invention are non-human animals and the production of transgenic humans is specifically excluded.


Techniques for producing transgenic animals are well known in the art. A useful general textbook on this subject is Houdebine, Transgenic animals—Generation and Use (Harwood Academic, 1997)—an extensive review of the techniques used to generate transgenic animals from fish to mice and cows.


Advances in technologies for embryo micromanipulation now permit introduction of heterologous DNA into, for example, fertilized mammalian ova. For instance, totipotent or pluripotent stem cells are transformed by microinjection, calcium phosphate mediated precipitation, liposome fusion, retroviral infection or other means, the transformed cells are then introduced into the embryo, and the embryo then develops into a transgenic animal. In a highly preferred method, developing embryos are infected with a retrovirus containing the desired DNA, and transgenic animals produced from the infected embryo. In a most preferred method, however, the appropriate DNAs are coinjected into the pronucleus or cytoplasm of embryos, preferably at the single cell stage, and the embryos allowed to develop into mature transgenic animals. Those techniques as well known. See reviews of standard laboratory procedures for microinjection of heterologous DNAs into mammalian fertilized ova, including Hogan et al., Manipulating the Mouse Embryo, (Cold Spring Harbor Press 1986); Krimpenfort et al., Bio/Technology 9:844 (1991); Palmiter et al., Cell, 41: 343 (1985); Kraemer et al., Genetic manipulation of the Mammalian Embryo, (Cold Spring Harbor Laboratory Press 1985); Hammer et al., Nature, 315: 680 (1985); Wagner et al., U.S. Pat. No. 5,175,385; Krimpenfort et al., U.S. Pat. No. 5,175,384, the respective contents of which are incorporated herein by reference


Another method used to produce a transgenic animal involves microinjecting a nucleic acid into pro-nuclear stage eggs by standard methods. Injected eggs are then cultured before transfer into the oviducts of pseudopregnant recipients.


Transgenic animals may also be produced by nuclear transfer technology as described in Schnieke, A. E. et al., 1997, Science, 278: 2130 and Cibelli, J. B. et al., 1998, Science, 280: 1256. Using this method, fibroblasts from donor animals are stably transfected with a plasmid incorporating the coding sequences for a binding domain or binding partner of interest under the control of regulatory. Stable transfectants are then fused to enucleated oocytes, cultured and transferred into female recipients.


Analysis of animals which may contain transgenic sequences would typically be performed by either PCR or Southern blot analysis following standard methods.


By way of a specific example for the construction of transgenic mammals, such as cows, nucleotide constructs comprising a sequence encoding a binding domain fused to GFP are microinjected using, for example, the technique described in U.S. Pat. No. 4,873,191, into oocytes which are obtained from ovaries freshly removed from the mammal. The oocytes are aspirated from the follicles and allowed to settle before fertilization with thawed frozen sperm capacitated with heparin and prefractionated by Percoll gradient to isolate the motile fraction.


The fertilized oocytes are centrifuged, for example, for eight minutes at 15,000 g to visualize the pronuclei for injection and then cultured from the zygote to morula or blastocyst stage in oviduct tissue-conditioned medium. This medium is prepared by using luminal tissues scraped from oviducts and diluted In culture medium. The zygotes must be placed in the culture medium Within two hours following microinjection.


Oestrous is then synchronized in the intended recipient mammals, such as cattle, by administering coprostanol. Oestrous is produced within two days and the embryos are transferred to the recipients 5-7 days after estrous. Successful transfer are evaluated in the offspring by Southern blot.


Alternatively, the desired constructs are introduced into embryonic stem cells (ES cells) and the cells cultured to ensure modification by the transgene. The modified cells are then injected into the blastula embryonic stage and the blastulas replaced into pseudopregnant hosts. The resulting offspring are chimeric with respect to the ES and host cells, and nonchimeric strains which exclusively comprise the ES progeny are obtained using conventional cross-breeding. This technique is described, for example, in WO91/10741.


In another embodiment, transgenic animals of the present invention are transgenic “knock-out” animals where a specific gene corresponding to a polynucleotide referred to in Tables 1-3 has been rendered non-functional by homologous recombination. The generation of “knock-out” animals is similar to the production of other transgenic animals except that the polynucleotide constructs are designed to integrate into the endogenous genes and disrupt the function of the endogenous sequences. The generation of “knock-out” animals is known in the art, including the design of suitable constructs that will recombine at the appropriate site in the genome.


In one embodiment, the heterologous sequence which it is desired to recombine into the genome of a target animal comprises a functional sequence but under the control of an inducible promoter so that expression of the gene are regulated by administration of an endogenous molecule. This are advantageous where disruption of the gene is embryonic-lethal.


“Knock-out” animals are used as animal models for the study of gene function.


Therapeutic Peptides


In accordance with this embodiment, ovarian cancer-associated proteins of the present invention are administered therapeutically to patients for a time and under conditions sufficient to ameliorate the growth of a tumor in the subject or to prevent tumor recurrence.


It is preferred to use peptides that do not consisting solely of naturally-occurring amino acids but which have been modified, for example to reduce immunogenicity, to increase circulatory half-life in the body of the patient, to enhance bioavailability and/or to enhance efficacy and/or specificity.


A number of approaches have been used to modify peptides for therapeutic application. One approach is to link the peptides or proteins to a variety of polymers, such as polyethylene glycol (PEG) and polypropylene glycol (PPG)—see for example U.S. Pat. Nos. 5,091,176, 5,214,131 and U.S. Pat. No. 5,264,209.


Replacement of naturally-occurring amino acids with a variety of uncoded or modified amino acids such as D-amino acids and N-methyl amino acids may also be used to modify peptides


Another approach is to use bifunctional crosslinkers, such as N-succinimidyl 3-(2 pyridyidithio)propionate, succinimidyl 6-[3-(2 pyridyidithio)propionamido]hexanoate, and sulfosuccinimidyl 6-[3-(2 pyridyldithio)propionamido]hexanoate (see U.S. Pat. No. 5,580,853).


It are desirable to use derivatives of the ovarian cancer-associated proteins of the invention which are conformationally constrained. Conformational constraint refers to the stability and preferred conformation of the three-dimensional shape assumed by a peptide. Conformational constraints include local constraints, involving restricting the conformational mobility of a single residue in a peptide; regional constraints, involving restricting the conformational mobility of a group of residues, which residues may form some secondary structural unit; and global constraints, involving the entire peptide structure.


The active conformation of the peptide are stabilized by a covalent modification, such as cyclization or by incorporation of gamma-lactam or other types of bridges. For example, side chains are cyclized to the backbone so as create a L-gamma-lactam moiety on each side of the interaction site. See, generally, Hruby et al., “Applications of Synthetic Peptides,” in Synthetic Peptides: A User's Guide: 259-345 (W. H. Freeman & Co. 1992). Cyclization also are achieved, for example, by formation of cystine bridges, coupling of amino and carboxy terminal groups of respective terminal amino acids, or coupling of the amino group of a Lys residue or a related homolog with a carboxy group of Asp, Glu or a related homolog. Coupling of the .alpha-amino group of a polypeptide with the epsilon-amino group of a lysine residue, using iodoacetic anhydride, are also undertaken. See Wood and Wetzel, 1992, Int'l J. Peptide Protein Res. 39: 533-39.


Another approach described in U.S. Pat. No. 5,891,418 is to include a metal-ion complexing backbone in the peptide structure. Typically, the preferred metal-peptide backbone is based on the requisite number of particular coordinating groups required by the coordination sphere of a given complexing metal ion. In general, most of the metal ions that may prove useful have a coordination number of four to six. The nature of the coordinating groups in the peptide chain includes nitrogen atoms with amine, amide, imidazole, or guanidino functionalities; sulfur atoms of thiols or disulfides; and oxygen atoms of hydroxy, phenolic, carbonyl, or carboxyl functionalities. In addition, the peptide chain or individual amino acids are chemically altered to include a coordinating group, such as for example oxime, hydrazino, sulfhydryl, phosphate, cyano, pyridino, piperidino, or morpholino. The peptide construct are either linear or cyclic, however a linear construct is typically preferred. One example of a small linear peptide is Gly-Gly-Gly-Gly which has four nitrogens (an N4 complexation system) in the back bone that can complex to a metal ion with a coordination number of four.


A further technique for improving the properties of therapeutic peptides is to use non-peptide peptidomimetics. A wide variety of useful techniques are used to elucidating the precise structure of a peptide. These techniques include amino acid sequencing, x-ray crystallography, mass spectroscopy, nuclear magnetic resonance spectroscopy, computer-assisted molecular modeling, peptide mapping, and combinations thereof. Structural analysis of a peptide generally provides a large body of data which comprise the amino acid sequence of the peptide as well as the three-dimensional positioning of its atomic components. From this information, non-peptide peptidomimetics are designed that have the required chemical functionalities for therapeutic activity but are more stable, for example less susceptible to biological degradation. An example of this approach is provided in U.S. Pat. No. 5,811,512.


Techniques for chemically synthesising therapeutic peptides of the invention are described in the above references and also reviewed by Borgia and Fields, 2000, TibTech 18: 243-251 and described in detail in the references contained therein.


Assays for Therapeutic Compounds


The ovarian cancer proteins, nucleic acids, and antibodies as described herein are used in drug screening assays to identify candidate compounds for use in treating ovarian cancer. The ovarian cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing ovarian cancer sequences are used in drug screening assays or by evaluating the effect of drug candidates on a “gene expression profile” or expression profile of polypeptides. In a preferred embodiment, the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes after treatment with a candidate agent (e.g., Zlokarnik, et al., 1998, Science 279: 84-88); Heid, 1996, Genome Res 6: 986-94).


In a preferred embodiment, the ovarian cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing the native or modified ovarian cancer-associated proteins are used in screening assays. That is, the present invention provides methods for screening for compounds/agents which modulate the ovarian cancer phenotype or an identified physiological function of a ovarian cancer-associated protein. As above, this are done on an individual gene level or by evaluating the effect of drug candidates on a “gene expression profile”. In a preferred embodiment, the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes after treatment with a candidate agent, see Zlokarnik, supra.


Having identified the differentially expressed genes herein, a variety of assays are executed. In a preferred embodiment, assays are run on an individual gene or protein level. That is, having identified a particular gene as up regulated in ovarian cancer, test compounds are screened for the ability to modulate gene expression or for binding to the ovarian cancer-associated protein. “Modulation” thus includes both an increase and a decrease in gene expression. The preferred amount of modulation will depend on the original change of the gene expression in normal versus tissue undergoing ovarian cancer, with changes of at least 10%, preferably 50%, more preferably 100-300%, and in some embodiments 300-1000% or greater. Thus, if a gene exhibits a 4-fold increase in ovarian cancer tissue compared to normal tissue, a decrease of about four-fold is often desired; similarly, a 10-fold decrease in ovarian cancer tissue compared to normal tissue often provides a target value of a 10-fold increase in expression to be induced by the test compound.


The amount of gene expression are monitored using nucleic acid probes and the quantification of gene expression levels, or, alternatively, the gene product itself are monitored, e.g., through the use of antibodies to the ovarian cancer-associated protein and standard immunoassays. Proteomics and separation techniques may also allow quantification of expression.


In a preferred embodiment, gene expression or protein monitoring of a number of entities, i.e., an expression profile, is monitored simultaneously. Such profiles will typically involve a plurality of those entities described herein.


In this embodiment, the ovarian cancer nucleic acid probes are attached to biochips as outlined herein for the detection and quantification of ovarian cancer sequences in a particular cell. Alternatively, PCR are used. Thus, a series are used with dispensed primers in desired wells. A PCR reaction can then be performed and analyzed for each well.


Expression monitoring are performed to identify compounds that modify the expression of one or more ovarian cancer-associated sequences, e.g., a polynucleotide sequence set out in Tables 1-3. In a preferred embodiment, a test modulator is added to the cells prior to analysis. Moreover, screens are also provided to identify agents that modulate ovarian cancer, modulate ovarian cancer-associated proteins, bind to a ovarian cancer-associated protein, or interfere with the binding of a ovarian cancer-associated protein and an antibody or other binding partner.


The term “test compound” or “drug candidate” or “modulator” or grammatical equivalents as used herein describes any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for the capacity to directly or indirectly alter the ovarian cancer phenotype or the expression of a ovarian cancer sequence, e.g., a nucleic acid or protein sequence. In preferred embodiments, modulators alter expression profiles, or expression profile nucleic acids or proteins provided herein. In one embodiment, the modulator suppresses a ovarian cancer phenotype, e.g. to a normal tissue fingerprint. In another embodiment, a modulator induced a ovarian cancer phenotype. Generally, a plurality of assay mixtures are run in parallel with different agent concentrations to obtain a differential response to the various concentrations. Typically, one of these concentrations serves as a negative control, i.e., at zero concentration or below the level of detection.


Drug candidates encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 100 and less than about 2,500 daltons. Preferred small molecules are less than 2000, or less than 1500 or less than 1000 or less than 500 Daltons. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Particularly preferred are peptides.


In one aspect, a modulator will neutralize the effect of a ovarian cancer-associated protein. By “neutralize” is meant that activity of a protein is inhibited or blocked and the consequent effect on the cell.


In certain embodiments, combinatorial libraries of potential modulators will be screened for an ability to bind to a ovarian cancer polypeptide or to modulate activity. Conventionally, new chemical entities with useful properties are generated by identifying a chemical compound (called a “lead compound”) with some desirable property or activity, e.g., inhibiting activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds. Often, high throughput screening (HTS) methods are employed for such an analysis.


In one preferred embodiment, high throughput screening methods involve providing a library containing a large number of potential therapeutic compounds (candidate compounds). Such “combinatorial chemical libraries” are then screened in one or more assays to Identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional “lead compounds” or can themselves be used as potential or actual therapeutics.


A combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical “building blocks” such as reagents. For example, a linear combinatorial chemical library, such as a polypeptide (e.g., mutein) library, is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds are synthesized through such combinatorial mixing of chemical building blocks (Gallop et al., 1994, J. Med. Chem. 37(9):1233-1251).


Preparation and screening of combinatorial chemical libraries is well known to those of skill in the art. Such combinatorial chemical libraries include, but are not limited to, peptide libraries, peptoids, encoded peptides, random bio-oligomers, nonpeptidal peptidomimetics, analogous organic syntheses of small compound libraries, nucleic acid libraries, peptide nucleic acid libraries, antibody libraries, carbohydrate libraries and small organic molecule libraries.


The assays to identify modulators are amenable to high throughput screening. Preferred assays thus detect enhancement or inhibition of ovarian cancer gene transcription, inhibition or enhancement of polypeptide expression, and inhibition or enhancement of polypeptide activity.


High throughput assays for the presence, absence, quantification, or other properties of particular nucleic acids or protein products are well known to those of skill in the art. Similarly, binding assays and reporter gene assays are similarly well known. Thus, e.g., U.S. Pat. No. 5,559,410 discloses high throughput screening methods for proteins, U.S. Pat. No. 5,585,639 discloses high throughput screening methods for nucleic acid binding (i.e., in arrays), while U.S. Pat. Nos. 5,576,220 and 5,541,061 disclose high throughput methods of screening for ligand/antibody binding.


In addition, high throughput screening systems are commercially available (see, e.g., Zymark Corp., Hopkinton, Mass.; Air Technical Industries, Mentor, Ohio; Beckman Instruments, Inc. Fullerton, Calif.; Precision Systems, Inc., Natick, Mass., etc.). These systems typically automate entire procedures, including all samisle and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detectors) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols for various high throughput systems. Thus, e.g., Zymark Corp. provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.


In one embodiment, modulators are proteins, often naturally occurring proteins or fragments of naturally occurring proteins. Thus, e.g., cellular extracts containing proteins, or random or directed digests of proteinaceous cellular extracts, are used. In this way libraries of proteins are made for screening in the methods of the invention. Particularly preferred in this embodiment are libraries of bacterial, fungal, viral, and mammalian proteins, with the latter being preferred, and human proteins being especially preferred. Particularly useful test compound will be directed to the class of proteins to which the target belongs, e.g., substrates for enzymes or ligands and receptors.


In a preferred embodiment, modulators are peptides of from about 5 to about 30 amino acids, with from about 5 to about 20 amino acids being preferred, and from about 7 to about 15 being particularly preferred. The peptides are digests of naturally occurring proteins as is outlined above, random peptides, or “biased” random peptides. By “randomized” or grammatical equivalents herein is meant that each nucleic acid and peptide consists of essentially random nucleotides and amino acids, respectively. Since generally these random peptides (or nucleic acids, discussed below) are chemically synthesized, they may incorporate any nucleotide or amino acid at any position. The synthetic process are designed to generate randomized proteins or nucleic acids, to allow the formation of all or most of the possible combinations over the length of the sequence, thus forming a library of randomized candidate bioactive proteinaceous agents.


In one embodiment, the library is fully randomized, with no sequence preferences or constants at any position. In a preferred embodiment, the library is biased. That is, some positions within the sequence are either held constant, or are selected from a limited number of possibilities. For example, in a preferred embodiment, the nucleotides or amino acid residues are randomized within a defined class, e.g., of hydrophobic amino acids, hydrophilic residues, sterically biased (either small or large) residues, towards the creation of nucleic acid binding domains, the creation of cysteines, for cross-linking, prolines for SH-3 domains, serines, threonines, tyrosines or histidines for phosphorylation sites, etc., or to purines, etc.


Modulators of ovarian cancer can also be nucleic acids, as defined below. As described above generally for proteins, nucleic acid modulating agents are naturally occurring nucleic acids, random nucleic acids, or “biased” random nucleic acids. For example, digests of procaryotic or eucaryotic genomes are used as is outlined above for proteins.


In certain embodiments, the activity of a ovarian cancer-associated protein is down-regulated, or entirely inhibited, by the use of antisense polynucleotide, i.e., a nucleic acid complementary to, and which can preferably hybridize specifically to, a coding mRNA nucleic acid sequence, e.g., a ovarian cancer-associated protein mRNA, or a subsequence thereof. Binding of the antisense polynucleotide to the mRNA reduces the translation and/or stability of the mRNA.


In the context of this invention, antisense polynucleotides can comprise naturally-occurring nucleotides, or synthetic species formed from naturally-occurring subunits or their close homologs. Antisense polynucleotides may also have altered sugar moieties or inter-sugar linkages. Exemplary among these are the phosphorothioate and other sulfur containing species which are known for use in the art. Analogs are comprehended by this invention so long as they function effectively to hybridize with the ovarian cancer-associated protein mRNA. See, e.g., Isis Pharmaceuticals, Carlsbad, Calif.; Sequitor, Inc., Natick, Mass.


Such antisense polynucleotides can readily be synthesized using recombinant means, or are synthesized in vitro. Equipment for such synthesis is sold by several vendors, including Applied Biosystems. The preparation of other oligonucleotides such as phosphorothioates and alkylated derivatives is also well known to those of skill in the art.


Antisense molecules as used herein include antisense or sense oligonucleotides. Sense oligonucleotides can, e.g., be employed to block transcription by binding to the anti-sense strand. The antisense and sense oligonucleotide comprise a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target mRNA (sense) or DNA (antisense) sequences for ovarian cancer molecules. Antisense or sense oligonucleotides, according to the present invention, comprise a fragment generally at least about 14 nucleotides, preferably from about 14 to 30 nucleotides. The ability to derive an antisense or a sense oligonucleotide, based upon a cDNA sequence encoding a given protein is described in, e.g., Stein & Cohen (Cancer Res. 48:2659 (1988 and van der Krol et ai. (BioTechniques 6:958 (1988)).


In addition to antisense polynucleotides, ribozymes are used to target and inhibit transcription of ovarian cancer-associated nucleotide sequences. A ribozyme is an RNA molecule that catalytically cleaves other RNA molecules. Different kinds of ribozymes have been described, including group I ribozymes, hammerhead ribozymes, hairpin ribozymes, RNase P, and axhead ribozymes (see, e.g., Castanotto et al., Adv. in Pharmacology 25: 289-317 (1994) for a general review of the properties of different 5 ribozymes).


Methods of preparing ribozymes are well known to those of skill in the art (see, e.g., WO 94/26877; Ojwang et al., Proc. Natl. Acad. Sci. USA 90:6340-6344 (1993); Yamada et al., Human Gene Therapy 1:39-45 (1994); Leavitt et al., Proc. Natl. Acad. Sci. USA 92:699-703 (1995); Leavitt et al., Human Gene Therapy 5:1151-120 (1994); and Yamada et al., Virology 205: 121-126 (1994)).


Polynucleotide modulators of ovarian cancer are introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand binding molecule, as described in WO 91/04753. Suitable ligand binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell. Alternatively, a polynucleotide modulator of ovarian cancer are introduced into a cell containing the target nucleic acid sequence, e.g., by formation of an polynucleotide-lipid complex, as described in WO 90/10448. It is understood that the use of antisense molecules or knock out and knock in models may also be used in screening assays as discussed above, in addition to methods of treatment.


As noted above, gene expression monitoring is conveniently used to test candidate modulators (e.g., protein, nucleic acid or small molecule). After the candidate agent has been added and the cells allowed to incubate for some period of time, the sample containing a target sequence to be analyzed is added to the biochip. If required, the target sequence is prepared using known techniques. For example, the sample are treated to lyse the cells, using known lysis buffers, electroporation, etc., with purification and/or amplification such as PCR performed as appropriate. For example, an in vitro transcription with labels covalently attached to the nucleotides is performed. Generally, the nucleic acids are labeled with biotin-FITC or PE, or with cy3 or cyS.


In a preferred embodiment, the target sequence is labeled with, e.g., a fluorescent, a chemiluminescent, a chemical, or a radioactive signal, to provide a means of detecting the target sequence's specific binding to a probe. The label also are an enzyme, such as, alkaline phosphatase or horseradish peroxidase, which when provided with an appropriate substrate produces a product that are detected. Alternatively, the label are a labeled compound or small molecule, such as an enzyme inhibitor, that binds but is not catalyzed or altered by the enzyme. The label also are a moiety or compound, such as, an epitope tag or biotin which specifically binds to streptavidin. For the example of biotin, the streptavidin is labeled as described above, thereby, providing a detectable signal for the bound target sequence. Unbound labeled streptavidin is typically removed prior to analysis.


As will be appreciated by those in the art, these assays are direct hybridization assays or can comprise “sandwich assays”, which include the use of multiple probes, as is generally outlined in U.S. Pat. Nos. 5,681,702, 5,597,909, 5,545,730, 5,594,117, 5,591,584, 5,571,670, 5,580,731, 5,571,670, 5,591,584, 5,624,802, 5,635,352, 5,594,118, 5,359,100, 5,124,246 and 5,681,697, all of which are hereby Incorporated by reference. In this embodiment, in general, the target nucleic acid is prepared as outlined above, and then added to the biochip comprising a plurality of nucleic acid probes, under conditions that allow the formation of a hybridization complex.


A variety of hybridization conditions are used in the present invention, including high, moderate and low stringency conditions as outlined above. The assays are generally run under stringency conditions which allows formation of the label probe hybridization complex only in the presence of target. Stringency are controlled by altering a step parameter that is a thermodynamic variable, including, but not limited to, temperature, formamide concentration, salt concentration, chaotropic salt concentration pH, organic solvent concentration, etc.


These parameters may also be used to control non-specific binding, as is generally outlined in U.S. Pat. No. 5,681,697. Thus it are desirable to perform certain steps at higher stringency conditions to reduce non-specific binding.


The reactions outlined herein are accomplished in a variety of ways. Components of the reaction are added simultaneously, or sequentially, in different orders, with preferred embodiments outlined below. In addition, the reaction may include a variety of other reagents. These include salts, buffers, neutral proteins, e.g. albumin, detergents, etc. which are used to facilitate optimal hybridization and detection, and/or reduce non-specific or background interactions. Reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may also be used as appropriate, depending on the sample preparation methods and purity of the target.


The assay data are analyzed to determine the expression levels, and changes in expression levels as between states, of individual genes, forming a gene expression profile.


Screens are performed to identify modulators of the ovarian cancer phenotype. In one embodiment, screening is performed to identify modulators that can induce or suppress a particular expression profile, thus preferably generating the associated phenotype. In another embodiment, e.g., for diagnostic applications, having identified differentially expressed genes important in a particular state, screens are performed to identify modulators that alter expression of individual genes. In an another embodiment, screening is performed to identify modulators that alter a biological function of the expression product of a differentially expressed gene. Again, having identified the importance of a gene in a particular state, screens are performed to identify agents that bind and/or modulate the biological activity of the gene product.


In addition screens are done for genes that are induced in response to a candidate agent. After identifying a modulator based upon its ability to suppress a ovarian cancer expression pattern leading to a normal expression pattern, or to modulate a single ovarian cancer gene expression profile so as to mimic the expression of the gene from normal tissue, a screen as described above are performed to identify genes that are specifically modulated in response to the agent. Comparing expression profiles between normal tissue and agent treated ovarian cancer tissue reveals genes that are not expressed in normal tissue or ovarian cancer tissue, but are expressed in agent treated tissue. These agent-specific sequences are identified and used by methods described herein for ovarian cancer genes or proteins. In particular these sequences and the proteins they encode find use in marking or identifying agent treated cells. In addition, antibodies are raised against the agent induced proteins and used to target novel therapeutics to the treated ovarian cancer tissue sample.


Thus, in one embodiment, a test compound is administered to a population of ovarian cancer cells, that have an associated ovarian cancer expression profile. By “administration” or “contacting” herein is meant that the candidate agent is added to the cells in such a manner as to allow the agent to act upon the cell, whether by uptake and intracellular action, or by action at the cell surface. In some embodiments, nucleic acid encoding a proteinaceous candidate agent (i.e., a peptide) are put into a viral construct such as an adenoviral or retroviral construct, and added to the cell, such that expression of the peptide agent is accomplished. Regulatable gene administration systems can also be used.


Once the test compound has been administered to the cells, the cells are washed if desired and are allowed to incubate under preferably physiological conditions for some period of time. The cells are then harvested and a new gene expression profile is generated, as outlined herein.


Thus, e.g., ovarian cancer tissue are screened for agents that modulate, e.g., induce or suppress the ovarian cancer phenotype. A change in at least one gene, preferably many, of the expression profile indicates that the agent has an effect on ovarian cancer activity. By defining such a signature for the ovarian cancer phenotype, screens for new drugs that alter the phenotype are devised. With this approach, the drug target need not be known and need not be represented in the original expression screening platform, nor does the level of transcript for the target protein need to change.


In a preferred embodiment, as outlined above, screens are done on individual genes and gene products (proteins). That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of either the expression of the gene or the gene product itself are done. The gene products of differentially expressed genes are sometimes referred to herein as “ovarian cancer-associated proteins” or a “ovarian cancer modulatory protein”. The ovarian cancer modulatory protein are a fragment, or alternatively, be the full length protein to the fragment encoded by the nucleic acids referred to in Tables 1-3. Preferably, the ovarian cancer modulatory protein is a fragment. In a preferred embodiment, the ovarian cancer amino acid sequence which is used to determine sequence identity or similarity is encoded by a nucleic acid referred to in Tables 1-3. In another embodiment, the sequences are naturally occurring allelic variants of a protein encoded by a nucleic acid referred to in Tables 1-3. In another embodiment, the sequences are sequence variants as further described herein.


Preferably, the ovarian cancer modulatory protein is a fragment of approximately 14 to 24 amino acids long. More preferably the fragment is a soluble fragment. Preferably, the fragment includes a non-transmembrane region. In a preferred embodiment, the fragment has an N-terminal Cys to aid in solubility. In one embodiment, the C-terminus of the fragment is kept as a free acid and the N-terminus is a free amine to aid in coupling, i.e., to cysteine.


In one embodiment the ovarian cancer-associated proteins are conjugated to an immunogenic agent as discussed herein. In one embodiment the ovarian cancer-associated protein is conjugated to BSA.


Measurements of ovarian cancer polypeptide activity, or of ovarian cancer or the ovarian cancer phenotype are performed using a variety of assays. For example, the effects of the test compounds upon the function of the ovarian cancer polypeptides are measured by examining parameters described above. A suitable physiological change that affects activity are used to assess the influence of a test compound on the polypeptides of this invention. When the functional consequences are determined using intact cells or animals, one can also measure a variety of effects such as, in the case of ovarian cancer associated with tumours, tumour growth, tumour metastasis, neovascularization, hormone release, transcriptional changes to both known and uncharacterized genetic markers (e.g., northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as cGMP. In tire assays of the invention, mammalian ovarian cancer polypeptide is typically used, e.g., mouse, preferably human.


Assays to identify compounds with modulating activity are performed in vitro. For example, a ovarian cancer polypeptide is first contacted with a potential modulator and incubated for a suitable amount of time, e.g., from 0.5 to 48 hours. In one embodiment, the ovarian cancer polypeptide levels are determined in vitro by measuring the level of protein or mRNA. The level of protein is measured using immunoassays such as western blotting, ELISA and the like with an antibody that selectively binds to the ovarian cancer polypeptide or a fragment thereof. For measurement of mRNA, amplification, e.g., using PCR, LCR, or hybridization assays, e.g., northern hybridization, RNAse protection, dot blotting, are preferred. The level of protein or mRNA is detected using directly or indirectly labeled detection agents, e.g., fluorescently or radioactively labeled nucleic acids, radioactively or enzymatically labeled antibodies, and the like, as described herein.


Alternatively, a reporter gene system are devised using the ovarian cancer-associated protein promoter operably linked to a reporter gene such as luciferase, green fluorescent protein, CAT, or (beta-gal. The reporter construct is typically transfected into a cell. After treatment with a potential modulator, the amount of reporter gene transcription, translation, or activity is measured according to standard techniques known to those of skill in the art.


In a preferred embodiment, as outlined above, screens are done on individual genes and gene products (proteins). That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of the expression of the gene or the gene product itself are done. The gene products of differentially expressed genes are sometimes referred to herein as “ovarian cancer-associated proteins.” The ovarian cancer-associated protein are a fragment, or alternatively, be the full length protein to a fragment shown herein.


In one embodiment, screening for modulators of expression of specific genes is performed. Typically, the expression of only one or a few genes are evaluated. In another embodiment, screens are designed to first find compounds that bind to differentially expressed proteins. These compounds are then evaluated for the ability to modulate differentially expressed activity. Moreover, once initial candidate compounds are identified, variants are further-screened to better evaluate structure activity relationships.


In a preferred embodiment, binding assays are done. In general, purified or isolated gene product is used; that is, the gene products of one or more differentially expressed nucleic acids are made. For example, antibodies are generated to the protein gene products, and standard immunoassays are run to determine the amount of protein present. Alternatively, cells comprising the ovarian cancer-associated proteins are used in the assays.


Thus, in a preferred embodiment, the methods comprise combining a ovarian cancer-associated protein and a candidate compound, and determining the binding of the compound to the ovarian cancer-associated protein. Preferred embodiments utilize the human ovarian cancer-associated protein, although other mammalian proteins may also be used, e.g. for the development of animal models of human disease. In some embodiments, as outlined herein, variant or derivative ovarian cancer-associated proteins are used.


Generally, in a preferred embodiment of the methods herein, the ovarian cancer-associated protein or the candidate agent is non-diffusably bound to an insoluble support having isolated sample receiving areas (e.g. a microtiter plate, an array, etc.). The insoluble supports are made of any composition to which the compositions are bound, is readily separated from soluble material, and is otherwise compatible with the overall method of screening. The surface of such supports are solid or porous and of any convenient shape. Examples of suitable insoluble supports include microtiter plates, arrays, membranes and beads. These are typically made of glass, plastic (e.g., polystyrene), polysaccharides, nylon or nitrocellulose, teflon™, etc. microtitre plates and arrays are especially convenient because a large number of assays are carried out simultaneously, using small amounts of reagents and samples. The particular manner of binding of the composition is not crucial so long as it is compatible with the reagents and overall methods of the invention, maintains the activity of the composition and is nondiffusable. Preferred methods of binding include the use of antibodies (which do not sterically block either the ligand binding site or activation sequence when the protein is bound to the support), direct binding to “sticky” or ionic supports, chemical crosslinking, the synthesis of the protein or agent on the surface, etc. Following binding of the protein or agent, excess unbound material is removed by washing. The sample receiving areas may then be blocked through incubation with bovine serum albumin (BSA), casein or other innocuous protein or other moiety.


In a preferred embodiment, the ovarian cancer-associated protein is bound to the support, and a test compound is added to the assay. Alternatively, the candidate agent is bound to the support and the ovarian cancer-associated protein is added. Novel binding agents include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc. Of particular interest are screening assays for agents that have a low toxicity for human cells. A wide variety of assays are used for this purpose, including labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.


The determination of the binding of the test modulating compound to the ovarian cancer-associated protein are done in a number of ways. In a preferred embodiment, the compound is labeled, and binding determined directly, e.g., by attaching all or a portion of the ovarian cancer-associated protein to a solid support, adding a labeled candidate agent (e.g., a fluorescent label), washing off excess reagent, and determining whether the label is present on the solid support. Various blocking and washing steps are utilized as appropriate.


In some embodiments, only one of the components is labeled, e.g., the proteins (or proteinaceous candidate compounds) are labeled. Alternatively, more than one component are labeled with different labels, e.g., 125I for the proteins and a fluorophor for the compound. Proximity reagents, e.g., quenching or energy transfer reagents are also useful.


In one embodiment, the binding of the test compound is determined by competitive binding assay. The competitor is a binding moiety known to bind to the target molecule (i.e., a ovarian cancer-associated protein), such as an antibody, peptide, binding partner, ligand, etc. Under certain circumstances, there are competitive binding between the compound and the binding moiety, with the binding moiety displacing the compound. In one embodiment, the test compound is labeled. Either the compound, or the competitor, or both, is added first to the protein for a time sufficient to allow binding, if present. Incubations are performed at a temperature which facilitates optimal activity, typically between 4 and 40° C. Incubation periods are typically optimized, e.g., to facilitate rapid high throughput screening. Typically between 0.1 and 1 hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.


In a preferred embodiment, the competitor is added first, followed by the test compound. Displacement of the competitor is an indication that the test compound is binding to the ovarian cancer-associated protein and thus is capable of binding to, and potentially modulating, the activity of the ovarian cancer-associated protein. In this embodiment, either component are labeled. Thus, e.g., if the competitor is labeled, the presence of label in the wash solution indicates displacement by the agent. Alternatively, if the test compound is labeled, the presence of the label on the support indicates displacement.


In an alternative preferred embodiment, the test compound is added first, with incubation and washing, followed by the competitor. The absence of binding by the competitor may indicate that the test compound is bound to the ovarian cancer-associated protein with a higher affinity. Thus, if the test compound is labeled, the presence of the label on the support, coupled with a lack of competitor binding, may indicate that the test compound is capable of binding to the ovarian cancer-associated protein.


In a preferred embodiment, the methods comprise differential screening to identity agents that are capable of modulating the activity of the ovarian cancer-associated proteins. In this embodiment, the methods comprise combining a ovarian cancer-associated protein and a competitor in a first sample. A second sample comprises a test compound, a ovarian cancer-associated protein, and a competitor. The binding of the competitor is determined for both samples, and a change, or difference in binding between the two samples indicates the presence of an agent capable of binding to the ovarian cancer-associated protein and potentially modulating its activity. That is, if the binding of the competitor is different in the second sample relative to the first sample, the agent is capable of binding to the ovarian cancer-associated protein.


Alternatively, differential screening is used to identify drug candidates that bind to the native ovarian cancer-associated protein, but cannot bind to modified ovarian cancer-associated proteins. The structure of the ovarian cancer-associated protein are modeled, and used in rational drug design to synthesize agents that interact with that site. Drug candidates that affect the activity of a ovarian cancer-associated protein are also identified by screening drugs for the ability to either enhance or reduce the activity of the protein.


Positive controls and negative controls are used in the assays. Preferably control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of all samples is for a time sufficient for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radiolabel is employed, the samples are counted in a scintillation counter to determine the amount of bound compound.


A variety of other reagents are included in the screening assays. These include reagents like salts, neutral proteins, e.g. albumin, detergents, etc. which are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., are used. The mixture of components are added in an order that provides for the requisite binding.


In a preferred embodiment, the invention provides methods for screening for a compound capable of modulating the activity of a ovarian cancer-associated protein. The methods comprise adding a test compound, as defined above, to a cell comprising ovarian cancer-associated proteins. Preferred cell types include almost any cell. The cells contain a recombinant nucleic acid that encodes a ovarian cancer-associated protein. In a preferred embodiment, a library of candidate agents are tested on a plurality of cells.


In one aspect, the assays are evaluated in the presence or absence or previous or subsequent exposure of physiological signals, e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells (i.e. cell-cell contacts). In another example, the determinations are determined at different stages of the cell cycle process.


In this way, compounds that modulate ovarian cancer agents are identified. Compounds with pharmacological activity are able to enhance or interfere with the activity of the ovarian cancer-associated protein. Once identified, similar structures are evaluated to identify critical structural feature of the compound.


In one embodiment, a method of inhibiting ovarian cancer cell division is provided. The method comprises administration of a ovarian cancer inhibitor. In another embodiment, a method of inhibiting ovarian cancer is provided. The method comprises administration of a ovarian cancer inhibitor. In a further embodiment, methods of treating cells or individuals with ovarian cancer are provided. The method comprises administration of a ovarian cancer inhibitor.


In one embodiment, a ovarian cancer inhibitor is an antibody as discussed above. In another embodiment, the ovarian cancer inhibitor is an antisense molecule.


A variety of cell growth, proliferation, and metastasis assays are known to those of skill in the art, as described below.


Soft Agar Growth or Colony Formation in Suspension


Normal cells require a solid substrate to attach and grow. When the cells are transformed, they lose this phenotype and grow detached from the substrate. For example, transformed cells can grow in stirred suspension culture or suspended in semi-solid media, such as semi-solid or soft agar. The transformed cells, when transfected with tumour suppressor genes, regenerate normal phenotype and require a solid substrate to attach and grow. Soft agar growth or colony formation in suspension assays are used to identify modulators of ovarian cancer sequences, which when expressed in host cells, inhibit abnormal cellular proliferation and transformation. A therapeutic compound would reduce or eliminate the host cells' ability to grow in stirred suspension culture or suspended in semisolid media, such as semi-solid or soft.


Techniques for soft agar growth or colony formation in suspension assays are described in Freshney, Culture of Animal Cells a Manual of Basic Technique (3rd ed., 1994), herein incorporated by reference. See also, the methods section of Garkavtsev et al. (1996), supra, herein incorporated by reference.


Contact Inhibition and Density Limitation of Growth


Normal cells typically grow in a flat and organized pattern in a petri dish until they touch other cells. When the cells touch one another, they are contact inhibited and stop growing. When cells are transformed, however, the cells are not contact inhibited and continue to grow to high densities in disorganized foci. Thus, the transformed cells grow to a higher saturation density than normal cells. This are detected morphologically by the formation of a disoriented monolayer of cells or rounded cells in foci within the regular pattern of normal surrounding cells. Alternatively, labeling index with (3H)-thymidine at saturation density are used to measure density limitation of growth. See Freshney (1994), supra. The transformed cells, when transfected with tumour suppressor genes, regenerate a normal phenotype and become contact inhibited and would grow to a lower density.


In this assay, labeling index with (3H)-thymidine at saturation density is a preferred method of measuring density limitation of growth. Transformed host cells are transfected with a ovarian cancer-associated sequence and are grown for 24 hours at saturation density in non-limiting medium conditions. The percentage of cells labeling with (3H)-thymidine is determined autoradiographically. See, Freshney (1994), supra.


Growth Factor or Serum Dependence


Transformed cells have a lower serum dependence than their normal counterparts (see, e.g., Temin, J. Natl. Cancer Insti. 37:167-175 (1966); Eagle et al., J. Exp. Med. 131:836-879 (1970)); Freshney, supra. This is in part due to release of various growth factors by the transformed cells. Growth factor or serum dependence of transformed host cells are compared with that of control. Tumor specific markers levels Tumor cells release an increased amount of certain factors (hereinafter “tumour specific markers”) than their normal counterparts. For example, plasminogen activator (PA) is released from human glioma at a higher level than from normal brain cells (see, e.g., Gullino, Angiogenesis, tumour vascularization, and potential interference with tumour growth. in Biological Responses in Cancer, pp. 178-184 (Mihich (ed.) 1985)). Similarly, Tumor angiogenesis factor (TAF) is released at a higher level in tumour cells than their normal counterparts. See, e.g., Folkman, Angiogenesis and Cancer, Sem Cancer Biol. (1992)). Various techniques which measure the release of these factors are described in Freshney (1994), supra. Also, see, Unkless et al., J. Biol. Chem. 249:4295-4305 (1974); Strickland & Beers, J. Biol. Chem. 251:5694-5702 (1976); Whur et al., Br. J. Cancer 42:305 312 (1980); Gullino, Angiogenesis, tumour vascularization, and potential interference with tumour growth. in Biological Responses in Cancer, pp. 178-184 (Mihich (ed.) 1985); Freshney Anticancer Res.5:111-130 (1985).


Invasiveness into Matrigel


The degree of invasiveness into Matrigel-or some other extracellular matrix constituent are used as an assay to identify compounds that modulate ovarian cancer-associated sequences. Tumor cells exhibit a good correlation between malignancy and invasiveness of cells into Matrigel or some other extracellular matrix constituent. In this assay, tumourigenic cells are typically used as host cells. Expression of a tumour suppressor gene in these host cells would decrease invasiveness of the host cells.


Techniques described in Freshney (1994), supra, are used. Briefly, the level of invasion of host cells are measured by using filters coated with Matrigel or some other extracellular matrix constituent. Penetration into the gel, or through to the distal side of the filter, is rated as invasiveness, and rated histologically by number of cells and distance moved, or by prelabeling the cells with 125 1 and counting the radioactivity on the distal side of the filter or bottom of the dish. See, e.g., Freshney (1984), supra.


Tumor Growth In Vivo


Effects of ovarian cancer-associated sequences on cell growth are tested in transgenic or immune-suppressed mice. Knock-out transgenic mice are made, in which the ovarian cancer gene is disrupted or in which a ovarian cancer gene is inserted. Knock-out transgenic mice are made by insertion of a marker gene or other heterologous gene into the endogenous ovarian cancer gene site in the mouse genome via homologous recombination. Such mice can also be made by substituting the endogenous ovarian cancer gene with a mutated version of the ovarian cancer gene, or by mutating the endogenous ovarian cancer gene, e.g., by exposure to carcinogens.


A DNA construct is introduced Into the nuclei of embryonic stem cells. Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is re-implanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells partially derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (see, e.g., Capecchi et al., Science 244:1288 (1989)). Chimeric targeted mice are derived according to Hogan et al., Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory (1988) and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, ed., IRL Press, Washington, D.C., (1987).


Alternatively, various immune-suppressed or immune-deficient host animals are used. For example, genetically athymic “nude” mouse (see, e.g., Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)), a SCID mouse, a thymectomized mouse, or an irradiated mouse (see, e.g., Bradley et al., Br. J. Cancer 38:263 (1978); Selby et al., Br. J. Cancer 41:52 (1980)) are used as a host. Transplantable tumour cells (typically about 106 cells) injected into isogenic hosts will produce invasive tumours in a high proportions of cases, while normal cells of similar origin will not. In hosts which developed invasive tumours, cells expressing a ovarian cancer-associated sequences are injected subcutaneously. After a suitable length of time, preferably 4 to 8 weeks, tumour growth is measured (e.g. by volume or by its two largest dimensions) and compared to the control. Tumours that have a statistically significant reduction (using, e.g. Student's T test) are said to have inhibited growth.


Administration


therapeutic reagents of the invention are administered to patients, therapeutically. Typically, such proteins/polynucleotides and substances may preferably be combined with various components to produce compositions of the invention. Preferably the compositions are combined with a pharmaceutically acceptable carrier or diluent to produce a pharmaceutical composition (which are for human or animal use). Suitable carriers and diluents include isotonic saline solutions, for example phosphate-buffered saline. The composition of the invention are administered by direct injection. The composition are formulated for parenteral, intramuscular, intravenous, subcutaneous, intraocular, oral, vaginal or transdermal administration. Typically, each protein are administered at a dose of from 0.01 to 30 mg/kg body weight, preferably from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.


Polynucleotides/vectors encoding polypeptide components for use in modulating the activity of the ovarian cancer-associated proteins/polynucleotides are administered directly as a naked nucleic acid construct. When the polynucleotides/vectors are administered as a naked nucleic acid, the amount of nucleic acid administered may typically be in the range of from 1 μg to 10 mg, preferably from 100 μg to 1 mg.


Uptake of naked nucleic acid constructs by mammalian cells is enhanced by several known transfection techniques' for example those including the use of transfection agents. Example of these agents include cationic agents (for example calcium phosphate and DEAE-dextran) and lipofectants (for example lipofectam™ and transfectam™). Typically, nucleic acid constructs are mixed with the transfection agent to produce a composition.


Preferably the polynucleotide or vector of the invention is combined with a pharmaceutically acceptable carrier or diluent to produce a pharmaceutical composition. Suitable carriers and diluents include isotonic saline solutions, for example phosphate-buffered saline. The composition are formulated for parenteral, intramuscular, intravenous, subcutaneous, oral, intraocular or transdermal administration.


The pharmaceutical compositions are administered in a range of unit dosage forms depending on the method of administration. For example, unit dosage forms suitable for oral administration include, powder, tablets, pills, capsules and lozenges. Orally administered dosage forms will typically be formulated to protect the active ingredient from digestion and may therefore be complexed with appropriate carrier molecules and/or packaged in an appropriately resistant carrier. Suitable carrier molecules and packaging materials/barrier materials are known in the art.


The compositions of the invention are administered for therapeutic or prophylatic treatments. In therapeutic applications, compositions are administered to a patient suffering from a disease (e.g. ovarian cancer) in an amount sufficient to cure or at least partially ameliorate the disease and its complications. An amount adequate to accomplish this is defined as a “therapeutically effective dose”. An amount of the composition that is capable of preventing or slowing the development of cancer in a patient is referred to as a “prophylactically effective dose”.


The routes of administration and dosages described are intended only as a guide since a skilled practitioner will be able to determine readily the optimum route of administration and dosage for any particular patient and condition.


The present invention is further described with reference to the accompanying drawings and the following non-limiting examples.


EXAMPLE 1
Gene Expression Profiling to Identify Differentially-Expressed Genes in Ovarian Cancer

1. Tissue Bank and Database


Tissue was collected from patients undergoing treatment at the GCC, we have established an Ovarian Cancer Tissue Bank and Clinical Database that currently holds data on over 400 cases treated at the GCC between 1986 and 2002. Tissue (currently 149 fresh/frozen and 292 archival fixed paraffin-embedded samples) was acquired from patients undergoing cytoreductive surgery and does not interfere with the collection of tissue for the normal processing of diagnostic specimens. Patient consent, included in all our studies, was collected prior to surgery. Tissue specimens and their associated pathology reports were coded in order to maintain patient confidentiality. Uncoded data was electronically and/or physically locked with restricted access by appropriate senior investigators only. Clinical (diagnosis, treatment, residual disease) and pathological data (tumour grade, stage) were collected and updated (disease recurrence, patient survival) at regular intervals. This study has ethical approval from the South Eastern Sydney Area Health Service Research Ethics Committee, Australia. Clinical data and tissue collection are ongoing.


2. Genetic Profiling of Ovarian Cancers


In order to identify those genes differentially regulated in epithelial ovarian cancer 51 ovarian cancer tumor samples were manually dissected from biological samples derived from subjects undergoing cytoreductive surgery. These samples comprised 8 endometrioid tumors, 4 mucinous tumors and 31 serous epithelial ovarian tumors, 12 corresponding omental deposits and 8 borderline (low-malignant potential) tumors.


RNA was isolated from the tumor samples in addition to 4 normal ovary samples using Trizol reagent (Life Technologies, Rockville, Md., USA) essentially according to manufacturer's instructions. RNA was then reverse transcribed using an oligo(dT) anchored oligonucleotide that additionally comprised a T7 promoter sequence. Isolated cDNA was then transcribed in vitro using the T7 MEGAscript kit (Ambion, Austin, Tex., USA) according to manufacturers instructions. Transcription was performed with biotinylated nucleotides (Bio-11-CTP and Bio-16-UTP) to enable detection of the transcribed cRNA.


Levels of gene expression in the cancer samples was then determined by analysing the transcribed cDNA samples using customized Affymetrix GeneChip® microarrays that comprise 59,618 oligonucleotide probe sets. These probe sets facilitate analysis of 46,000 gene clusters, representing over 90% of the predicted expressed human genome.


Data were normalized, and changes in gene expression detected using a ranked penalized t-statistic with p-values adjusted for multiple testing using the Holm procedure. Analysis was performed using the LIMMA package (available from Bioconductor, Biostatistics Unit of the Dana Farber Cancer Institute at the Harvard Medical School/Harvard School of Public Health).


Gene expression in 186 samples representing 52 different tissues of the body was also determined using the previously described methods to facilitate the identification of changes in gene expression that are specific for ovarian cancer.


Using this method 284 up-regulated transcripts and 186 down-regulated transcripts were identified.


In order to determine the efficacy of such a method of analysis for determining gene expression changes associated with ovarian cancer, those genes identified were compared to results of published expression profile studies. Using this method, 71 genes were identified in the present study that had been previously identified, including, for example, genes known to be over-expressed in ovarian cancer, such as, for example MUC1 and E-cadherin.


The ovarian cancer-associated genes and proteins set forth in Table 1 include sequences that are up-regulated or down-regulated in ovarian cancer subjects, including subjects suffering specifically from serous, encodmetrioid, mucinous or clear cell ovarian cancer, or non-invasive (borderline) ovarian cancers of any phenotype, and subjects that suffered from recurrences of ovarian cancer in the medium term, or died within the medium term.


Data presented in Table 2 indicate those genes that are expressed at significantly higher levels or significantly reduced levels in patients suffering from serous cancer relative tot he level of expression of the same genes in a normal or healthy subject.


EXAMPLE 2
Validation of Gene Expression Profiling Results using Tissue Microarrays

Each of the transcripts identified as being differentially-expressed specifically in ovarian cancer was then further analysed using in situ hybridization or immunohistochemical staining of tissue microarrays constructed from a large cohort of primary ovarian tumor tissue. Such analysis confirms upregulation, down-regulation or total loss of expression of the transcripts identified in the microarray analysis of tumor samples.


Furthermore, as each of the samples in the tissue microarray have been clinicopathologically characterized (for example to identify cancer grade and/or disease stage) and the subjects from whom the tumors were isolated continuously monitored (to detect for example, death or relapse of cancer), changes with gene expression were also analysed for correlation with such parameters in order to determine predictive changes in gene expression.


The relative intensity and percentage of cells staining was determined and evaluated for associations with clinical stage and grade of disease and disease relapse using the Kaplan Meier method and log-rank test, and by univariate and bivariate analyses in a Cox proportional hazards model for gene expression and other clinical and pathologic predictors of outcome to determine the potential independent prognostic value of the markers being assessed.


Immunohistochemical analysis has been performed on several genes identified in gene profiling analysis of ovarian cancer samples. For example, SOX17, Ep-CAM and claudin 3 were shown by gene profiling analysis to be specifically up-regulated in ovarian cancer compared to normal ovaries (FIG. 1 and FIG. 2). Using immunohistochemical analysis, it was determined that SOX17, EP-CAM and claudin 3 are upregulated in serous cancer, mucinous cancer, endometroid cancer and clear cell ovarian cancer.


Furthermore, immunohistochemical analysis has been used to analyse the expression of several other genes that are specifically upregulated in mucinous ovarian cancer. In particular the expression of LI-cadherin (cadherin 17), meprin alpha and Galectin 4 as detected using immunohistochemistry is shown in FIG. 3. There was a significant increase in protein detected in the mucinous ovarian cancer samples compared to the normal ovary sample and serous ovarian cancer sample.


Immunohistochemical analysis was also performed to analyse the expression of three, genes that are known to be upregulated in ovarian cancer (CA125, MUC-1 and E-cadherin) (FIGS. 1 and 2).


EXAMPLE 3′
Identification of Prognostic Markers of Ovarian Cancer

Using a classical survival analysis to mine expression profiling data several genes that are associated with poor patient outcome (ie death or cancer relapse) have been identified (Tables 2 and 3). Such genes have clinical utility as prognostic indicators of disease.


Using detailed clinicopathological and postoperative data on all of the 51 patients included in our transcriptional profiling studies, including details of biochemical (eg. rising serum CA-125) and/or clinical recurrence of disease and overall survival, expression profiles were correlates with clinical parameters.


A preliminary survival analysis was performed on the 33 serous cancers within this cohort. The median follow-up time for these patients was 25.5 months from the date of primary laparotomy to the date of last follow-up or the date of death, and 21 of these patients (66%) were deceased from causes related to their malignancy.


Preliminary analysis of the expression profiles of these tumors identified several potential gene clusters that were associated with an increased risk of biochemical and clinical recurrence and overall survival, including the EDD gene (SEQ ID NO: 63). Exemplary prognostic markers for detecting ovarian cancer are shown in Tables 1 and 3. Preferred markers are indicated in Table 3.


Using immunohistochemical analysis two genes have been confirmed to be upregulated in serous ovarian cancer. In particular, sFRP4, a negative signalling protein of the Wnt pathway, and SOCS3, a negative signaller of IL-6 induced signalling are specifically upregulated in serous ovarian cancer when compared to normal ovarian tissue (FIG. 4A).


Furthermore, using clinical patient data and correlating this information with gene expression levels using a Cox proportional hazards model, it has been shown that high expression of sFRP4 correlates with a poor outcome in patients (n=127) with serous ovarian cancer (p=0.0056) (FIG. 4B).


EXAMPLE 4
Validation of Gene Expression Profiling Results Using Quantitative RT-PCR

Candidate diagnostic genes are screened by quantitative RT-PCR against ovarian cancer cell lines to both validate the transcript profiling data (ie check their up- or down-regulation). Candidate diagnostic genes are screened using mRNA isolated from a panel of 9 ovarian tumour cell lines, (A2780, SKOV3, OVCAR-3, IGROV-1, CAOV3, OV-90, SW626, TOV-21 G and TOV-112D), in addition to several other tumour cell lines including lines derived from breast, prostate and colorectal tumours, and immortalised (non-transformed) human ovarian surface epithelial cells and a primary normal breast epithelial cell line (184).


Total RNA is isolated from the normal and tumour cell lines, reverse transcribed into cDNA and used as template in a quantitative PCR using a LightCycler system (Roche Diagnostics). The relative amount of each gene product is determined by comparison to a standard housekeeping gene (GAPDH).


EXAMPLE 5
Identification of Novel Genes for Diagnosis of Ovarian Cancer

We identified candidate genes with diagnostic potential from our list of aberrantly regulated genes by applying the following selection procedure: genes with a good transcript profile and low p-value (ie highly significantly up- or down-regulated in ovarian cancer, as determined in Example 1); and mapping to areas of the genome that have been shown to be amplified or lost in ovarian cancer. Accordingly, it is likely that these genes are involved in the development and progression of ovarian cancer (ie putative oncogenes and tumour suppressor genes). Additional parameters for analysis included known or putative function in oncogenesis (eg signal transduction, regulation of cellular proliferation, apoptosis etc); and association with other forms of other tumours. Genes identified in this analysis are shown in Table 3.


One method for the diagnosis of cancer comprises detecting modified DNA shed by the developing tumour into the blood stream. This can include the detection of mutations in both oncogenes and tumour suppressor genes involved in the development and progression of ovarian cancer. Furthermore, it has been recently shown that aberrant methylation of tumour suppressor genes, specifically hypermethylation of their gene promoters, frequently accompanies gene silencing in cancers, and indeed in some cases appears to be the predominant mechanism of gene silencing.


Combined with the knowledge of tumour nucleic acids circulating in the blood that reflect the biological characteristics of a tumour, the detection of methylation-specific tumour suppressor gene signatures for any given tumour type has promise as a specific and sensitive molecular test for detecting and monitoring cancer. Aberrant methylation is a frequent epigenetic event in epithelial ovarian cancer and many candidate tumour suppressor genes of epithelial ovarian cancer have been shown to be hypermethylated in epithelial ovarian cancer, such as, for example BRCA1.


In particular, expression of the candidate tumor suppressor gene MCC, has been shown to be down-regulated in epithelial ovarian cancer compared to normal ovarian tissue. MCC appears to be involved in critical cell growth regulatory processes and maps to a chromosomal region hypothesised as containing a tumor suppressor gene in ovarian cancer. Furthermore, we have identified a CpG island within the predicted promoter sequence of the MCC gene, a critical feature of genes that are subject to gene silencing by hypermethylation and a known characteristic of tumor suppressor genes. Taken together these data strongly implicate MCC as a candidate tumor suppressor gene involved in epithelial ovarian cancer.

TABLE 1Genes having modified expression in subjects suffering from ovarian cancerAccession numberUniGene MappingGene symbol and titlePutative FunctionP valuea. upregulated genesNM_002354Hs.692:235Ep-CAM; TACSTD1, tumor-associated calciumLymphocyte antigen, plasma membrane, tumor antigen. Member of0signal transducer 1; epithelial gylcoproteinthe GA733 family. C arcinoma-associated antigen expressed on mostnormal epithelial cells and gastrointestinal carcinomas and functionsas a homotypic calcium-independent cell adhesion molecule. Theantigen is being used as a target for immunotherapy treatment of human carcinomas.BC006428Hs.15093:210;HSPC195, hypothetical protein HSPC195Homo sapiens cDNA FLJ10920 fis, clone OVARC1000384-resourcerer.0Hs.290304:1NM_017697Hs.24743:94FLJ20171, hypothetical protein FLJ20171contains 3 RNA recognition motifs0AW419196Hs.257924:13FLJ13782, Hypothetical protein FLJ13782weakly similar to a drosophila transcription factor0AW630088Hs.76550:164MAL2Mal2 T-cell differentiation protein; found thru interaction with TPD520which is overexpressed in breast cancer; 4 TM are involved in vesicle transportNM_004360Hs.194657:233CDH1, cadherin 1, type 1, E-cadherin (epithelial)Tumor suppressor. Ca2+-dependent glycoprotein, mediates cell—cell0interactions in epithelial cells. Mutations correlated with gastric,breast, colorectal, thyroid and ovarian cancer. Loss of functionthought to contribute to progression in cancer by increasingproliferation, invasion, and/or metastasis. The ectodomain of thisprotein mediates bacterial adhesion to mammalian cells and thecytoplasmic domain is required for internalization.NM_003761Hs.172684:89VAMP8, vesicle-associated membrane protein 8Early endosome, membrane fraction, non-selective vesicle docking,0(endobrevin)non-selective vesicle transport, protein complex assembly, synapticvesicle. Member of a family involved in docking or fusion of synapticvesicles. Associated with the perinuclear vesicular structures of theearly endocytic compartment.NM_004415Hs.349499DSP, desmoplakin (DPI, DPII)Cell shape and cell size control, cell—cell adherens junction,0epidermal differentiation, intermediate filament, structural constituentof cytoskeleton. Acts as a site of attachment for intermediatefilaments in desmosomes (intercellular junction in vertebrateepithelial cells). Compound heterozygosity for non-sense and mis-sense mutations underlies skin fragility/woolly hair syndrome.NM_013230Hs.286124:357;CD24: CD24 antigen (small cell lung carcinomaPlasma membrane, humoral defense mechanism. Cell surface0Hs.375108cluster 4 antigen)antigen; glycosyl phosphatidylinositol (GPI)-linked glycoprotein thatdifferentiates and activates granulocytes and B lymphocytes.NM_003710Hs.233950:84, Hs.182265:2,SPINT1, serine protease inhibitor, Kunitz type 1.Extracellular, membrane fraction, serine protease inhibitor. Member0Hs.7771:1Hepatocyte growth factor activator inhibitor.of the Kunitz family of serine protease inhibitors. Hepatocyte growthfactor activator inhibitor is a potent inhibitor specific for HGF activatorand is thought to be involved in regulation of proteolytic activation ofHGF in injured tissues.NM_153345Hs.17558:16FLJ90586, hypothetical proteinFunction unknown0.0001NM_015238Hs.21543:36KIAA0869, KIAA0869 protein; KIBRAFunction unknown0.0002AI282759Hs.242463:1KRT8, keratin 8Cell structure, Cytoskeletal. May form intermediate filaments; type II0.0002keratin, member of a family of structural proteins. Disruption ofmechanisms that normally regulate keratin expression in vivo couldbe related to inflammatory and neoplastic pancreatic disorders(Casanova 1999).AI393742Hs.199067:46ERBB3, v-erb-b2 erythroblastic leukemia viralTransmembrane receptor protein tyrosine kinase, epidermal growth0.0002oncogene homolog 3 (avian)factor receptor, integral plasma membrane protein, protein aminoacid phosphorylation. Member of the ERBB gene family of receptortyrosine kinases, elevated levels in certain human mammary tumorcell lines. A receptor for heregulin, capable of mediating HGL-stimulated tyrosine phosphorylation of itself. Epidermal growth factorcontains both positive and negative determinants for interaction withErbB-2/ErbB-3 heterodimers (Stortelers 2002)AW957300Hs.294142:167ESTs, Weakly similar to CYL1_HUMAN CYLICIN IFunction unknown0.0002[H. sapiens]NM_012474; W70171Hs.75939:33, Hs.170864:1UMPK, uridine monophosphate kinaseCatalyzes the phosphorylation of uridine monophosphate to uridine0.0003diphosphate. First step in production of pyrimidine nucleosidetriphosphates required for RNA and DNA synthesis. An allele of thisgene may play a role in mediating nonhumoral immunity toHemophilus influenzae type B.AA165082Hs.146388:47, Hs.113919:3MAP7, microtubule-associated protein 7Establishment and/or maintenance of cell polarity, microtubule0.0004associated protein, microtubule cytoskeleton organization andbiogenesis, structural molecule. Predominantly expressed in cells ofepithelial origin. Involved in microtubule dynamics and cellpolarization and differentiation. Stabilizes microtubules, and maymodulate microtubule functions. Studies of the related mouse proteinsuggest an essential role in microtubule function required forspermatogenesis.AA284679Hs.25640:264, Hs.5372:2CLDN3, claudin 3Integral plasma membrane protein, pathogenesis, tight junction,0.0004transmembrane receptor. Member of the claudin family of integralmembrane proteins; receptor for Clostridium perfringens enterotoxin;NM_004433Hs.166096:170ELF3, E74-like factor 3 (ets domain transcriptionEmbryogenesis and morphogenesis, transcription co-activator,0.0004factor, epithelial-specific)transcription factor, transcription from Pol II promoter. ETS domaintranscriptional activator; activates expression of epithelial cellspecific genes.AW247252Hs.75514:181NP, nucleoside phosphorylaseDNA modification, nucleobase nucleoside nucleotide and nucleic0.0004acid metabolism, purine-nucleoside phosphorylase. Enzyme purinenucleoside phosphorylase together with adenosine deaminase(ADA) serves a key role in purine catabolism, referred to as thesalvage pathway. Mutations in either enzyme result in a severecombined immunodeficiency (SCID).NM_015925Hs.361379,LISCH7, Liver-specific bHLH-Zip transcriptionLISCH protein0.0004Hs.95697:59, Hs.93649:1factorNM_022454Hs.97984:22SOX17, SRY (sex determining region Y)-box 17Likely ortholog of mouse SRY-box containing gene 17; alias SOX170.0005AI124756Hs.5337:191IDH2, Isocitrate dehydrogenase 2 (NADP+),Carbohydrate metabolism, mitochondrion0.0006mitochondrialNM_003064Hs.313:273, Hs.297895:1SPP1, secreted phosphoprotein 1 (osteopontin,Osteopontin (bone sialoprotein); bone and blood vessel extracellular0.0006bone sialoprotein I, early T-lymphocyte activationmatrix protein involved in calcification and atherosclerosis. Increased1)expression is associated with breast tumor metastasis (Urquidl2002). Role in HCC, especially in cancer-stromal interactions (Gotoh2002). Association between levels of a biomarker, osteopontin, andovarian cancer suggest its clinical usefulness (Kim 2002).BE382756Hs.169902:319, Hs.275406:1SLC2A1, Solute carrier family 2 (facilitated glucoseGlucose transporter, membrane fraction. SLC2A1/GLUT1 -0.0006transporter), member 1facilitated glucose transporter. Glucose transporter is an integralmembrane glycoprotein that is involved in transporting glucose intomost cells. 12 TMs. Role in transport of glucose across the blood-brain barrier. Consistent marker of ovarian epithelial malignancy(Kalir 2002). Marker for discriminating hepatocellular carcinoma fromother carcinomas (Zimmerman 2002).BE512730Hs.65114:718, Hs.279437:1KRT18, keratin 18Cell shape and cell size control, embryogenesis and morphogenesis,0.0006intermediate filament, structural constituent of cytoskeleton.Component of intermediate filaments; type I epidermal keratin,strongly similar to murine Endo B. Expressed in single layerepithelial tissues of the body. Mutations linked to cryptogeniccirrhosis.NM_001769Hs.1244:227, Hs.230559:1,CD9: CD9 antigen (p24)Plasma membrane, integral plasma membrane protein. Member of0.0006Hs.242020:1the transmembrane 4 superfamily (TM4SF); may mediate plateletactivation and aggregation. Cell surface glycoprotein that is known tocomplex with integrins and other transmembrane 4 superfamilyproteins.AI791905;Hs.95549:147, Hs.229556:1FLJ20273, RNA-binding proteinContains four RNA recognition motifs (RRM, RBD, or RNP)0.0007NM_019027NM_006103Hs.2719:108, Hs.54451:1WFDC2, WAP four-disulfide core domain 2Endopeptidase inhibitor, extracellular space, proteolysis and0.0009peptidolysis, spermatogenesis. Epididymis-specific secreted protein;may have a role in sperm maturation; arelong to a family ofextracellular proteinase inhibitors. Expressed in pulmonary epithelialcells, and also expressed in some ovarian cancers.U81961Hs.438580SCNN1A, sodium channel, nonvoltage-gated 1Amiloride-sensitive sodium channel, excretion, integral plasma0.0009alphamembrane protein, membrane fraction, sodium transport. Alphasubunit of the amiloride-sensitive epithelial sodium channel;functions in nonvoltage-gated channelX69699; NM_013952Hs.73149:72, Hs.213008:1PAX8, paired box gene 8Histogenesis and organogenesis, embryogenesis and0.0009morphogenesis, thyroid-stimulating hormone receptor, transcriptionfactor. Member of the paired domain family of nuclear transcriptionfactors; are involved in the ribosome assembly, required for normalthyroid development. PAX genes play critical roles during fetaldevelopment and cancer growth.AI027643Hs.120912:12ESTsFunction unknown0.001AA173992Hs.7956:28ESTsFunction unknown0.0011AB018249Hs.10458:10SCYA16, small inducible cytokine subfamily AAntimicrobial humoral response (sensu invertebrata), cell—cell0.0011(Cys—Cys), member 16.signaling, chemokine chemotaxis. Cytokine A16; lymphocyte andmonocyte chemoattractant.NM_014791Hs.184339:27MELK, likely ortholog of maternal embryonicKIAA0175 gene product; serine/threonine protein kinase domain0.0011leucine zipper kinase.NM_030674Hs.18272:81SLC38A1, solute carrier family 38, member 1amino acid transporter A1 (ATA1), likely ortholog of mouse N-system0.0012amino acid transporter protein NAT2.NM_005682Hs.6527:201GPR56, G protein-coupled receptor 56cell adhesion, cell—cell signalling, G-protein linked receptor, integral0.0012plasma membrane protein, G-protein linked receptor proteinsignalling pathway. Member of the G protein-coupled receptor family;similar to secretin and calcitonin receptors. 7 transmembranedomains, a mucin-like domain and cysteine box in the N-terminalregion. Expressed in range of tissues, highest levels in thyroid,selectively within the monolayer of cuboidal epithelial cells of thesmaller, more actively secreting follicies of human thyroid.Differentially expressed in melanoma cell lines with differentmetastatic potential (Zendman et al 1999).AI669760Hs.188881:6, Hs.199354:1ESTsdbEST Library Tissue Type restricted to prostate0.0013NM_001730Hs.84728:127KLF5, Kruppel-like factor 5 (Intestinal)RNA polymerase II transcription factor, transcription from Pol II0.0014promoter. Zinc finger transcriptional activator; localizes to thenucleus and binds the epidermal growth factor response element,binds GC boxes.AI355761Hs.242463:2KRT8, keratin 8Cell structure, Cytoskeletal. May form Intermediate filaments; type II0.0014keratin, member of a family of structural proteins. Disruption ofmechanisms that normally regulate keratin expression in vivo couldbe related to inflammatory and neoplastic pancreatic disorders(Casanova 1999).BE019020Hs.85838:171SLC16A3, solute carrier family 16 (monocarboxylicIntegral plasma membrane protein, membrane fraction,0.0015acid transporters), member 3 (MCT3)monocarboxylic acid transport, monocarboxylic acid transporter.Member of monocarboxylate transporter family; may function as atransporter (MCT3).NM_001307Hs.278562:101CLDN7, claudin 7Integral membrane protein, tight junction. Similar to murine Cldn7;0.0016NM_002266Hs.159557:394KPNA2, karyopherin alpha 2 (RAG cohort 1,DNA metabolism, G2 phase of mitotic cell cycle. NLS-bearing0.0016importin alpha 1)substrate-nucleus import, cytoplasm, importin alpha-subunit, nuclearlocalization sequence binding, nucleoplasm, regulation of DNArecombination, spindle pole body and microtubule cycle (sensuSaccharomyces). Karyopherin alpha 2 (importin alpha 1); subunit ofthe NLS (nuclear localization signal) receptor. KPNA2 proteininteracts with the NLSs of DNA heilcase Q1 and SV40 T antigen andare involved in the nuclear transport of proteins. KPNA2 also mayplay a role in V(D)J recombination.AW176120Hs.9061:77MGC2477, hypothetical protein MGC2477function unknown0.0016BE265489Hs.3123:49LLGL2, lethal giant larvae (Drosophila) homolog 2Cytoskeleton, structural molecule. May associate with nonmuscle0.0016myosin II heavy chain. cDNA source cancer cell lines. 57% ID tom. musculus 1920362A tumor suppressor gene mgl1BE279383Hs.26557:77PKP3, plakophilin 3Cell adhesion, intercellular junction. Desmosomal plaque proteins0.0016are members of the ‘armadillo-repeat’ multigene family and haveimportant functions in cytoskeleton/cell membrane interactions.J05581; NM_002456Hs.89603:128, Hs.296789:1MUC1, mucin 1, transmembraneIntegral plasma membrane protein. Cell surface mucin glycoprotein0.0016expressed by most glandular and ductal epithelial cells and somehematapoietic cell lineages. Alterations in glycosylation in epithelialcancer cells. Marker for hepatocellular carcinoma. MUC1 metaboliccomplex conserved in tumor-derived and normal epithelial cells.Expression predictor of surgical outcome in mass-formingintrahepatic cholangiocarcinoma. Tyrosine kinase c-Src constitutes abridge between cystic fibrosis transmembrane regulator channelfailure and MUC1 overexpression in cystic fibrosis.AA531276Hs.59509:9ESTs (unnamed protein product)Function unknown0.0017AW167128Hs.231934:3ESTs; weakly similar to A57717 transcription factorFunction unknown0.0018EC2AW368226Hs.67928:25, Hs.229840:1Ets-related transcription factor, ESX, epithelium-Embryogenesis and morphogenesis, transcription co-activator,0.0021restricted Ets protein ESX-not in Unigene, buttranscription factor, transcription from Pol II promoter.found using resourcerer.AK000733Hs.23900:82RACGAP1, Rac GTPase activating protein 1Strongly similar to murine Racgap1 GTPase-activating protein for0.0024rac. The plexin-B1/Rac interaction inhibits PAK activation andenhances Sema4D ligand bindingNM_014736Hs.81892:95KIAA0101 gene productfunction unknown; no signficant hits with Superfamily0.0025NM_014586Hs.109437:17HUNK, hormonally upregulated neu tumor-Developmental processes, protein serine/threonine kinase, signal0.0025associated kinasetransduction, protein kinase containing SNF1 (fam ofserine/threonine kinases) domain; progesterone and estradiolregulated. Similar to murine Hunk.AI885516Hs.95612:31, Hs.251688:1desmocollin type 2a, desmocollin 2, isoform Dsc2bCell adhesion, intercellular junction0.0027preproprotein; desmosomal glycoprotein II/III;desmocollin-3-not in Unigene, but found usingresourcerer.AW194426Hs.20726:17ESTsFunction unknown0.0027NM_001982Hs.199067:83, Hs.167386:1ERBB3, HER3 (c-erb-B3), v-erb-b2 erythroblasticEpidermal growth factor receptor, integral plasma membrane protein,0.0028leukemia viral oncogene homalog 3 (avian)protein amino acid phosphorylation. Member of the ERBB genefamily of receptor tyrosine kinases, elevated levels in certain humanmammary tumor cell lines. A receptor for heregulin, capable ofmediating HGL-stimulated tyrosine phosphorylation of itself.NM_007019Hs.93002:85UBE2C, ubiquitin carrier protein E2-CUbiquitin-dependent protein degradation, degradation of cyclin,0.0031protein modification, positive control of cell proliferation. Subunit of acomplex with ubiquitin ligase activity; complex that exhibits cyclin-selective ubiquitin ligase activity.BE184455Hs.251754:128, Hs.245742:1SLPI, secretory leukocyte protease inhibitorPlasma protein, proteinase inhibitor. Secreted inhibitor which0.0034(antileukoproteinase)protects epithelial tissues from serine proteases. Found in varioussecretions including seminal plasma, cervical mucus, and bronchialsecretions, has affinity for trypsin, leukocyte elastase, and cathepsinG. Its inhibitory effect contributes to the immune response byprotecting epithelial surfaces from attack by endogenous proteolyticenzymes; the protein is also thought to have broad-spectrum anti-biotic activity.Y00815; NM_002840Hs.75216:262, Hs.228792:1,PTPRF, protein tyrosine phosphatase, receptorCell adhesion, integral plasma membrane protein, transmembrane0.0035Hs.245063:1type, Freceptor protein, tyrosine phosphatase signaling pathway. Receptor-type protein tyrosine phosphatase F; interacts with the insulinreceptor; has Ig-like and FN-III repeats in the extracellular domainAA706017Hs.119944:14ESTsFunction unknown0.0038AA256641Hs.238894:24ESTs, Highly similar to S02392 alpha-2-Function unknown0.0041macroglobulin receptor precursorAW055308Hs.31803:15ESTs, Weakly similar to TRHY_HUMANFunction unknown0.0043TRICHOHYALI [H. sapiens]AI301558Hs.290801:35,ESTFunction unknown0.0044Hs.356228T18997Hs.180372:119;BCL2-like 1, Homo sapiens cDNA FLJ20750 fis,Function unknown0.0044Hs.394609clone HEP05174 (hypothetical proteinAI798863Hs.87191:8ESTsFunction unknown0.0049J03258Hs.2062:146VDR, vitamin D (1,25-dihydroxyvitamin D3)DNA binding, signal transduction, vitamin D3 receptor. Zinc-finger0.0049receptorDNA-binding transcription factor. Genetic polymorphism determinesbone mineral density. Stat1-vitamin D receptor interactionsantagonize 1,25-dihydroxyvitamin D transcriptional activity andenhance stat1-mediated transcription.AA151647Hs.68877:141, Hs.228686:1CYBA, cytochrome b-245, alpha polypeptideCytochrome b, membrane, mitochondrion, superoxide metabolism.0.005Alpha-subunit of cytochrome b245, primary component of themicrobicidal oxidase system of phagocytes. CYBA deficiency isassociated with chronic granulomatous disease (CGD).AI538613Hs.135657:8TMPRSS3 Transmembrane protease, serine 3Integral membrane protein, proteolysis and peptidolysis. Contains a0.0051serine protease domain, a transmembrane domain, a LDL receptor-like domain, and a scavenger receptor cysteine-rich domain. Serineproteases are known to be involved in a variety of biologicalprocesses, whose malfunction often leads to human diseases anddisorders. Expressed in fetal cochlea and many other tissues, and isthought to be involved in the development and maintenance of theinner ear or the contents of the perilymph and endolymph. Missensemutations in autosomal recessive sensorineural deafness. Identifiedas a tumor associated gene that is overexpressed in ovarian tumors.NM_018000Hs.79741:18FLJ10116, hypothetical protein FLJ10116Function unknown0.0051NM_144724Hs.124740:18hypothetical protein FLJ3053259% identity to human Zinc finger protein 910.0051AJ278016Hs.55565:35ANKRD3, ankyrin repeat domain 3ATP binding, protein amino acid phospharylation, protein binding,0.0055protein serine/threonine kinase.NM_013994Hs.75562:147DDR1, discoldin domain receptor family, member 1Cell adhesion, integral plasma membrane protein, transmembrane0.0055receptor, protein tyrosine kinase. Epithelial-specific receptor proteintyrosine kinase; are involved in cell adhesion; has putative discoldinmotifs in extracellular domain. DDR1 (CD167a) is a RTK that iswidely expressed in normal and transformed epithelial cells and isactivated by various types of collagen.T09997:NM_001312Hs.70327:196, Hs.211478:1CRIP-2, cysteine-rich protein 2Zn-finger LIM domain protein; 208-amino acid protein containing 20.0055LIM domainsBE302796Hs.105097:115TK1, thymidine kinase 1, solubleCytoplasm, thymidine kinase. Generates thymidylate for DNA0.006synthesis. TK1 gene expression together with TS, TP and DPD geneexpression may play important roles in influencing the malignantbehavior of epithelial ovarian cancer (Fujiwaki R 2002).NM_001067Hs.156346:184, Hs.270810:2TOP2A, topoisomerase (DNA) II alpha (170 kD)DNA binding, DNA topoisomerase (ATP-hydrolyzing), nucleus. DNA0.006topoisomerase II alpha; may relax DNA torsion upon replication ortranscription. Involved in processes such as chromosomecondensation, chromatid separation, and the relief of torsional stressthat occurs during DNA transcription and replication. Catalyzes thetransient breaking and rejoining of two strands of duplex DNA. Thegene encoding this enzyme functions as the target for severalanticancer agents and a variety of mutations in this gene have beenassociated with the development of drug resistance. Reducedactivity of this enzyme may also play a role in ataxia-telangiectasia.U46455Hs.252189:148, Hs.248217:1SDC4, syndecan 4 (amphiglycan, ryudocan)Integral plasma membrane. proteoglycan syndecan. Syndecans are0.0061transmembrane heparan sulfate proteoglycans that appear to act asreceptors or coreceptors involved in intracellular communication.Members of the MYC gene family and 4 members of the syndecangene family are closely situated on 4 different chromosomes.M79141Hs.13234:39ESTsFunction unknown0.0062AI955040Hs.301584:5, Hs.265398:3ESTs, Moderately similar to hypothetical proteinFunction unknown0.0065FLJ20378 [Homo sapiens] [H. sapiens]NM_005560Hs.11669:81, Hs.231010:1LAMA5, laminin, alpha 5Basement lamina, structural molecule. Widely expressed in adult0.0066tissues, with highest levels in lung, heart, and kidney. Fifth memberof the alpha subfamily of vertebrate laminin chains. Possiblebasement membrane protein; contains laminin EGF-like domain, twoextracellular laminin G domains.BE563085Hs.833:97ISG15, interferon-stimulated protein, 15 kDaCell-cell signaling, cytoplasm, extracellular space, protein binding.0.0068Protein that is induced by interferon.BE278288Hs.155048:119LU, Lutheran blood group (Auberger b antigenBlood group antigen, cell adhesion, integral plasma membrane0.0069included)protein, signal transduction, transmembrane receptor. Lutheranblood group glycoprotein; may play role in cell—cell, cell-matrixadhesion, signal transduction; member of the Ig superfamily, hasintegrin-binding motifs, SH3 domains.NM_020859Hs.278628:52ShrmL, Shroom-related protein (KIAA1481Amiloride-sensitive sodium channel (weakly similar to Mus musculus0.0074protein)PDZ domain actin binding protein)AI262789Hs.93659:52ERP70, protein disulfide isomerase related proteinEndoplasmic reticulum lumen, protein secretion. Strongly similar to0.008(calcium-binding protein, intestinal-related)rat Rn.4070 (CABP2); may bind calcium.NM_006147Hs.11801:77IRF6, interferon regulatory factor 6Member 6 of the interferon regulatory factor transcription factor0.0082family; has low similarity to IRF4, which is a lymphocytic transcriptionfactor that stimulates B cell proliferation.R61463Hs.16165:50LAK-4P, expressed in activated T/LAKexpressed in activated T/LAK lymphocytes0.0082lymphocytesAI878857;Hs.109706:285HN1, hematological and neurological expressed 1Strongly similar to murine Hn10.0087NM_016185proteinAK001783Hs.73239:37FLJ10901, hypothetical protein FLJ10901B link shows some homology to KIAA1294 but no known function0.009AC004770Hs.4756:99FEN1, flap structure-specific endonuclease 1DNA repair enzyme, DNA replication, UV protection, double-strand0.0093break repair, double-stranded DNA binding, double-stranded DNAspecific exodeoxyribonuclease, endonuclease, fatty aciddesaturation, membrane fraction. Removes 5′ overhanging flaps inDNA repair and processes the 5′ ends of Okazaki fragments inlagging strand DNA synthesis.AI567421Hs.273330:137AGRN: agrinAgrin is a neuronal aggregating factor that induces the aggregation0.0093of acetylcholine receptors and other postsynaptic proteins on musclefibers and is crucial for the formation of the neuromuscular junction.Acts at the nerve-muscle synapse in the glomerular basal membraneand on T-lymphocytes.AW161386Hs.13561:49MGC4692: hypothetical protein MGC4692Function unknown0.0103M85430Hs.155191:546VIL2, villin 2 (ezrin)Cytoskeletal anchoring, microvillus. Regulates cell adhesion and0.0106cortical morphogenesis. The cytoplasmic peripheral membraneprotein encoded by this gene functions as a protein-tyrosine kinasesubstrate in microvilli. As a member of the ERM protein family, thisprotein serves as an intermediate between the plasma membraneand the actin cytoskeleton. It plays a key role in cell surface structureadhesion, migration, and organization.AW250380Hs.109059:124, Hs.24756:11MRPL12, mitochondrial ribosomal protein L12Protein synthesis, General cellular role, Ribosomal subunit,0.0114Mitochondrial, RNA-binding protein, Ribosome-associated.AI733848;Hs.71935:13ZNF339, zinc finger protein 339Zinc finger protein0.0115NM_021220AF111856;Hs.105039:48SLC34A2, solute carrier family 34 (sodiumSLC34A2: solute carrier family 34 (sodium phosphate), member 2;0.0121NM_006424phosphate), member 2contains 8 predicted TMs and a cysteine-rich N-terminal region.Type 2 sodium-dependent phosphate transporter. member of therenal type II co-transporter family.BE386983;Hs.343214CKLFSF7; chemokine-like factor super family 7chemokine-like factor gene superfamily; transmb 4 superfamily0.0131NM_138410AA433988Hs.98502:8MUC16, mucin 16, CA125Mucin 16. Allas CA125 ovarian cancer antigen0.0137AW248314Hs.9622:83MRPS18A, mitochondrial ribosomal protein S18AMitochondrial small ribosomal subunit, protein biosynthesis,0.0149structural constituent of ribosomeribosomal mitochondrial proteinS18AAA454501Hs.43666:65PTP4A3, protein tyrosine phosphatase type IVA,Prenylated protein tyrosine phosphatase. PTPs are cell signaling0.016member 3molecules that play regulatory roles in a variety of cellular processes.Strong similarity to murine Ptp4a3 (Mm.4124). Overexpression ofthis gene in mammalian cells was reported to inhibit angiotensin-IIinduced cell calcium mobilization and promote cell growth. PRL3(PTP4A3) expressed at high levels cancer metastases (Saha et al.2001), PRL3 gene is important for colorectal cancer metastasis.U33446Hs.75799:116PRSS8, protease, serine, 8 (prostasin)Extracellular space, plasma membrane, serine type peptidase. A0.0166trypsinogen, member of the trypsin family of serine proteases. Highlyexpressed in prostate epithelia, one of several proteolytic enzymesfound in seminal fluid. Protease-mediated regulation of sodiumabsorption is a function of human airway epithelia, and prostasin is alikely candidate for this activity.X98654Hs.93837:43PITPNM, phosphatidylinositol transfer protein,Brain development, lipid metabolism, membrane fraction,0.0167membrane-associatedphosphatidylinositol transporter, phototransduction. Catalyzes thetransfer of phosphatidylinositol between membranes; similar toDrosophila rdgB.AI660149Hs.44865:39, Hs.300819:19,LEF1, Lymphoid enhancer-binding factor-1Very strongly similar to murine Lef1; may act as a transcription0.0172Hs.293904:14factor. Expressed in pre-B and T cells. Binds to T-cell receptor-alphaenhancer and confers maximal enhancer activity. A target geneectopically activated in colon cancer, from selective activation of apromoter for a full-length LEF1 isoform that binds beta-cateniri(HOVANES 2001).AF098158;Hs.9329:152C20orf1, chromosome 20 open reading frame 1ATP binding, GTP binding, cell proliferation, mitosis, nucleus spindle.0.0183NM_012112Proliferation-associated nuclear protein; associates with the spindlepole and mitotic spindle during mitosisAB014551Hs.155120:101,ARHGEF2, rho/rac guanine nucleotide exchangeCell shape and cell size control, cell surface receptor linked signal0.0206Hs.337774factor (GEF) 2transduction, guanyl-nucleotide exchange factor, microtubulecytoskeleton. Rho GTPases play a fundamental role in numerouscellular processes that are initiated by extracellular stimuli that workthrough G protein coupled receptors. The encoded protein may formcomplex with G proteins and stimulate Rho-dependent signals.Rho/Rac guanine nucleotide exchange factor (GEF) 2; associateswith microtubules, stimulates GTP binding on Rac and RhoAI278023Hs.89986:24, Hs.290780:1ESTsFunction unknown0.0208Z95152Hs.178695:25, Hs.79107:1MAPK13, mitogen-activated protein kinase 13MAP kinase, antimicrobial humoral response (sensu invertebrata),0.0217cell surface receptor, signal transduction, chemotaxis, stressresponse. MAP kinases act as an integration point for multiplebiochemical signals, and are involved in a wide variety of cellularprocesses such as proliferation, differentiation, transcriptionregulation and development. Are activated by proinflammatorycytokines and cellular stress. Transcription factor ATF2, andmicrotubule dynamics regulator stathmin are substrates of thiskinase.AW840171Hs.265398:7ESTs, Moderately similar to hypothetical proteinFunction unknown0.0222FLJ20378 [Homo sapiens] [H. sapiens]D49441Hs.155981:53MSLN, mesothelinCell adhesion, cell surface antigen, membrane. Pre-pro-0.0225megakaryocyte potentiating factor. An antibody that reacts withovarian cancers and mesotheliomas was used to isolate a cellsurface antigen named mesothelin. Although the function ofmesothelin is unknown, it may play a role in cellular adhesion and ispresent on mesothelium, mesotheliomas, and ovarian cancers.AW797437Hs.69771:262, Hs.444:1,EST, CM1-UM0039-030400-173-a09Function unknown0.0229Hs.294163:1BE396290Hs.5097:261SYNGR2, synaptogyrin 2Integral plasma membrane protein, member of a family of0.0229transmembrane synaptic vesicle proteins, specialized secretoryorganelles that store neurotransmitters in nerve terminals, andrelease them by fusing with the presynaptic plasma membraneduring exocytosis.AI656166;Hs.7331ASRGL1; asparaginase like 1glycoprotein catabolism0.02NM_025080NM_002145Hs.2733:25HOXB2, homeo box B2, Hox2H proteinCirculation, developmental processes, transcription factor. Member0.024of homeodomain family of DNA binding proteins; may regulate geneexpression, morphogenesis, and differentiation. Genes of the HOXB(or HOX2) complex are expressed specifically inerythromegakaryocytic cell lines, some are expressed only inhematopoietic progenitors.AW959311Hs.87019:8;Hypothetical protein DKFZp434J037probable serine/threonine protein kinase; KIAA05370.0251Hs.172012NM_000269Hs.118638:166, Hs.276104:1,NME1, non-metastatic cells 1, protein (NM23A)Transcription factor and nucleoside diphosphate kinase; has a role in0.0257Hs.276127:1, Hs.276246:1the transcriptional regulation of c-myc expression. Mutations inNME1 have been identified in aggressive neuroblastomas.AA379597Hs.5199:87, Hs.277192:1HSPC150, HSPC150 protein similar to ubiquitin-Similar to ubiquitin conjugating enzyme0.0259conjugating enzymeBE148235Hs.193063:100Homo sapiens cDNA FLJ14201 fis, clonehigh homology to ARP-3 actin-like protein0.0259NT2RP3002955AI683243; AI587638Hs.97258ESTsMod similarity to S29539 ribosomal protein L13a0.03AF111713Hs.286218:64JAM1, junctional adhesion moleculeCell motility, inflammatory response, intercellular junction. Role in the0.0261regulation of tight junction assembly in epithelia. Ligation of JAM isrequired for reovirus-induced activation of NF-kappa-B andapoptosis. Role in lymphocyte homing.BE391635Hs.75725:450, Hs.274751:1,TAGLN2, transgelin 2Complex assembly protein. Homolog of the protein transgelin, which0.0275Hs.277482:1, Hs.277468:1is one of the earliest markers of differentiated smooth muscle.Function not yet determined. Are an actin-binding protein.D14697Hs.77393:201, Hs.247769:1FDPS, famesyl diphosphate synthase (famesylFamesyl pyrophosphate synthetase (famesyl diphosphate0.0276pyrophosphate synthetase,synthase); part of the cholesterol synthesis pathway.dimethylallyltranstransferase,geranyltranstransferase)AW194364Hs.94814MGC2865, Hypothetical protein MGC2865Function unknown.0.0295T47364Hs.278613:145IFI27, interferon, alpha-inducible protein 27Integral membrane protein. Isolated from estradiol-treated human0.03breast carcinoma cells. Induced by interferon-alpha in human celllines of different origin, expression is independent of the presence ofestradiol receptor in the cells.U17760Hs.301103:71, Hs.75517:24,LAMB3, Laminin, beta 3 (nicein (125 kD), kalininEpidermal differentiation, laminin-5, structural molecule. Member of0.0304Hs.199068:1(140 kD), BM600 (125 kD)) (Accn NM_000228)a family of basement membrane proteins. LAMB3 serves as the betachain in laminin-5. Mutations in LAMB3 have been identified as thecause of various types of epidermolysis bullosa.AU076517Hs.184276:142SLC9A3R1, solute carrier family 9Actin cytoskeleton, protein complex assembly. Regulatory cofactor of0.0312(sodium/hydrogen exchanger), isoform 3the NHE3 (SLC9A3) sodium/hydrogen antiporter, interacts withregulatory factor 1merlin (NF2) and ERM family members; has two PDZ domains.Structural determinants in interaction of beta 2 adrenergic andplatelet-derived growth factor receptorsAW880841Hs.96908,PIG11, p53-induced proteinNegative control of cell proliferation, stress response. May generate0.0314Hs.74427:112or respond to oxidative stress, may have a role in p53-dependentapoptosis Polyak K, Xia Y, Zweier JL, Kinzier KW, Vogelstein B. Amodel for p53-induced apoptosis. Nature. 1997 Sep18; 389(6648): 300-5.H24185Hs.92918:91BM-009, hypothetical protein BM-009Function unknown0.0314BE614410Hs.23044:51MGC16386, hypothetical protein, similar to RIKENFunction unknown.0.0326cDNAH16423Hs.82685:37CD47: CD47 antigen (Rh-related antigen, integrin-Oncogenesis, plasma membrane, plasma glycoprotein, cell—cell0.0336associated signal transducer)matrix adhesion, integral plasma membrane proteoglycan, integrinreceptor signal signalling pathway. Similar to Rh-antigen; mayinteract with integrins and have a role in intracellular calciumincrease during cell adhesion.AU076611;Hs.154672:123MTHFD2, methylene tetrahydrofolateElectron transporter, methenyltetrahydrofolate cyclohydrolase,0.0342NM_006636dehydrogenase (NAD+ dependent);mitochondrion. encodes a nuclear-encoded mitochondrialmethenyltetrahydrofolate cyclohydrolasebifunctional enzyme with methylenetetrahydrofolate dehydrogenaseand methenyltetrahydrofolate cyclohydrolase activities. may provideformyltetrahydrofolate for formylmethionyl tRNA synthesis; involvedin initiation of mitochondrial protein synthesis.AI859390Hs.288940:49TMEM8, five-span transmembrane protein M83;Integral plasma membrane protein. Type I transmembrane protein;0.0345type I transmembrane proteincontains five membrane-spanning domainsAA159216Hs.55505:57FLJ20442, hypothetical protein FLJ20442Contains a dual specificity protein phosphatase catalytic domain;0.035434% similar to protein-tyrosine phosphataseAF119665;Hs.184011:156PP, pyrophosphatase (inorganic)Inorganic diphosphatase, phosphate metabolism. Catalyzes the0.0358NM_021129hydrolysis of pyrophosphate to inorganic phosphateBE513613;Hs.11538:275ARPC1B, actin related protein 2/3 complex,Cell motility, structural constituent of cytoskeleton. Arp2/3 complex,0.0387NM_005720subunit 1A (41 kD)subunit 1A; involved in assembly of the actin cytoskeleton, may havea role in protrusion of lamellipodiaNM_012153Hs.182339EHF: ets homologous factorDNA binding, tumor suppressor, cell proliferation, developmental0.0404processes, transcription activating factor. Member of the ESEsubfamily of Ets transcription factorsAW772298Hs.21103:40, Hs.266784:2,Homo sapiens mRNA; cDNA DKFZp564B076Alias coat protein gamma-cop0.0423Hs.102950:1(from clone DKFZp564B076)H16646Hs.118666:66PP591, hypothetical protein PP591Hypothetical protein PP591 (Novel Human cDNA clones with0.043function of inhibiting cancer cell growth; unpublished)AA279661Hs.83753:244, Hs.301236:3SNRPB, small nuclear ribonucleoproteinSpliceosome, mRNA splicing, small nuclear ribonucleoprotein. U10.0446polypeptides B and B1and U2 snRNP protein; component of snRNP complexes, requiredunits of the spliceosomeBE001596Hs.85266:102ITGB4, integrin, beta 4Cell adhesion receptor, integrin, invasive growth, oncogenesis. Beta0.04534 subunit of integrin; involved in cell—cell and cell-matrix interactions;member of a family of cell-surface proteins. Binding of beta 4 toplectin is essential for the proper formation and function ofhemidesmosomes.BE246444Hs.283685:148, Hs.232028:2FLJ20396, hypothetical protein FLJ20396100%/175aa unnamed protein g70204680.0453X54942Hs.83758:34CKS2, CDC28 protein kinase 2Cell proliferation, regulation of CDK activity. Similar to S. pombe0.0478p13suc1; binds and regulates CDK-cyclin complexes. expressed indifferent patterns through the cell cycle in HeLa cells, which reflectsspecialized role for the encoded protein.AA305599Hs.238205:36PRO2013, hypothetical protein PRO2013Function unknown0.0483AF019226Hs.8036:84RAB3D, member RAS oncogene familyRAB small monomeric GTPase, hemocyte development. GTP-0.0485binding protein; are involved in vesicle transport; member of the RABfamily of small GTPases. Alias GOV, that is overexpressed inglioblastoma multiforme tissue as compared to normal brain tissue.GOV is also highly expressed in recurrent glioma, colon tumormetastatic to brain, breast tumors, prostate tumors, and severaltumor cell linesNM_001949Hs.1189:65, Hs.296939:2E2F3, E2F transcription factor 3Protein binding, transcription factor, transcription initiation from Pol II0.049promoter. Involved in cell cycle regulation, binds retinoblastomaprotein (Rb). E2F family plays a crucial role in the control of cellcycle and action of tumor suppressor proteins and is also a target ofthe transforming proteins of small DNA tumor viruses.AF217513Hs.279905:73, Hs.283649:4ANKT, nucleolar protein ANKTclone HQ0310 PRO0310p1 nucleolar protein ANKT - no functional0.0504dataAW513143Hs.98367:8ESTsExpressed in uterus0.0535AJ245671Hs.12844:73EGFL6, EGF-like-domain; multiple 6Cell cycle, oncogenesis, integrin ligand, extracellular space. Member0.0568of the epidermal growth factor (EGF) repeat superfamily; contains anEGF-like-domain. Expressed early during development, and itsexpression has been detected in lung and meningioma tumors.AA084248Hs.85339:64GPR39, G protein-coupled receptor 39G-protein linked receptor, G-protein coupled receptor protein0.19signaling pathway, integral plasma membrane protein.U62801Hs.79361:65KLK6, kallikrein 6 (neurosin, zyme)Serine type peptidase, pathogenesis. Neurosin (protease M, zyme);0.0159a serine protease that cleaves amyloid precursor protein (APP).Growing evidence suggests that many kallikreins are implicated incarcinogenesis and some have potential as novel cancer and otherdisease biomarkers.D49441Hs.155981:53MSLN, mesothelinCell adhesion, cell surface antigen, membrane. Pre-pro-0.147megakaryocyte potentiating factor. An antibody that reacts withovarian cancers and mesotheliomas was used to isolate a cellsurface antigen named mesothelin. Although the function ofmesothelin is unknown, it may play a role in cellular adhesion and ispresent on mesothelium, mesotheliomas, and ovarian cancers.X51630Hs.1145:22, Hs.296851:1WT1, Wilms tumor 1Nucleus, transcription factor, transcription regulation. 4 Zn finger0.2938domains. Functions in kidney and gonad proliferation anddifferentiation. Mutations in this gene are associated with thedevelopment of Wilms tumors in the kidney or with abnormalities ofthe genitourinary tract.AB018305Hs.5378:149SPON1, spondin 1, (f-spondin) extracellular matrixExtracellular matrix protein. Very strongly similar to rat F-spondin0.3394protein(Rn.7546); may have a role in the growth and guidance of axons.AA433988Hs.98502:8MUC16, mucin 16, CA125Mucin 16. Alias CA125 ovarian cancer antigen0.6568NM_006149Hs.5302:132LGALS4, lectin, galactoside-binding, soluble, 4Lectin, cytosol, cell adhesion, plasma membrane. Binds to beta0.0001(galectin 4)galactoside, involved in cell adhesion, cell growth regulation,inflammation, immunomodulation, apoptosis and metastasis;member of a family of lectins. LGALS4 is an S-type lectin that isstrongly underexpressed in colorectal cancer.AA315933Hs.120879:17Homo sapiens, clone MGC: 32871Function unknown0.0001IMAGE: 4733535, mRNA, complete cdsU47732Hs.84072:110TM4SF3, transmembrane 4 superfamily member 3Integral plasma membrane protein, lysosome, pathogenesis, protein0.0028amino acid glycosylation, signal transducer, tumor antigen. Cellsurface glycoprotein defined by the monoclonal antibody CO-029 isa 27- to 34-kD membrane protein expressed in gastric, colon, rectal,and pancreatic carcinomas but not in most normal tissuesNM_005588Hs.179704MEP1A, meprin A alpha, PABA peptide hydrolasemetalloprotease located apically and secreted by epithelial cells in0.01normal colon; degrades broad range of ECM components in vitro;proposed role in tumour progression by facilitating migration,intravasation and metastasisAW503395Hs.5541:112ATP2A3, ATPase, Ca++ transporting, ubiquitousEndoplasmic reticulum, adenosinetriphosphatase, small molecule0.0154transport, calcium-transporting ATPase, integral plasma membraneprotein. Sarco/endoplasmic reticulum Ca2+-ATPase; pumps calcium.NM_004063Hs.89436:50CDH17, cadherin 17, LI cadherin (liver-intestine)Cell adhesion, integral plasma membrane protein, membrane0.0172fraction, small molecule transport, transporter. Member of thecadherin family of calcium-dependent glycoproteins; facilitatesuptake of peptide-based drugs, may mediate cell—cell interactions.Component of the gastrointestinal tract and pancreatic ducts,intestinal proton-dependent peptide transporter in the first step inoral absorption of many medically important peptide-based drugs.AI073913Hs.100686:20LOC155465, anterior gradient protein 3Oncogenesis0.0266AI928445Hs.92254:80SYTL2: synaptotagmin-like 2Synaptotagmin-like protein of the C2 domain-containing family of0.08proteins. Although the specific function of the synaptotagmin-likeproteins is unknown, a role in regulation of synaptic vesicletrafficking via their C2 domains has been suggested. Region of weaksimilarity to murine GphW40460Hs.144442:5PLA2G10: phospholipase A2, group XExtracellular, secreted phospholipase A2. Group X secretory0.1888phospholipase_a2; hydrolyzes the phospholipid sn-2 ester bond;member of the phospholipase familyAA132961Hs.212533:4Homo sapiens cDNA: FLJ22572 fis, cloneFunction unknown0.1965HSI02313AF111856Hs.105039:48SLC34A2, solute carrier family 34 (sodiumSLC34A2: solute carrier family 34 (sodium phosphate), member 2;0.5078phosphate), member 2contains 8 predicted TMs and a cysteine-rich N-terminal region.Type 2 sodium-dependent phosphate transporter. member of therenal type II co-transporter family.AA143654zo65a02.r1 Stratagene pancreas (#937208) HomoFunction unknown0.036sapiens cDNA clone IMAGE: 591722 5′, mRNAsequenceb. prognostic IndicatorsAA046217Hs.105370:2ESTsFunction unknown0.00NM_015902EDD: Homo sapiens progestin induced proteinSoluble fraction, cell proliferation, ubiquitin- protein ligase, ubiquitin0.00(DD5), mRNA. VERSION NM_020967.1 GIconjugating enzyme, ubiquitin-dependent protein degradation.Member of the HECT family of proteins; may function as an E3ubiquitin-protein ligase. This gene is localized to chromosome 8q22,a locus disrupted in a variety of cancers. This gene potentially has arole in regulation of cell proliferation or differentiation.T83882Hs.97927:20ESTsFunction unknown0.01#(NOCAT)NM_001615*: Homo sapiens actin, gamma 2,Structural protein of muscle. Gamma 2 actin; enteric-type, smooth0.01smooth muscle, enteric (ACTG2), mRNA. variantmuscle cell actin.1, mRNA.AB040888Homo sapiens mRNA for KIAA1455 protein, partialFunction unknown0.01cdsAA628980Hs.192371:3DSCR8Function unknown0.01down syndrome critical region protein DSCR8AI623351Hs.172148:51ESTsFunction unknown0.01AW614420Hs.204354:383ARHBRHO small monomeric GTPase, RHO protein signal transduction,0.01ras homolog gene family, member Bperipheral plasma membrane protein. Ras-related GTP bindingprotein of the rho subfamily, member B; may regulate assembly ofactin stress fibers and focal adhesions; very strongly similar tomurine Arhb.AA243499Hs.104800:23hypothetical protein FLJ10134Highly similar to murine p19.5; are a membrane protein0.01AF251237Hs.112208:16GAGED2GAGE genes are expressed in a variety of tumors and in some fetal0.01XAGE-1 proteinand reproductive tissues. This gene is strongly expressed in Ewing'ssarcoma, alveolar rhabdomyosarcoma and normal testis. The proteinencoded by this gene contains a nuclear localization signal andshares a sequence similarity with other GAGE/PAGE proteins.Because of the expression pattern and the sequence similarity, thisprotein also belongs to a family of CT (cancer-testis) antigens.AI970797Hs.64859:16ESTsFunction unknown0.01AF145713Hs.61490:51SCHIP1Cytoplasm. Associates with the neurofibromatosis type 2 protein0.01schwannomin-interacting protein 1schwannomin (NF2); contains a colled-coil domain|ProteomeX78565Hs.289114:173, Hs.74637:1TNCCell adhesion, extracellular matrix, cell adhesion receptor, ligand0.01hexabrachion (tenascin C, cytotactin)binding or carrier. Hexabrachion (tenascin c), an extracellular matrixglycoprotein; has epidermal growth factor-like repeatsT97307gb:ye53h05.s1 Soares fetal liver spleen 1NFLSFunction unknown0.01Homo sapiens cDNA clone IMAGE: 121497 3′,mRNA sequence.BE243845Hs.75511:418CTGFCell motility, plasma membrane, soluble fraction, response to0.01connective tissue growth factorwounding, extracellular matrix, extracellular space, epidermaldifferentiation, cell growth and maintenance, insulin-like growthfactor binding, insulin-like growth factor receptor binding protein.Connective tissue growth factor; binds IGF, may have a role inregulating normal and neoplastic cell growthAW068302Hs.182183:214, Hs.325474:172,CALD1Cytoskeleton, actin binding, calmodulin binding, tropomyosin0.01Hs.283080:7caldesmon 1binding. Protein of unknown function. Actomyosin regulatory protein,non-muscle formAL133561Hs.241426:5DKFZP434B061 proteinFunction unknown0.01BE313555Hs.7252:158RAI17Function unknown0.02retinoic acid induced 17X07820Hs.2258:1MMP10Zinc binding, extracellular space, extracellular matrix,0.02matrix metalloproteinase 10 (MMP10; stromelysinmetalloendopeptidase, proteolysis and peptidolysis. Stromelysin 2;2)matrix metalloprotease that degrades connective tissueAI973016Hs.15725:77IER5Function unknown. A related mouse gene may play an important role0.02immediate early response 5in mediating the cellular response to mitogenic signals.AF084545Homo sapiens versican Vint isoform, mRNA,Function unknown0.02partial cdsU41518Hs.74602:146, Hs.767:1AQP1Excretion, water transport, water transporter, Integral plasma0.02aquaporin 1 (channel-forming integral protein,membrane protein. Aquaporin 1 (channel-forming integral protein);28 kD)member of a family of water-transportersZ11894H. sapiens rearranged mRNA for immunoglobulin0.02kappa chain (VNJ)AW138190Hs.180248:8ZNF124DNA binding. C2H2 zinc-finger protein 1240.02zinc finger protein 124 (HZF-16)BE086548Hs.42346:83, Hs.6975:42MYOZ2calcineurin-binding protein calsarcin-10.02myozenin 2W47196Hs.166172:50ARNTNucleus, transcription factor, transcription co-activator, transcription,0.02aryl hydrocarbon receptor nuclear translocatorDNA-dependent, protein-nucleus import, translocation, arylhydrocarbon receptor nuclear translocator. Aryl hydrocarbonreceptor nuclear translocator; used in receptor translocation fromcytosol to nucleusAI796870Hs.54277:76DXS9928ENucleus. Has many charged residues and a possible nuclear0.02DNA segment on chromosome X (unique) 9928localization signalexpressed sequenceX02761Hs.287820:73, Hs.321592:1FN1Cell adhesion, cell motility, cell adhesion, soluble fraction, signal0.02fibronectin 1transduction, extracellular matrix, extracellular space. Fibronectin 1;member of family of proteins found in plasma and extracellularmatrixAW968613Hs.79428:166BNIP3Anti-apoptosis, apoptosis inhibitor. Bcl2-related protein 3; binds0.02BCL2/adenovirus E1B 19 kD-interacting protein 3antiapoptotic viral E1B 19 kDa protein and cellular Bcl2 proteinAW972565Hs.32399:24ESTs, Weakly similar to S51797 vasodilator-Function unknown0.02stimulated phosphoprotein [H. sapiens]AF045229Hs.82280:81RGS10Regulator of G protein signaling (RGS) family members are0.02regulator of G-protein signalling 10regulatory molecules that act as GTPase activating proteins (GAPs)for G alpha subunits of heterotrimeric G proteins. RGS proteins areable to deactivate G protein subunits of the Gi alpha, Go alpha andGq alpha subtypes. They drive G proteins into their inactive GDP-bound forms.AW953853Hs.292833:19PAEPDevelopmental processes. Placental protein 14 (Glycodelin);0.02progestagen-associated endometrial proteinmember of lipocalin superfamily, highly similar to beta-lactoglobulins(placental protein 14, pregnancy-associatedendometrial alpha-2-globulin, alpha uterineprotein)U52426Hs.74597:75, Hs.157615:3STIM1Integral plasma membrane protein, positive control of cell0.02stromal interaction molecule 1proliferation. Very strongly similar to murine Stim1; are atransmembrane stromal cell proteinF06700Hs.7879:115IFRD1Myoblast determination. Strongly similar to rat interferon-related0.02interferon-related developmental regulator 1developmental regulator 1; may play a role in muscle differentiationAI798863Hs.87191:8ESTsFunction unknown0.03NAC4001170: gi|6863176|gb|AAF30402.1|AF109924_10.03(AF109924) sulfatase 1 precursor [Helix pomaH52761Hs.141475:24Homo sapiens cDNA clone IMAGE: 178663Function unknown0.03BE546947Hs.44276:43HOXC10Embryogenesis and morphogenesis, positive control of cell0.03homeo box C10proliferation, RNA polymerase II transcription factor. Homeobox C10,member of the homeobox developmental regulator family; binds withHOXA13 and HOXC13 to the Lamin B2 origin; ortholog of DrosophilaAbdominal-BAU076643Hs.313:257, Hs.329910:1SPP1Ossification, extracellular matrix, skeletal development. Osteopontin0.03secreted phosphoprotein 1 (osteopontin, bone(bone sialoprotein); bone and blood vessel extracellular matrixsialoprotein I, early T-lymphocyte activation 1)protein involved in calcification and atherosclerosis#(NOCAT)NM_015902*: Homo sapiens progestin induced0.03protein (DD5), mRNA. VERSION NM_020967.1 GIU20536Hs.3280:20CASP6Induction of apoptosis, cysteina-type peptidase, proteolysis and0.03caspase 6, apoptosis-related cysteine proteasepeptidolysis. Caspase 6; a cysteine (thiol) protease; related to theICE-subfamily of caspasesAA581602Hs.41840:7ESTsFunction unknown0.03AJ245210gb: Homo sapiens mRNA for ImmunoglobulinFunction unknown0.03gamma heavy chain variable region, partial, clone1A-4G21.X65965H. sapiens SOD-2 gene for manganese superoxide0.03dismutaseAI806770Hs.30258:9ESTsFunction unknown0.03BE386490Hs.279663:51PIRNucleus, transcription co-factor, transcription from Pol II promoter.0.03PirinPutative cofactor of the NFI/CTF1 transcriptional activatorAW581992Hs.301434:104, Hs.329017:1KIAA1387Function unknown0.03KIAA1387 proteinU77534Human clone 1A11 immunoglobulin variableFunction unknown0.03region (VH5-D-JH4) gene, partial cdsAL034417Hs.11169:194, Hs.10958:1,Gene 33/Mig-6Function unknown0.03Hs.74137:1L10343Hs.112341:96, Hs.1968:1Homo sapiens elafin precursor, gene, completeFunction unknown0.03cdsAW518944Hs.76325:80, Hs.231299:1IGJLinker protein for immunoglobulin alpha and mu polypeptides0.03immunoglobulin J polypeptide, linker protein forimmunoglobulin alpha and mu polypeptidesW28729Hs.236510:6Human retina cDNA randomly primed sublibraryFunction unknown0.03Homo sapiens cDNA, mRNA sequenceAI640160Hs.74131:4ARSEArylsulfatase, skeletal development. Arylsulfatase E; likely involved0.03arylsulfatase E (chondrodysplasia punctata 1)in warfarin embryopathy.U11862Hs.75741:62ABP1Metabolism, peroxisome, amine oxidase, drug binding. Diamine0.03amiloride binding protein 1 (amine oxidaseoxidase (D-amino-acid oxidase histaminase, amiloride-binding(copper-containing))protein); deaminates putrescine and histamineAW295980Hs.252741:3ESTsFunction unknown0.03X59135Hs.156110:4H. sapiens mRNA for immunoglobulin 0-81VL0.03BE466173Hs.379794Homo sapiens mRNA; cDNA DKFZp686N0118Function unknown0.03(from clone DKFZp686N0118)#(NOCAT)Target Exon0.03AI354722Hs.127216:24hypothetical protein FLJ13465Function unknown0.04M90464Hs.169825:45, Hs.408:1Human collagen type IV alpha 5 chain (COL4A5)Function unknown0.04gene, 5′ endAA829286Hs.332053:48, Hs.336462:10SAA1Inflammatory response, high-density lipoprotein. Member of the0.04serum amyloid A1serum amyloid A protein family; member of high densityapolipoproteins.AI333771Hs.82204:8, Hs.228363:1ESTsFunction unknown0.04BE465867;Hs.197751:66DAAM1The protein encoded by this gene contains FH domains and belongs0.04NM_014992dishevelled associated activator of morphogenesis 1to a novel FH protein subfamily implicated in cell polarity, thought tofunction as a scaffolding protein.BE616902Hs.285313:145, Hs.4055:43COPEBA trancriptional activator, capable of activating transcription0.04core promoter element binding proteinapproximately 4-fold either on homologous or heterologouspromoters. The DNA binding and transcriptional activity of thisprotein, in conjunction with its expression pattern, suggests that thisprotein may participate in the regulation and/or maintenance of thebasal expression of pregnancy-specific glycoprotein gene andpossibly other TATA box-less genes.AA430373gb: zw20f11.s1 Soares ovary tumor NbHOT HomoFunction unknown0.04sapiens cDNA clone IMAGE: 769869 3′ similar togb: M63438 IG KAPPA CHAIN PRECURSOR V-IIIREGION (HUMAN);, mRNA sequence.R27430Hs.271565:3ESTsFunction unknown0.04BE387335Hs.283713:68CTHRC1Function unknown0.04collagen triple helix repeat containing 1AW264102Hs.39168:16ESTsFunction unknown0.04NATarget ExonFunction unknown0.04AW952323Hs.129908:39KIAA0591 proteinFunction unknown0.04AA088177Hs.172870:13ESTsFunction unknown0.04BE614567Hs.19574:123MGC5469Function unknown0.04hypothetical protein MGC5469AL079658Hs.338207:139, Hs.146559:1FRAP1DNA repair, DNA recombination, cell cycle control, 1-0.04FK506 binding protein 12-rapamycin associatedphosphatidylinositol 3-kinase, inositol/phosphatidylinositol kinase,protein 1FKBP-rapamycin associated protein; phosphatidylinositol kinase thatmay mediate rapamycin inhibition of the cell cycle progressionthrough G1NM_002776Hs.69423:46, Hs.275464:1KLK10Extracellular, serine-type peptidase. Putative serine protease0.04kallikrein 10 (KLK10) (PRSSL1) (nes1)BE261944Hs.118625:62CYB561Energy pathways, secretory vesicle, cytochrome b5 reductase,0.04cytochrome b-561secretory vesicle membrane, integral plasma membrane protein.Cytochrome b561; serves as a biological marker for adrenergicsecretory vesiclesNM_006379Hs.171921:50SEMA3CDrug resistance, immune response, cell growth and maintenance.0.04sema domain, immunoglobulin domain (Ig), shortSemaphorin E; member of a protein family involved in neuronalbasic domain, secreted, (semaphorin) 3Cgrowth cone guidanceAI002238Hs.11482:19SFRS11Nucleus, mRNA splicing, mRNA processing, pre-mRNA splicing0.04splicing factor, arginine/serine-rich 11factor, May have a role in pre-mRNA splicing; containsarginine/serine-rich domain and an RRM domain#(NOCAT)ENSP00000231844*: Ecotropic virus integration 10.04site protein.X81789Hs.77897:149SF3A3Nucleus, spliceosome, mRNA splicing, mRNA processing, pre-0.04splicing factor 3a, subunit 3, 60 kDmRNA splicing factor. Spliceosome-associated protein 3a, subunit 3;component of the essential heterotrimeric splicing factor SF3a;contains a zinc fingerNM_002122Hs.198253:21HLA-DQA1Pathogenesis, class II major histocompatibility complex antigen.0.00major histocompatibility complex, class II, DQAlpha 1 chain of HLA-DQ1 class II molecule (Ia antigen); complexalpha 1binds peptides and presents them to CD4+ T lymphocytes|ProteomeAB001914Homo sapiens PACE4 gene, exon 23-25,Function unknown0.04complete cdsAA311919Hs.69851:24NOLA1Involved in various aspects of rRNA processing and modification.0.04nucleolar protein family A, member 1 (H/ACA smallLocalize to the dense fibrillar components of nucleoli and to coilednucleolar RNPs)(Cajal) bodies in the nucleus.AI381750Hs.283437:122, Hs.10065:58HTGN29 proteinFunction unknown0.04#(NOCAT)NM_000636*: Homo sapiens superoxide dismutaseMitochondrion, oxidative stress response, manganese superoxide0.042, mitochondrial (SOD2), mRNA. expression)dismutase. Manganese superoxide dismutase; intramitochondrial(RFX2), mRNA.free radical scavenging enzyme; has strong similarity to murineSod2.AA292998Hs.163900:25ESTsFunction unknown0.04BE439580Hs.75498:40SCYA20Chemokine, chemotaxis, immune response, signal transduction,0.04small inducible cytokine subfamily A (Cys—Cys),extracellular space, cell—cell signalling, inflammatory response,member 20antimicrobial humoral response. Cytokine A20 (exodus); chemotacticfactor for lymphocytes, but not a chemotactic factor for monocytesAI677897Hs.76640:124RGC32Cytoplasm, cell cycle regulator, regulation of CDK activity. Strongly0.04RGC32 proteinsimilar to RGC-32.#(NOCAT)Target ExonFunction unknown0.04N72403Homo sapiens cDNA clone IMAGE: 245132Function unknown0.05BE003054Hs.1695:46MMP12Zinc binding, cell motility, macrophage elastase, extracellular matrix,0.05matrix metalloproteinase 12 (macrophageproteolysis and peptidolysis. Matrix metalloprotease; degradeselastase)elastinAL035588Hs.153203:26, Hs.23391:1Human DNA sequence from clone 696P19 onFunction unknown0.05chromosome 6p12.3-21.2. Contains the gene forTFEB, an NPM1 (Nucleophosmin, Numatrin)pseudogene and the MDFI gene for MyoD familyinhibitor (myogenic repressor I-MF). ContainsESTs, STSs, GSSs and two putative CpG islands,complete sequenceAI080491Hs.93270:3ESTs, Moderately similar to S65657 alpha-1C-Function unknown0.05adrenergic receptor splice form 2 [H. sapiens]AW770994Hs.30340:125hypothetical protein KIAA1165Function unknown0.05H24177Hs.75262:69, Hs.238912:1CTSOCysteine-type endopeptidase, proteolysis and peptidolysis.0.05cathepsin OCathepsin O; cysteine (thiol) proteaseAF146781Hs.20450:29BCM-like membrane protein precursorFunction unknown0.05NM_001955Hs.2271:45, Hs.306:1EDN1Circulation, peptide hormone, soluble fraction, signal transduction,0.05endothelin 1extracellular space, cell—cell signalling, blood pressure regulation,positive control of cell proliferation. Preproendothelin 1; precursor ofthe hormone endothelin 1AI680737Hs.289068:204, Hs.326198:1TCF4Nucleus, RNA polymerase II transcription factor, transcription0.05transcription factor 4regulation from Pol II promoter. Transcriptional activator; interactswith ITF1 (TCF3); contains basic helix-loop-helix domain|ProteomeAI752666Hs.76669:183NNMTNicotinamide N-methyltransferase; catalyzes the N-methylation of0.05nicotinamide N-methyltransferasenicotinamide and other pyridines, structurally-related drugs andxenobiotics|ProteomeAA505445Hs.300697:21IGHG3Constant region of heavy chain of IgG30.05immunoglobulin heavy constant gamma 3 (G3mmarker)BE246649;Hs.345728SOCS3suppression of IL-6 mediated signalling0.02NM_003955STAT induced STAT-inhibitor 3; suppressor ofcytokine signalling 3M86849Hs.323733:62, Hs.300816:5GJB2 gap junction protein, beta 2, 26 kD (connexinHearing, connexon, plasma membrane, connexon channel, cell—cell0.0026)signalling, small molecule transport. Connexin 26; gap junctionprotein expressed in various tissues including cochlea.AW963419Hs.155223:20STC2 stanniocalcin 2Peptide hormone, cell—cell signalling, glycopeptide hormone,0.00nutritional response pathway, cell surface receptor linked signaltransduction. Stanniocalcin 2; may regulate metal ion homeostasisand inhibits phosphate uptake.BE298665Hs.14846:132Homo sapiens mRNA; cDNA DKFZp564D016Function unknown0.00(from cloneAK000637Hs.46624:11HSPC043 HSPC043 proteinFunction unknown0.00BE077546Hs.31447:27ESTs, Moderately similar to A46010 X-linkedFunction unknown0.00retinopathy protein [H. sapiens]T97307gb: ye53h05.s1 Soares fetal liver spleen 1NFLSFunction unknown0.00Homo sapiens cDNA clone IMAGE: 121497 3′,mRNA sequence.R24601Hs.108300:46Homo sapiens adenylosuccinate synthetaseFunction unknown0.00isozyme (ADSS) mRNA, complete cdsBE090176Hs.179902:95Interim-CDw92 antigencholine transporter-like protein0.00AA393907Hs.97179:22ESTsFunction unknown0.00W28729Hs.236510:6Homo sapiens mRNA; cDNA DKFZp666D074Function unknown0.00(from clone DKFZp666D074)BE313754Hs.13350:52Homo sapiens mRNA; cDNA DKFZp586D0918Function unknown0.01AW673081Hs.54828:9ESTsFunction unknown0.01AA356694Hs.94011:42, Hs.7744:2,HCA4 Hepatocellular carcinoma-associatedFunction unknown0.01Hs.231043:1protein HCA4L08239Hs.5326:11MG61 Porcupineamino acid system N transporter 2;0.01BE397649Hs.94109:40Homo sapiens cDNA FLJ34399 fis, cloneFunction unknown0.01HCHON2001359NM_012317Hs.45231:36LDOC1 Leucine zipper, down-regulated in cancer 1Nucleus, negative control of cell proliferation. Nuclear protein;0.01contains a leucine zipper-like motifNM_000947Hs.74519:20PRIM2A primase, polypeptide 2A (58 kD)DNA primase, DNA replication, priming, alpha DNA0.01polymerase: primase complex. Subunit of DNA primase polypeptide2A; part of the DNA polymerase alpha-primase complexAJ250562Hs.82749:133Homo sapiens partial TM4SF2 gene forFunction unknown0.01tetraspanin protein, exon 1 and joined CDSAL040183Hs.123484:24, Hs.326906:1Homo sapiens mRNA; cDNA DKFZp686E1934Function unknown0.01(from clone DKFZp686E1934)BE207573Hs.83321:32NMB neuromedin BPeptide hormone, soluble fraction, signal transduction, cell—cell0.01signalling. Precursor of neuromedin B, a C-terminally amidatedpeptide hormone; similar to bombesinBE564162Hs.250820:45FLJ14827 hypothetical protein FLJ14827Function unknown0.01BE439580Hs.75498:40SCYA20 Small inducible cytokine subfamily AChemokine, chemotaxis, immune response, signal transduction,0.01(Cys—Cys), member 20extracellular space, cell—cell signalling, inflammatory response,antimicrobial humoral response. Cytokine A20 (exodus); chemotacticfactor for lymphocytes, but not a chemotactic factor for monocytesAW067800Hs.155223:52STC2 stanniocalcin 2Peptide hormone, cell—cell signalling, glycopeptide hormone,0.01nutritional response pathway, cell surface receptor linked signaltransduction. Stanniocalcin 2; may regulate metal ion homeostasisand inhibits phosphate uptake.AA569756Hs.87803:10Homo sapiens cDNA FLJ30156 fis, cloneFunction unknown0.01BRACE2000487AW138190Hs.180248:8ZNF124 zinc finger protein 124 (HZF-16)DNA binding. C2H2 zinc-finger protein 1240.01AF126245Hs.14791:48ACAD8 acyl-Coenzyme A dehydrogenase family,Lipid metabolism, acyl-CoA dehydrogenase. Member of the acyl-0.01member 8Coenzyme A dehydrogenase family; alpha, beta-dehydrogenatesacyl-CoA estersL10343Hs.112341:96, Hs.1968:1Homo sapiens elafin precursor, gene, completeelastase-specific inhibitor in bronchial secretions0.01cdsNM_002514Hs.235935:38NOV nephroblastoma overexpressed geneInsulin-like growth factor receptor binding protein. Insulin-like growth0.01factor binding protein; may play a role in nephrogenesisAI863735Hs.186755:3ESTsFunction unknown0.01NM_005397Hs.16426:160, Hs.248780:1PODXL podocalyxin-likeIntegral plasma membrane protein. Transmembrane protein similar0.01to rodent podocalyxinsW26391Hs.301206:100KIF3B kinesin family member 3BPlus-end kinesin, microtubule motor, anterograde axon cargo0.01transport, plus-end-directed kinesin ATPase, determination of left-right asymmetry. Similar to murine Kif3b; may have a role inintracellular organelle transport, may act in left-right determination inembryogenesis; are a microtubule-associated motor proteinH15474Hs.132898:156FADS1 fatty acid desaturase 1C-5 sterol desaturase, fatty acid desaturation, integral membrane0.01protein. Delta-5 desaturase; catalyzes production of polyenoic fattyacids such as arachldonic acidU51166Hs.173824:106TDG Thymine-DNA glycosylaseDNA repair, nucleoplasm, damaged DNA binding, base-excision0.01repair, G/T-mismatch-specific thymine-DNA glycosylase. Thymine-DNA glycosylase; excises uracil and thymine from mispairs withguanidineAA243499Hs.104800:23FLJ10134 hypothetical protein FLJ10134Highly similar to murine p19.5; are a membrane protein0.01AW408807Hs.34497:46FLJ22116 hypothetical protein FLJ22116Function unknown0.01AI738719Hs.198427:98HK2 Hexokinase 2Hexokinase, cell cycle control, glucose catabolism, glucose0.01metabolism, mitochondrial outer membrane. Hexokinase II; convertsaldo- and keto-hexose sugars to the hexose-6-phosphateAB040888Hs.41793:110Homo sapiens mRNA for KIAA1455 protein, partialFunction unknown0.01cdsBE313077Hs.93135:40, Hs.228357:1Homo sapiens cDNA FLJ39971 fis, cloneFunction unknown0.01SPLEN2028066AI677897Hs.76640:124RGC32 RGC32 proteinCytoplasm, cell cycle regulator, regulation of CDK activity. Strongly0.01similar to RGC-32C14898Hs.192986:5ESTsFunction unknown0.01AI821730Hs.116524:7Homo sapiens cDNA FLJ35800 fis, cloneFunction unknown0.01TESTI2005933AF007393Hs.177574:111PRKRIR protein-kinase, interferon-inducibleStress response, protein binding, signal transduction, translational0.01double stranded RNA dependent inhibitor,regulation, negative control of cell proliferation. Regulates interferon-repressor of (P58 repressor)induced protein kinase PKR (PRKR) activity by binding and inhibitingthe PKR-regulator P58IPK (PRKRI)H65423Hs.17631:42DKFZP434E2135 hypothetical proteinFunction unknown0.01DKFZp434E2135N46243Hs.110373:26ESTs, Highly similar to T42626 secreted leucine-Function unknown0.01rich repeat-containing protein SLIT2 - mouse(fragment) [M. musculus]AA095971Hs.198793:56, Hs.309674:7Homo sapiens cDNA: FLJ22463 fis, cloneFunction unknown0.01HRC10126U20350Hs.78913:33CX3CR1 chemokine (C—X3—C) receptor 1Virulence, chemotaxis, coreceptor, cell adhesion, plasma0.01membrane, chemokine receptor, response to wounding, cellulardefense response, integral plasma membrane protein, G-proteinlinked receptor protein signalling pathway. CX3C chemokinereceptor; G protein-coupled receptor, mediates leukocyte migrationand adhesion, binds the CX3C chemokine fractalkine and signalsthrough a pertussis toxin sensitive G-proteinNM_005756Hs.184942:18GPR64 G protein-coupled receptor 64Spermatogenesis, G-protein linked receptor, integral plasma0.01membrane protein, G-protein linked receptor protein signallingpathway. Member of the G protein-coupled receptor familyD19589Hs.13453:87FLJ14753 hypothetical protein FLJ14753Function unknown0.02AW957446Hs.301711:74ESTsFunction unknown0.02AW294647Hs.233634:40C20orf39 chromosome 20 open reading frame 39Function unknown0.02BE159718Hs.85335:46Homo sapiens, clone IMAGE: 4513159, mRNAFunction unknown0.02AI888490Hs.55902:22EDG3 endothelial differentiation, sphingolipid G-Lipid binding, plasma membrane, inflammatory response, G-protein0.02protein-coupled receptor, 3linked receptor, embryogenesis and morphogenesis, integral plasmamembrane protein, positive control of cell proliferation, cytostoliccalcium ion concentration elevation, G-protein linked receptor proteinsignalling pathway. Lysosphingolipid receptor, a G protein-coupledreceptor; activates calcium flux and serum response element driventranscriptionAA022569Hs.29802:35, Hs.271785:1ESTsFunction unknown0.02BE147740Hs.104558:21ESTs, Moderately similar to hypothetical proteinFunction unknown0.02FLJ20378 [Homo sapiens]AI798863Hs.87191:8ESTsFunction unknown0.02BE464341Hs.21201:18Interim-DKFZP566B0846: nectin 3Low similarity to PVRL1; are a membrane glycoprotein; contains an0.02immunoglobulin (Ig) domainAL080235Hs.35861:34, Hs.289068:1RIS1 Ras-induced senescence 1Rat brain specific binding protein0.02AI557212Hs.17132:102, Hs.330782:1ESTsFunction unknown0.02X75208Hs.2913:41EPHB3 EphB3Signal transduction, integral plasma membrane protein,0.02transmembrane receptor protein tyrosine kinase. Eph-relatedreceptor tyrosine kinase B3AA628980Hs.192371:3DSCR8 Down syndrome critical region proteinMelanoma-testis-associated protein 20.02DSCR8BE242587Hs.118651:39HHEX hematopoietically expressed homeoboxNucleus, DNA binding, transcription factor, developmental0.02processes, antimicrobial humoral response. Member of thehomeodomain family of DNA binding proteins; may regulate geneexpression, morphogenesis, and differentiationNM_005512Hs.151641:65GARP glycoprotein A repetitions predominantIntegral plasma membrane protein. Putative transmembrane cell0.02surface protein; has an extracellular domain comprised largely ofleucine-rich repeatsAW953853Hs.292833:19PAEP progestagen-associated endometrial proteinDevelopmental processes. Placental protein 14 (Glycodelin);0.02(placental protein 14, pregnancy-associatedmember of lipocalin superfamily, highly similar to beta-lactoglobulinsendometrial alpha-2-globulin, alpha uterineprotein)AU076611Hs.154672:122MTHFD2 methylene tetrahydrofolateMitochondrion, electron transporter, methenyltetrahydrofolate0.02dehydrogenase (NAD dependent),cyclohydrolase, methylenetetrahydrofolate dehydrogenase. NAD-methenyltetrahydrofolate cyclohydrolasedependent methylene tetrahydrofolate dehydrogenase-cyclohydrolase; may provide formyltetrahydrofolate forformylmethionyl tRNA synthesis; involved in initiation ofmitochondrial protein synthesisAW968613Hs.79428:166BNIP3 BCL2/adenovirus E1B 19 kD-interactingAnti-apoptosis, apoptosis inhibitor. Bcl2-related protein 3; binds0.02protein 3antiapoptotic viral E1B 19 kDa protein and cellular Bcl2 proteinAL353944Hs.50115:14Homo sapiens mRNA; cDNA DKFZp761J1112Function unknown0.02(from clone DKFZp761J1112)BE614149Hs.20814:29, Hs.306626:27LOC51072: C21orf19-like proteinFunction unknown0.02AA292998Hs.163900:25ESTsHighly similar to winged helix/forkhead transcription factor0.02H12912Hs.274691:138AK3 adenylate kinase 3Nucleobase, nucleoside, nucleotide and nucleic acid metabolism.0.02Adenylate kinase 3; strongly similar to murine Ak4AA188763Hs.36793:4SLC12A8 solute carrier family 12Solute carrier family 12 (potassium/chloride transporters), member 80.02(potassium/chloride transporters), member 8AK000596Hs.3618:56HPCAL1 hippocalcin-like 1Calcium-binding protein with similarity to hippocalin (human HPCA);0.02expressed only in the brain.AI970797Hs.64859:16ESTsFunction unknown0.02AW519204Hs.40808:22ESTsFunction unknown0.02Z42387Hs.83883:114TMEPAI transmembrane, prostate androgenFunction unknown0.02induced RNAAF145713Hs.61490:51SCHIP1 schwannomin-interacting protein 1Cytoplasm. Associates with the neurofibromatosis type 2 protein0.02schwannomin (NF2); contains a coiled-coil domainAA972412Hs.13755:41FBXW2 f-box and WD-40 domain protein 2Protein modification, ubiquitin-protein ligase, proteolysis and0.02peptidolysis, ubiquitin conjugating enzyme. F-box and WD-40domain protein 2; putative SCF ubiquitin ligase subunit involved inprotein degradation; contains a WD-40 domain and an F-boxAK001564Hs.104222:139, Hs.296267:4Homo sapiens cDNA FLJ10702 fis, cloneMember of the ADP-ribosylation factor (ARF) family; putative GTP-0.02NT2RP3000759, weakly similar to ADP-binding protein involved in protein traffickingRIBOSYLATION FACTORAW959861Hs.290943:28ESTsFunction unknown0.02BE313555Hs.7252:158RAI17 retinoic acid induced 17Function unknown0.02W25005Hs.24395:199zb67e02.r1 Soares_fetal_lung_NbHL19W HomoFunction unknown0.02sapiens cDNA clone IMAGE: 308666 5′, mRNAsequenceAI193356Hs.160316:3ESTsFunction unknown0.02AF111106Hs.3382:223PPP4R1 Protein phosphatase 4, regulatoryProtein phosphatase0.02subunit 1AI130740Hs.6241:116PIK3R1 phosphoinositide-3-kinase, regulatoryA family of enzymes that phosphorylate the 3′-hydroxyl of0.02subunit, polypeptide 1 (p85 alpha)phosphatidylinositol (Ptdins).AA985190Hs.246875:42FLJ20059 hypothetical protein FLJ20059Contains four Kelch motif domains0.02BE221880Hs.268555:144XRN2 5′-3′ exoribonuclease 2Nucleus, nuclease, recombination, RNA catabolism, RNA0.03processing. 5′-3′ Exoribonuclease; similar to Schizosaccharomycespombe Dhp1pAF084545Homo sapiens versican Vint isoform, mRNA,Function unknown0.03partial cdsR26584Hs.267993:43TAPBP-R: TAP binding protein relatedHas low similarity to TAPBP (Tapasin); contains two immunoglobulin0.03(Ig) domains|ProteomeAW247380Hs.12124:116ELAC2 elaC homolog 2 (E. coli)putative prostate cancer susceptibility protein0.03AA364261Hs.131365:7ESTsWeakly similar to T31613 hypothetical protein Y50E8A.I -0.03Caenorhabditis elegans [C. elegans]U25849Hs.75393:141ACP1 Human red cell-type low molecular weightAcid phosphatase0.03acid phosphatase (ACP1) gene, exon 6 and 7,complete cdsAF262992Hs.123159:14SPAG4 Sperm associated antigen 4Spermatogenesis, structural protein. Sperm associated antigen 4;0.03predicted ortholog of rat SPAG4, which interacts with rat ODF27, the27 kDa outer dense fiber protein of elongating spermatidsAW342140Hs.182545:1ESTs, Weakly similar to POL2_MOUSEFunction unknown0.03Retrovirus-related POL polyproteinAL133572Hs.199009:58PCCX2 protein containing CXXC domain 2DNA-binding protein with PHD finger and CXXC domain, is regulated0.03by proteolysis.AI497778Hs.20509:4HBXAP Hepatitis B virus x associated proteinWeakly similar to Drosophila CG86770.03AI745379Hs.42911:31TAF13 TAF13 RNA polymerase II, TATA boxTFIID complex, protein binding, transcription factor, general RNA0.03binding protein (TBP)-associated factor, 18 kDpolymerase II transcription factor. TBP-associated factor K;component of TFIID complexes containing TAFII30 (TAF2H)U51712Hs.13775:135LAGY: lung cancer-associated Y proteinThe protein encoded by this gene is a lung cancer associated0.03protein. The function of the protein is not known. Multiplealternatively spliced transcript variants have been described for thisgene but some of their full length sequence has not beendetermined.AW375974Hs.156704:4ESTsFunction unknown0.03AF251237Hs.112208:16GAGED2 G antigen, family D, 2GAGE genes are expressed in a variety of tumors and in some fetal0.03and reproductive tissues. This gene is strongly expressed in Ewing'ssarcoma, alveolar rhabdomyosarcoma and normal testis. The proteinencoded by this gene contains a nuclear localization signal andshares a sequence similarity with other GAGE/PAGE proteins.Because of the expression pattern and the sequence similarity, thisprotein also belongs to a family of CT (cancer-testis) antigens.NM_000636Homo sapiens superoxide dismutase 2,Mitochondrion, oxidative stress response, manganese superoxide0.02mitochondrial (SOD2), mRNA. expression)dismutase. Manganese superoxide dismutase; intramitochondrial(RFX2), mRNA.free radical scavenging enzyme; has strong similarity to murineSod2.AA130986Hs.271627:1ESTsFunction unknown0.01AA216363Hs.262958:48, Hs.327737:2DKFZP434B044 hypothetical proteinFunction unknown0.01DKFZp434B044AA628980Hs.192371:3DSCR8 down syndrome critical region proteinFunction unknown0.00DSCR8AA811657Hs.220913:9Homo sapiens cDNA FLJ40827 fis, cloneFunction unknown0.02TRACH2011500AA897108gb: am08a06.s1 Soares_NFL_T_GBC_S1 HomeFunction unknown0.01sapiens cDNA clone 3′, mRNA sequenceAB040888Hs.41793:110Homo sapiens mRNA for KIAA1455 protein, partialFunction unknown0.02cdsAF212225Hs.283693:104Homo sapiens BM022 mRNA, complete cdsFunction unknown0.02AI089575Hs.9071:52ESTsFunction unknown0.02AI282028Hs.25205:10ESTsFunction unknown0.02AI368826Hs.30654:15FLJ10849: hypothetical protein FLJ10849Moderately similar to members of the septin family0.02AI718702Hs.308026:11, Hs.194490:6HLA-DRB3 major histocompatibility complex, classSignal transduction, integral plasma membrane protein, class II0.02II, DR beta 5major histocompatibility complex antigen. Beta 3 chain of HLA-DR;subunit of MHC class II molecule, complex binds peptides andpresents them to CD4+ T lymphocytesAI827248Hs.224398:3Homo sapiens cDNA FLJ11469 fis, cloneFunction unknown0.01HEMBA1001658AK002039Hs.26243:38MRVI1 murine retrovirus integration site 1 homologOncogenesis, tumor suppressor, endoplasmic reticulum membrane.0.02Similar to human MLRP; may act as a tumor suppressorAL109791Hs.241559:3Homo sapiens mRNA full length insert cDNA cloneFunction unknown0.00EUROIMAGE 151432AW090198Hs.4779:29LOC127829: hypothetical protein BC015408Function unknown0.01AW296454Hs.24743:92FLJ20171: hypothetical protein FLJ2017Contains three RNA recognition motifs (RRM, RBD, or RNP)0.02AW445034Hs.256578:4ESTsFunction unknown0.00AW452948Hs.257631:3ESTsFunction unknown0.01AW470411Hs.288433:27HNT: neurotriminCell adhesion, neuronal cell recognition, integral plasma membrane0.02protein. Neurotrimin; may function as a GPI-anchored neural celladhesion molecule; member of the immunoglobulin superfamilyAW885727Hs.301570:22FST follistatinDevelopmental processes. Follistatin; inhibits the release of follicle-0.01stimulating hormone (FSH)AW970859Hs.313503:4ESTsFunction unknown0.02AW979189Hs.283367:3ESTsFunction unknown0.01BE165866Hs.83623:66Human XIST, coding sequence “a” mRNA (locusXIST mRNA0.01DXS399E)BE175582gb: RC5-HT0580-100500-022-C01 HT0580 HomoFunction unknown0.01sapiens cDNA, mRNA sequenceBE242587Hs.118651:39HHEX hematopoletically expressed homeoboxNucleus, DNA binding, transcription factor, developmental0.01processes, antimicrobial humoral response. Member of thehomeodomain family of DNA binding proteins; may regulate geneexpression, morphogenesis, and differentiation|BE271927Hs.87385:31, Hs.307940:4LOC115416: hypothetical protein BC012331Function unknown0.01BE439580Hs.75498:40SCYA20 small inducible cytokine subfamily AChemokine, chemotaxis, immune response, signal transduction,0.02(Cys—Cys), member 20extracellular space, cell—cell signalling, inflammatory response,antimicrobial humoral response. Cytokine A20 (exodus); chemotacticfactor for lymphocytes, but not a chemotactic factor for monocytesBE464016Hs.238956:35Homo sapiens cDNA FLJ37793 fis, cloneFunction unknown0.02BRHIP3000473D63216Hs.153684:137FRZB frizzled-related proteinMembrane, extracellular, skeletal development. Frizzled-related0.02protein; similar to frizzled family of receptorsF34856Hs.292457:120Homo sapiens, clone MGC: 16362Function unknown0.02IMAGE: 3927795, mRNA, complete cdsM83822Hs.62354:112LRBA LPS-responsive vesicle trafficking, beachMay mediate protein-protein interactions; contains two WD domains0.02and anchor containing(WD-40 repeats) and a beige/BEACH domain|ProteomeN33937Hs.10336:6ESTsFunction unknown0.01N49068Hs.93966:4ESTsFunction unknown0.01N51357Hs.260855:62NSE1: NSE1Function unknown0.02N80486Hs.39911:17Homo sapiens mRNA for FLJ00089 protein, partialFunction unknown0.02cdsNM_000954Hs.8272:265, Hs.332355:1PTGDS prostaglandin D2 synthase (21 kD, brain)Membrane, prostaglandin-D synthase. Glutathione-independent0.02prostaglandin D2 synthase; membrane associated, catalyzessynthesis of prostaglandin D; member of the lipocalin family oftransportersNM_005756Hs.184942:18GPR64 G protein-coupled receptor 64Spermatogenesis, G-protein linked receptor, integral plasma0.02membrane protein, G-protein linked receptor protein signallingpathway. Member of the G protein-coupled receptor familyNM_016652Hs.268281:61CRNKL1 Cm, crooked neck-like 1 (Drosophila)Function unknown0.02R26584Hs.267993:43TAPBP-R: TAP binding protein relatedHas low similarity to TAPBP (Tapasin); contains two immunoglobulin0.01(lg) domainsR31178Hs.287820:6FN1 fibronectin 1Cell adhesion, cell motility, cell adhesion, soluble fraction, signal0.02transduction, extracellular matrix, extracellular space. Fibronectin 1;member of family of proteins found in plasma and extracellularmatrixW05391Hs.83623:8Homo sapiens cDNA FLJ30298 fis, cloneFunction unknown0.02BRACE2003172W25005Hs.24395:199zb67e02.r1 Soares_fetal_lung_NbHL19W HomoFunction unknown0.01sapiens cDNA clone IMAGE: 308666 5′, mRNAsequenceW45393Hs.55888:15ATF7 activating transcription factor 7Transcription factor. Leucine zipper DNA-binding protein; recognizes0.02a cAMP response element (CRE), involved in the regulation ofadenovirus Ela-responsive and cellular cAMP-inducible promotersW68815Hs.301885:20Homo sapiens cDNA FLJ33794 fis, cloneFunction unknown0.01CTONG1000009X65965H. sapiens SOD-2 gene for manganese superoxideMitochondrion, oxidative stress response, manganese superoxide0.01dismutasedismutase. Manganese superoxide dismutase; intramitochondrialfree radical scavenging enzyme; has strong similarity to murineSod2.X76732Hs.3164:58NUCB2 nucleobindin 2Cytosol, DNA binding, plasma membrane, calcium binding,0.02extracellular space. Nucleobindin 2; may bind DNA and calcium; hasDNA-binding and EF-hand domains, and a leucine-zipperZ45051Hs.22920:25C20orf103 chromosome 20 open reading frameLow similarity to a region of murine Lamp1|Proteome0.02103c. downregulated genesNM_022117Hs.136164:23SE20-4, cutaneous T-cell lymphoma-associatedCutaneous T-cell lymphoma-associated tumor antigen se20-4se20-0tumor antigen se20-4se20-44; differentially expressed nucleolar TGF-beta1 target protein(DENTT); also known as CDA1NM_005460Hs.24948:32,SNCAIP, synuclein, alpha interacting proteinCytoplasm, pathogenesis, protein binding. Synphilin-1; promotes0Hs.300445:4(synphilin)formation of cytosolic inclusions in neurons (SNCAIP). Synucleinalpha interacting protein contains several protein-protein interactiondomains and interacts with alpha synuclein in neurons. Mutations ofSNCAIP have been linked to Parkinson disease.NM_002387Hs.1345:5MCC, mutated in colorectal cancersReceptor, signal transduction, tumor suppressor. Similar to the G0protein-coupled m3 muscarinic acetylcholine receptor. MCC is acandidate for the putative colorectal tumor suppressor gene. TheMCC gene product are involved in early stages of colorectalneoplasia in both sporadic and familial tumors.AI745249Hs.23650:30Homo sapiens, clone MGC: 9889 IMAGE: 3868330Function unknown0.0009AI694200Hs.356620,ESTsFunction unknown0.0442Hs.227913:11









TABLE 2










Genes having modified expression in serous ovarian cancer relative to normal ovarian tissue











Accession number
UniGene Mapping
Gene symbol and title
Putative Function
Ratio














M25809
Hs.64173
ATP6V1B1, ATPase, H+ transporting, lysosomal
Subunit B1 (beta subunit) of a vacuolar-type H+-ATPase 1; apical proton
1062.30




56/58 kD, V1 subunit B, isoform 1 (Renal tubular
pump that mediates distal nephron acid secretion




acidosis with deafness)


AW959311
Hs.172012
DKFZP434J037: hypothetical protein DKFZp434J037
Function unknown
227.83


H16423
Hs.82685

Homo sapiens mRNA; cDNA DKFZp313F0317 (from

Function unknown
74.54




clone DKFZp313F0317)


AI733848
Hs.71935
ZNF339, zinc finger protein 339
Zinc finger protein
55.13


AW055308
Hs.31803
NAC1, transcriptional repressor NAC1
Function unknown
52.63


AF034102
Hs.32951
SLC29A2, solute carrier family 29 (nucleoside
Nitrobenzylthioinosine-insensitive equilibrative nucleoside transporter 2;
44.34




transporters), member 2
may act in the uptake of purine and pyrimidine nucleosides


AI791905
Hs.95549
FLJ20273: RNA-binding protein
Contains four RNA recognition motifs (RRM, RBD, or RNP)
43.21


AW296454
Hs.24743
FLJ20171: hypothetical protein FLJ20171
Contains three RNA recognition motifs (RRM, RBD, or RNP)
38.91


Z43989
Hs.82141
Human clone 23612 mRNA sequence
Function unknown
37.89


AL043980
Hs.7886
PELI1, pellino homolog 1 (Drosophila)
Pellino protein
35.20


BE514982
Hs.38991
S100A2, S100 calcium binding protein A2
S100 calcium-binding protein A2; Interacts with target proteins to link
34.53





extracellular stimuli and cellular responses; member of the S100





tissue/cell specific Ca2+-binding protein family




Target Exon
Function unknown
34.02


AI811807
Hs.108646

Homo sapiens cDNA FLJ12534 fis, clone

Function unknown
32.34




NT2RM4000244


U90441
Hs.3622
P4HA2, procollagen-proline, 2-oxoglutarate 4-
Alpha 2 subunit of prolyl 4-hydroxylase; catalyzes the formation of 4-
32.24




dioxygenase (proline 4-hydroxylase), alpha
hydroxyproline in collagens


T98226
Hs.171952
OCLN, occludin
This gene encodes an integral membrane protein which is located at
31.56





tight junctions. This protein are involved in the formation and





maintenance of the tight junction.


R35343
Hs.24968
Human DNA sequence from clone RP1-233G16 on

31.22




chromosome Xq22.1-23. Contains the 5′ part of a




novel gene, ESTs, STSs, GSSs and a putative CpG




island


BE247295
Hs.78452
SLC20A1, solute carrier family 20 (phosphate
Sodium-dependent phosphate symporter; acts as a cell-surface receptor
30.16




transporter), member 1
for gibbon ape leukemia virus


AB037734
Hs.4993
PCDH19, protocadherin
Protocadherin
29.90




C5000394*: gi|12737280|ref|Xp_006682.2| keratin 18
Function unknown
29.30




[Homo sapiens]||6633


AF212223
Hs.25010

Homo sapiens BM025 mRNA, complete cds

Function unknown
28.85


AA902656
Hs.21943
NIF3L1, NIF3 (Ngg1 interacting factor 3, S. pombe
Amyotrophic lateral sclerosis 2 (juvenile) chromosome region, candidate 1
27.73




homolog)-like 1


X14008
Hs.234734
Human lysozyme gene (EC 3.2.1.17)
Lysozyme
27.66


AA570256

LOC116238: hypothetical protein BC014072
Function unknown
27.52


AA137152
Hs.286049
PSA, phosphoserine aminotransferase
The protein encoded by this gene is likely a phosphoserine
25.57





aminotransferase, based on similarity to proteins in mouse, rabbit, and






Drosophila. Alternative splicing of this gene results in two transcript






variants encoding different isoforms.


BE621807

TM4SF1, transmembrane 4 superfamily member 1
L6 antigen; member of the transmembrane 4 superfamily (TM4SF)
25.40


AB041036
Hs.57771
KLK11, kallikrein 11
Trypsin-like serine protease; has serine protease activity
25.05


F13386
Hs.7888

Homo sapiens clone 23736 mRNA sequence

Function unknown
22.50


AA158177
Hs.118722
FUT8, fucosyltransferase 8 (alpha (1,6)
N-linked glycosylation, oligosaccharide biosynthesis, glycoprotein 6-
21.90




fucosyltransferase)
alpha-L-fucosyltransferase. Alpha(1,6)fucosyltransferase (GDP-L-Fuc:N-





acetyl-beta-D-glucosaminide:alpha1-6 fucosyltransferase); transfers





fucose to N-linked type complex glycopeptides from GDP-Fuc; functions





in asparagine-linked glycoprotein oligosaccharide synthesis


BE267045
Hs.75064
TBCC, tubulin-specific chaperone c
Tubulin-specific chaperone c; cofactor in the folding pathway of beta-
21.49





tubulin, mediates the release of beta-tubulin polypeptides committed to





the native state




NM_005936: Homo sapiens myeloid/lymphoid or
Function unknown
20.46




mixed-lineage leukemia (trithorax (Drosophila)




homolog); translocated to, 4 (MLLT4), mRNA.


AA150864
Hs.790
MGST1, microsomal glutathione S-transferase 1
Microsome, glutathione transferase. Microsomal glutathione S-
20.35





transferase; catalyzes the conjugation of glutathione to electrophilic





compounds; member of a family of detoxication enzymes.


AW955632
Hs.66666
EST367702 MAGE resequences, MAGD Homo
Function unknown
20.26





sapiens cDNA, mRNA sequence



AW837046
Hs.6527
QV1-LT0037-150200-069-e09 LT0037 Homo sapiens
Function unknown
19.60




cDNA, mRNA sequence


AA286887
Hs.24724
MFHAS1, malignant fibrous histiocytoma amplified
The primary structure of its product includes an ATP/GTP-binding site,
19.16




sequence 1
three leucine zipper domains, and a leucine-rich tandem repeat, which





are structural or functional elements for interactions among proteins





related to the cell cycle, and which suggest that overexpression might be





oncogenic with respect to MFH.


AW401864
Hs.18720
PDCD8: programmed cell death 8 (apoptosis-inducing
Mitochondrial apoptosis-inducing factor; flavoprotein inducing chromatin
19.01




factor)
condensation and DNA fragmentation


AA196241
Hs.73980
zp98f03.r1 Stratagene muscle 937209 Homo sapiens
Function unknown
18.82




cDNA clone IMAGE: 628253 5′ similar to gb: M19309




TROPONIN T, SLOW SKELETAL MUSCLE




ISOFORMS (HUMAN);, mRNA sequence


NM_004998
Hs.82251
MYO1E, myosin IE
Highly similar to class I myosin; may bind proline-rich peptides; contains
18.62





an Src homology 3 (SH3) and myosin head domain (motor domain)


AW873704
Hs.320831
C20orf72: chromosome 20 open reading frame 72
Function unknown
18.19


AW361666
Hs.49500
KIAA0746: KIAA0746 protein
Function unknown
18.05


BE174595
Hs.366
PTS, 6-pyruvoyltetrahydropterin synthase
6-Pyruvoyltetrahydropterin synthase; synthesizes tetrahydrobiopterin,
17.28





activity requires sepiapterin reductase, Mg2+, and NADPH


M31669
Hs.1735
Human inhibin beta-B-subunit gene, exon 2, and
Function unknown
16.24




complete cds


AK001714
Hs.95744
FLJ10852, hypothetical protein similar to ankyrin
Are involved in protein-protein interactions; has five ankyrin repeats and
16.09




repeat-containing priotein AKR1
a DHHC-type zinc finger or NEW1 domain


AU076517
Hs.184276
AU076517 Sugano cDNA library Homo sapiens cDNA
Function unknown
16.05




clone ColF3365 similar to 5′-end region of Homo





sapiens ezrin-radixin-moesin binding phosphoprotein-





50 mRNA, mRNA sequence


NM_006456
Hs.288215
STHM, sialyltransferase
Low similarity to beta-galactosidase a-2,3-sialytransferase SIAT4B;
15.93





member of the sialyltransferase family


BE148235
Hs.193063

Homo sapiens cDNA FLJ14201 fis, clone

Function unknown
15.91




NT2RP3002955


AV653729
Hs.8185
SQRDL: sulfide dehydrogenase like (yeast)
Sulfide dehydrogenase like
15.35


AL119671
Hs.1420
FGFR3, fibroblast growth factor receptor 3
Fibroblast growth factor receptor 3; receptor tyrosine kinase that binds
14.62




(achondroplasia, thanatophoric dwarfism)
acidic and basic FGF


AA393071
Hs.182579
LAP3, leucine aminopeptidase
Leucine aminopeptidase
14.60


AL048753
Hs.303649
CCL2, chemokine (C—C motif) ligand 2
Cytokine A 2; chemotactic factor for monocytes
14.37


AI868872
Hs.282804
CP, ceruloplasmin (ferroxidase)
Ceruloplasmin; ferrous oxidase, binds copper in plasma and maintains
14.07





iron homeostasis


NM_004419
Hs.2128
DUSP5, dual specificity phosphatase 5
Mitogen inducible dual specificity protein phosphatase 5;
14.05





dephosphorylates extracellular signal-regulated kinase


AW969587
Hs.86366
EST381664 MAGE resequences, MAGK Homo
Function unknown
13.75





sapiens cDNA, mRNA sequence



AW161449
Hs.72290
WNT7A, wingless-type MMTV integration site family,
Very strongly similar to murine Wnt7a; may have a role in limb
13.48




member 7A
development and sexual dimorphism; member of the Wnt family of cell





signalling proteins


BE409838
Hs.194657
CDH1, cadherin 1, type 1, E-cadherin (epithelial)
E-cadherin (uvomorulin); Ca2+-dependent glycoprotein, mediates cell-
12.92





cell Interactions in epithelial cells


BE540274
Hs.239
FOXM1, forkhead box M1
Cell-cycle regulated HNF-3/fork head; a transcriptional regulator
12.86


AF022375
Hs.73793
VEGF, vascular endothelial growth factor
Vascular endothelial growth factor; induces endothelial cell proliferation
12.79





and vascular permeability


AW369278
Hs.23412
FLJ20160: hypothetical protein FLJ20160
Function unknown
12.73


AF147204
Hs.89414
CXCR4, chemokine (C—X—C motif), receptor 4 (fusin)
CXC chemokine receptor (fusin); G protein-coupled receptor binds CXC
12.56





cytokines, mediates intracellular calcium flux


BE242818
Hs.311609
DDX39, DEAD/H (Asp-Glu-Ala-Asp/His) box
Strongly similar to human D6S81E; member of the DEAD/H box ATP-
12.43




polypeptide 39
dependent RNA helicase family


NM_014791
Hs.184339
MELK, maternal embryonic leucine zipper kinase
Leucine zipper kinase
12.25


U38847
Hs.151518
TARBP1, TAR (HIV) RNA binding protein 1
Binds to the HIV-1 TAR RNA regulatory element, may function alone or
12.22





with HIV-1 Tat to disengage RNA polymerase II during transcriptional





elongation; has a leucine zipper


AW953575
Hs.303125
EST365645 MAGE resequences, MAGC Homo
Function unknown
12.21





sapiens cDNA, mRNA sequence



AI949095
Hs.67776
ESTs, Weakly similar to T22341 hypothetical protein

Homo sapiens, clone IMAGE: 5455669, mRNA, partial cds

12.08




F47B8.5 - Caenorhabditis elegans [C. elegans]


BE274530
Hs.273333
FLJ10986, hypothetical protein FLJ10986
Member of the FGGY carbohydrate kinase family
11.75


AB020676
Hs.21543
KIAA0869 protein
Function unknown
11.73




Target Exon
Function unknown
11.69


H48299
Hs.26126:33
CLDN10, claudin 10
Cell adhesion, integral plasma membrane protein, tight junction.
11.67


T34530
Hs.4210

Homo sapiens cDNA FLJ13069 fis, clone

Function unknown
11.50




NT2RP3001752


NM_022454
Hs.97984
SOX17, SRY (sex determining region Y)-box 17
SRY-related HMG-box transcription factor SOX17
11.42


AA737033
Hs.7155

Homo sapiens, clone IMAGE: 4428577, mRNA, partial

Function unknown
10.79




cds


AA433988
Hs.98502:8
MUC16, mucin 16, CA125
Mucin 16, Alias CA125 ovarian cancer antigen
10.52


H91282
Hs.286232

Homo sapiens cDNA: FLJ23190 fis, clone LNG12190

Function unknown
10.50


AW005054
Hs.47883
LOC57118: CamKI-like protein kinase
CamKI-like protein kinase; granulocyte-specific protein kinase that
10.49





activates ERK/MAP kinase activity; similar to Ca(2+)-calmodulin-





dependent kinase I (CamKI)


X69699
Hs.73149
PAX8, paired box gene 8
Member of the paired domain family of nuclear transcription factors; are
10.39





involved in the ribosome assembly, required for normal thyroid





development


AW382987
Hs.88474:42

Homo sapiens cDNA, mRNA sequence

Function unknown
10.21


AW957446
Hs.301711

Homo sapiens, clone MGC: 23936 IMAGE: 3838595,

Function unknown
10.12




mRNA, complete cds


AA361562
Hs.178761
POH1: 26S proteasome-associated pad1 homolog
Ubiquitin-dependent protein degradation
10.01


AA834626

RAD54L, RAD54 (S. cerevisiae)-like
Has likely roles in mitotic and meiotic DNA recombination and repair;
9.85





member of SNF2/SWI2 family of DNA-dependent ATPases


AI878927
Hs.79284
MEST, mesoderm specific transcript (mouse) homolog
Mesoderm specific protein; member of the alpha/beta hydrolase fold
9.83





family


AW074266
Hs.23071
LOC85439: stonin 2
Stonin 2
9.74


NM_000947
Hs.74519
PRIM2A, primase, polypeptide 2A (58 kD)
Subunit of DNA primase polypeptide 2A; part of the DNA polymerase
9.72





alpha-primase complex


NM_006187
Hs.56009
OAS3, 2′-5′-oligoadenylate synthetase 3 (100 kD)
Member of the 2′5′-oligoadenylate synthetase family
9.68


AW276858
Hs.81256
S100A4, S100 calcium binding protein A4 (calcium
Calcyclin (metastasis-associated) (S100 calcium-binding protein A4);
9.66




protein, calvasculin, metastasin, murine placental
interacts with targets to link extracellular stimull and cellular responses;




homolog)
member of the S100 family of tissue-specific calcium-binding proteins


T18997
Hs.180372
LOC139231: hypothetical protein BC016683
Function unknown
9.49


AA262294
Hs.180383
DUSP6, dual specificity phosphatase 6
Dual specificity protein phosphatase 6; selectively dephosphorylates and
9.48





inactivates MAP kinase


AA220238
Hs.94986
RPP38: ribonuclease P (38 kD)
Nucleus, ribonuclease P. Subunit p38 of ribonuclease P
9.41





ribonucleoprotein; processes 5′ ends of precursor tRNAs


AW505308
Hs.75812
PCK2, phosphoenolpyruvate carboxykinase 2
Phosphoenolpyruvate carboxykinase 2; forms phosphoenolpyruvate by
9.38




(mitochondrial)
decarboxylation of oxaloacetate at the rate-limiting step of





gluconeogenesis


AI186431
Hs.296638
PLAB: prostate differentiation factor
Macrophage inhibitory cytokine; member of a subgroup of the TGF-beta
9.12





superfamily


AI095718
Hs.135015

Homo sapiens cDNA FLJ40906 fis, clone

Function unknown
9.04




UTERU2004698, highly similar to Mus musculus




mRNA for thrombospondin type 1 domain


W70171
Hs.75939
UMPK, uridine monophosphate kinase
The protein encoded by this gene catalyzes the phosphorylation of
8.97





uridine monophosphate to uridine diphosphate. This is the first step in





the production of the pyrimidine nucleoside triphosphates required for





RNA and DNA synthesis. In addition, an allele of this gene may play a





role in mediating nonhumoral immunity to Hemophilus influenzae type B.


AI580935
Hs.105698

Homo sapiens cDNA FLJ31553 fis, clone

Function unknown
8.90




NT2RI2001178


AB040914
Hs.278628
ShrmL: Shroom-ralated protein
Shroom-related protein
8.87


AU076611
Hs.154672
MTHFD2, methylene tetrahydrofolate dehydrogenase
NAD-dependent methylene tetrahydrofolate dehydrogenase-
8.71




(NAD+ dependent), methenyltetrahydrofolate
cyclohydrolase; may provide formyltetrahydrofolate for formylmethionyl




cyclohydrolase
tRNA synthesis; Involved in initiation of mitochondrial protein synthesis


AI089660
Hs.323401
LOC84661: dpy-30-like protein
dpy-30-like protein
8.71


D13666
Hs.136348:228, Hs.80988:2
OSF-2: osteoblast specific factor 2 (fasciclin I-like)
Cell adhesion, skeletal development. Putative bone adhesion protein;
8.64





similar to the insect protein fasciclin I


AI798863
Hs.87191
ESTs
Function unknown
8.52


U78093
Hs.15154
SRPX, sushi-repeat-containing protein, X chromosome
Putative membrane protein with short consensus repeat (sushi) domains
8.51


AI669760
Hs.188881
ESTs
Function unknown
8.37


AI375726
Hs.279918
MGC2198: hypothetical protein MGC2198
Function unknown
8.37


AW271106
Hs.133294
ESTs
Function unknown
8.30


AK001782
Hs.15093
HSPC195: hypothetical protein HSPC195
Function unknown
8.18


AF019226
Hs.8036
RAB3D, RAB3D, member RAS oncogene family
GTP-binding protein; are involved in vesicle transport; member of the
7.94





RAB family of small GTPases


AW968343
Hs.24255
LOC150696: prominin-related protein
Prominin-related protein
7.90


AF111856
Hs.105039
SLC34A2, solute carrier family 34 (sodium phosphate),
Sodium-dependent phosphate transporter; member of the renal type II
7.87




member 2
co-transporter family


AA863360
Hs.26040

Homo sapiens, clone MGC: 40051 IMAGE: 5243005,

Function unknown
7.75




mRNA, complete cds


NM_005764
Hs.271473
DD96: epithelial protein up-regulated in carcinoma,
Up-regulated in malignant epithelial cells of renal cell carcinomas, and in
7.75




membrane associated protein 17
carcinomas of colon, breast and lung


AW360901
Hs.183047
MGC4399: mitochondrial carrier protein
Mitochondrial carrier protein MGC4399
7.71


AL353944
Hs.50115

Homo sapiens mRNA; cDNA DKFZp761J1112 (from

Function unknown
7.69




clone DKFZp761J1112)


H59799
Hs.42644
TXNL2, thioredoxin-like 2
Member of the thioredoxin family; has region of moderate similarity to
7.65





glutaredoxin-like proteins


NM_002984
Hs.75703
CCL4, chemokine (C—C motif) ligand 4
Cytokine A4
7.64


AA642452
Hs.130881
BCL11A, B-cell CLL/lymphoma 11A (zinc finger
May bind nucleic acids; contains three C2H2 type zinc finger domains
7.61




protein)


AA789081
Hs.4029
GAS41: giloma-amplified sequence-41
Similar to the transcription factors AF-9 and ENL
7.46


H13032
Hs.103378
MGC11034, hypothetical protein MGC11034
Function unknown
7.42


BE384836
Hs.3454
KIAA1821: KIAA1821 protein
KIAA1821 protein
7.40


AW067800
Hs.155223
STC2, stanniocalcin 2
Stanniocalcin 2; may regulate metal ion homeostasis and inhibits
7.36





phosphate uptake


T55979
Hs.115474
RFC3, replication factor C (activator 1) 3 (38 kD)
Subunit of replication factor C (activator 1) 3; activator of DNA
7.35





polymerases


AJ278016
Hs.55565
ANKRD3, ankyrin repeat domain 3
Ortholog of mouse protien kinase C-associated kinase, putative gene,
7.25





ankirin like, possible dual-specificity Ser/Thr/Tyr kinase domain




NM_025080: Homo sapiens hypothetical protein
Function unknown
7.22




FLJ22316 (FLJ22316), mRNA. VERSION




NM_025079.1 GI: 13376631


AA084248
Hs.85339:64
GPR39, G protein-coupled receptor 39
GPR39, G protein-coupled receptor 39
7.15


BE620738
Hs.173125
PPIF, peptidylprolyl isomerase F (cyclophilin F)
Cyclophilin F (peptidylprolyl isomerase F); binds the immunosuppressant
7.06





drug cyclosporin A


AF072873
Hs.114218
FZD6, frizzled (Drosophila) homolog 6
frizzled-6; may function in tissue polarity, development and
7.04





carcinogenesis; similar to frizzled receptor family, has seven





transmembrane domains


AA852773
Hs.334838
KIAA1866 protein
KIAA1866 protein
6.99


R07566
Hs.73817
CCL3, chemokine (C—C motif) ligand 3
Macrophage inflammatory protein 1 alpha; chemokine
6.98


NM_005211
Hs.174142
CSF1R, colony stimulating factor 1 receptor, formerly
Macrophage colony stimulating factor tyrosine kinase receptor; involved
6.79




McDonough feline sarcoma viral (v-fms) oncogene
in regulation of growth and differentiation of myeloid cells




homolog


AI752666
Hs.76669
NNMT, nicotinamide N-methyltransferase
Nicotinamide N-methyltransferase; catalyzes the N-methylation of
6.52





nicotinamide and other pyridines, structurally-related drugs and





xenobiotics


AF182294
Hs.241578
LOC51691: U6 snRNA-associated Sm-like protein
Member of the Sm family; core constituent of snRNP complexes
6.50




LSm8


AA457211
Hs.8858
BAZ1A, bromodomain adjacent to zinc finger domain,
May bind DNA and act as a chromatin-mediated transcriptional
6.48




1A
regulator; contains a bromodomain and a PHD-finger


W40262
Hs.146310
zc79f02.s1 Pancreatic Islet Homo sapiens cDNA clone
Function unknown
6.47




IMAGE: 328539 3′, mRNA sequence


AB033091
Hs.74313
KIAA1265 protein
Function unknown
6.45


AA292998
Hs.163900
ESTs, Highly similar to winged helix/forkhead
Function unknown
6.36




transcription factor [Homo sapiens] [H. sapiens]


BE613269
Hs.21893
DKFZp761N0624: hypothetical protein
Function unknown
6.35




DKFZp761N0624


H25836
Hs.301527
ESTs, Moderately similar to unknown [Homo sapiens]
Function unknown
6.27




[H. sapiens]


AL037228
Hs.82043
NUDT5, nudix (nucleoside diphosphate linked molety
NDP-sugar hydrolase; converts ADP-ribose to AMP or ribose 5-
6.25




X)-type motif 5
phosphate; contains a MulT motif


AV662037
Hs.124740
FLJ30532: hypothetical protein FLJ30532
Function unknown
6.21


AI674383
Hs.22891
wc38h08.x1 NCI_CGAP_Pr28 Homo sapiens cDNA
Function unknown
6.20




clone IMAGE: 2320959 3′, mRNA sequence


AW342140
Hs.182545
ESTs, Weakly similar to POL2_MOUSE Retrovirus-
Function unknown
6.18




related POL polyprotein [Contains: Reverse




transcriptase; Endonuclease] [M. musculus]


BE560135
Hs.5232
HSPC125, HSPC125 protein
Function unknown
6.17


BE409857
Hs.69499
HSPC132: hypothetical protein HSPC132
Moderately similar to a region of S. cerevisiae Ykl053c-ap
6.16


AW972542
Hs.289008
LOC116150: hypothetical protein, MGC: 7199
Function unknown
6.16


AI523755
Hs.59236
DKFZP434L0718: hypothetical protein
Function unknown
6.16




DKFZp434L0718


NM_014056
Hs.7917
DKFZP564K247: DKFZP564K247 protein
Function unknown
6.08


AI857607
Hs.181301
CTSS, cathepsin S
Cathepsin S; lysosomal cysteine (thiol) protease that cleaves elastin
6.04


AW247529
Hs.6793
PAFAH1B3, platelet-activating factor acetylhydrolase,
Platelet-activating factor acetylhydrolase gamma; may play a role in
5.98




isoform lb, gamma subunit (29 kD)
brain development


AK000868
Hs.5570

Homo sapiens cDNA FLJ10006 fis, clone

Function unknown
5.92




HEMBA1000168, weakly similar to CYLICIN I


AF053551
Hs.31584
MTX2, metaxin 2
Very strongly similar to murine metaxin 2 (Mm.12941); are involved in
5.91





mitochondrial protein import


AI538613
Hs.298241
TMPRSS3, Transmembrane protease, serine 3
The encoded protein contains a serine protease domain, a
5.86





transmembrane domain, a LDL receptor-like domain, and a scavenger





receptor cysteine-rich domain. This gene was identified as a tumor





associated gene that is overexpressed in ovarian tumors.


U48508
Hs.89631
Human skeletal muscle ryanodine receptor gene
Function unknown
5.86




(RYR1), exons 103, 104, 105, 106, and complete cds


T69387
Hs.76364
AIF1, allograft inflammatory factor 1
Allograft Inflammatory factor 1; cytokine inducible protein associated with
5.86





vascular injury


AC005954
Hs.25527

Homo sapiens chromosome 19, cosmid R28784,

Function unknown
5.86




complete sequence


AB037805
Hs.88442
KIAA1384 protein
Function unknown
5.84


AL031427
Hs.40094
Human DNA sequence from clone 167A19 on
Function unknown
5.83




chromosome 1p32.1-33. Contains three genes for




novel proteins, the DIO1 gene for type I iodothyronine




deiodinase (EC 3.8.1.4, TXDI1, ITDI1) and an HNRNP




A3 (Heterogenous Nuclear Ribonucleoprotein A3,




FBRNP) pseudogene.


AA340864
Hs.278562
CLDN7, claudin 7
Similar to murine Cldn7; are an integral membrane protein
5.76


X89984
Hs.211563
BCL7A, B-cell CLL/lymphoma 7A
Similar to the actin-binding protein caldesmon; serine-rich
5.74


AI355761
Hs.242463
qt94a11.x1 NCI_CGAP_Co14 Homo sapiens cDNA
Function unknown
5.73




clone IMAGE: 1962908 3′ similar to gb: X74929




KERATIN, TYPE II CYTOSKELETAL 8 (HUMAN);,




mRNA sequence


AA376409
Hs.10862

Homo sapiens cDNA: FLJ23313 fis, clone HEP11919

Function unknown
5.71


AA310162
Hs.169248
HCS: cytochrome c
Somatic cytochrome c (heart cytochrome c)
5.67


AW015534
Hs.217493
ANXA2, annexin A2
Annexin II (lipocortin-2); enhances osteoclast formation and bone
5.64





resorption; member of the annexin protein family


AA326108
Hs.53631:82
BHLHB3: basic helix-loop-helix domain containing,
Basic helix-loop-helix (bHLH) transcription factors (e.g., DEC1, also
5.64




class B, 3
called BHLHB2; 604256) are related to Drosophila hairy/enhancer of





split proteins. They are involved in the control of proliferation and





development during differentiation, particularly in neurons.


AA120865
Hs.23136
ESTs, Highly similar to THYA_HUMAN Prothymosin
Function unknown
5.62




alpha [H. sapiens]


AK000517
Hs.6844
NALP2: NALP2 protein
Protein with low similarity to murine Op1
5.54


Z36842
Hs.57548

H. sapiens (xs85) mRNA, 209 bp

Function unknown
5.53


AA831552
Hs.268016

Homo sapiens cDNA: FLJ21243 fis, clone COL01164


5.50


AL137578
Hs.27607

Homo sapiens mRNA; cDNA DKFZp564N2464 (from

Function unknown
5.50




clone DKFZp564N2464)


AA316181
Hs.61635
STEAP, six transmembrane epithelial antigen of the
Six transmembrane epithelial antigen of the prostate; prostate-specific
5.46




prostate
cell-surface antigen


X03635
Hs.1657
ESR1, estrogen receptor 1
Estrogen receptor, nuclear receptor transcription factor activated by
5.42





ligand-binding, involved in hormone-mediated inhibition of gene





expression


AI557280
Hs.184270
PT2.1_15_G11.r tumor2 Homo sapiens cDNA 3′,
Function unknown
5.41




mRNA sequence


AW248508
Hs.279727

Homo sapiens cDNA FLJ14035 fis, clone

Function unknown
5.40




HEMBA1004638


N90866
Hs.276770
CDW52, CDW52 antigen (CAMPATH-1 antigen)
CAMPATH-1 antigen; GPI-anchored protein
5.39


U83115
Hs.161002
AIM1, absent in melanoma 1
Member of the beta gamma-crystallin superfamily of proteins;
5.35





Interactions with the cytoskeleton


AB007860
Hs.12802
DDEF2, development and differentiation enhancing
GTPase-activating protein; Interacts with members of the Arf and Src
5.35




factor 2
family


Z46223
Hs.176663

H. sapiens DNA for immunoglobulin G Fc receptor IIIB

Immunoglobulin G Fc receptor
5.31


BE264974
Hs.6566
TRIP13; thyroid hormone receptor interactor 13
Interacts with ligand binding domain of thyroid hormone receptor and
5.30





with human papillomavirus type 16 (HPV16) E1


AA194422
Hs.22564
MYO6, myosin VI
Motor, hearing, myosin ATPase, structural protein. Class 6 myosin;
5.27





motor protein; very strongly similar to murine Myo6


AF134157
Hs.169487
MAFB, v-maf musculoaponeurotic fibrosarcoma
Very strongly similar to murine Krml; may function as a basic domain-
5.25




oncogene homolog B (avian)
leucine zipper transcription factor


AA232119
Hs.16085
SH120: putative G-protein coupled receptor
putative G-protein coupled receptor
5.25


W58353
Hs.285123
OSBPL10, oxysterol binding protein-like 10
Member of the oxysterol-binding protein (OSBP) family; may bind
5.21





oxygenated derivatives of cholesterol


AW167128
Hs.231934
ESTs, Weakly similar to A57717 transcription factor
Function unknown
5.19




EC2 - human [H. sapiens]


U70370
Hs.84136
PITX1, paired-like homeodomain transcription factor 1
Member of the homeodomain family of DNA binding proteins; may
5.18





regulate gene expression and control cell differentiation


N55669
Hs.333823
MRPL13, mitochondrial ribosomal protein L13
Protein of the large 60S ribosomal subunit
5.17


BE298446
Hs.305890
BCL2L1, BCL2-like 1
BCL2-related protein; alternative form bcl-xiong inhibits apoptosis and
5.17





bcl-xshort induces apoptosis


AW136551
Hs.181245

Homo sapiens cDNA FLJ12532 fis, clone

Function unknown
5.15




NT2RM4000200


AW250380
Hs.109059
HGS, hepatocyte growth factor-regulated tyrosine
Zinc-finger protein; interacts with STAM, undergoes tyrosine
5.13




kinase substrate
phosphorylation in response to IL2, CSF2, or HGF


AW002565
Hs.124660

Homo sapiens cDNA: FLJ21763 fis, clone COLF6967

Function unknown
5.13


AI697274
Hs.105435
GMDS, GDP-mannose 4,6-dehydratase
GDP-mannose-4,6-dehydratase; epimerase converts GDP-mannose to
5.11





GDP-mannose-4-keto-6-D-deoxymannose, plays a role in the synthesis





of fucosylated oligosaccharides


NM_003878
Hs.78619
GGH, gamma-glutamyl hydrolase (conjugase,
Gamma-glutamyl hydrolase; has greater exopeptidase activity on
5.11




folylpolygammaglutamyl hydrolase)
methotrexate pentaglutamate than on diglutamate


AF052112
Hs.12540
LYPLA1, lysophospholipase I
Lysophospholipid-specific lysophospholipase 1; hydrolyzes
5.09





lysophosphatidyl choline


AV654694
Hs.82316
IFI44, interferon-induced protein 44
Member of the family of interferon-alpha/beta inducible proteins; may
5.09





mediate the antiviral action of interferon


R24601

Home sapiens adenylosuccinate synthetase isozyme
Adenylosuccinate synthetase
5.07




(ADSS) mRNA, complete cds


BE019020
Hs.85838

Homo sapiens cDNA clone IMAGE: 2963945 5′ similar

Function unknown
5.04




to TR: O15427 O15427 MONOCARBOXYLATE




TRANSPORTER.;, mRNA sequence


AW163799
Hs.198365
BPGM, 2,3-bisphosphoglycerate mutase
2,3-bisphosphoglycerate mutase; has synthase, mutase, and
5.04





phosphatase activities, controls 2,3-diphosphoglycerate metabolism,





which is an effector for haemoglobin


AA278921
Hs.1908
PRG1, proteoglycan 1, secretory granule
Secretory granule proteoglycan 1
5.02


NM_003726
Hs.19126
SCAP1, src family associated phosphoprotein 1
Src kinase-associated phosphoprotein; acts as an adaptor protein;
5.02





contains a pleckstrin homology domain and an SH3 domain


AA281167
Hs.111911
ESTs, Weakly similar to T06291 extensin homolog
Function unknown
5.02




T9E8.80 - Arabidopsis thaliana [A. thaliana]




C9000306*: gi|12737280|ref|XP_006682.2| keratin 18
Function unknown
5.01




[Homo sapiens]||6633


AF098158
Hs.9329
C20orf1, chromosome 20 open reading frame 1
Proliferation-associated nuclear protein; associates with the spindle pole
5.00





and mitotic spindle during mitosis


AA101043
Hs.151254:19
KLK7, kallikrein 7 (chymotryptic; stratum comeum)
Epidermal differentiation. Stratum comeum chymotryptic enzyme; serine
4.87





protease. Growing evidence suggests that many kallikreins are





Implicated in carcinogenesis and some have potential as novel cancer





and other disease biomarkers. Thought to be involved in the proteolysis





of intercellular cohesive structures preceding desquamation, which is the





shedding of the outermost layer of the epidermis.


AF017986
Hs.31386:185

Homo sapiens secreted apoptosis related protein 1

Function unknown
4.12




(SARP1) mRNA, partial cds.


AW960564
Hs.3337:137
TM4SF1: transmembrane 4 superfamily member 1
Pathogenesis, plasma membrane, call proliferation, N-linked
3.62





glycosylation, integral membrane protein, integral plasma membrane





protein. L6 antigen; member of the transmembrane 4 superfamily





(TM4SF). The proteins mediate signal transduction events that play a





role in the regulation of cell development, activation, growth and motility.





This encoded protein is a cell surface antigen and is highly expressed in





different carcinomas.


W29092
Hs.7678:40
CRABP1 Cellular retinoic acid binding protein 1
Cytoplasm, retinoid binding, signal transduction, developmental
3.34





processes. Cellular retinoic acid-binding protein 1; are involved in





delivering retinoic acid to the nucleus, assumed to play an important role





in retinoic acid-mediated differentiation and proliferation processes.


H93366
Hs.7567:84

Homo sapiens cDNA: FLJ21962 fis, clone HEP05564

Function unknown
3.29


D49441
Hs.155981:53
MSLN, mesothelin
Cell adhesion, cell surface antigen, membrane. Pre-pro-megakaryocyte
3.14





potentiating factor. An antibody that reacts with ovarian cancers and





mesotheliomas was used to isolate a cell surface antigen named





mesothelin. Although the function of mesothelin is unknown, it may play





a role in cellular adhesion and is present on mesothelium,





mesothellomas, and ovarian cancers.


AA214228
Hs.127751:21, Hs.78006:5
C20orf180: chromosome 20 open reading frame 180
Region of high similarity to tyrosine-phosphorylated protein DOK1
2.99


M31126
Hs.272620:1
PSG9: pregnancy specific beta-1-glycoprotein 9
Pregnancy, extracellular, plasma glycoprotein. Member of the
2.82





pregnancy-specific glycoprotein (PSG) and CEA families.


U62801
Hs.79361:65
KLK6, kallikrein 6 (neurosin, zyme)
Serine type peptidase, pathogenesis. Neurosin (protease M, zyme); a
2.77





sarine protease that cleaves amyloid precursor protein (APP). Growing





evidence suggests that many kallikreins are implicated in carcinogenesis





and some have potential as novel cancer and other disease biomarkers.


AK001536
Hs.285803:6
Hamo sapiens cDNA FLJ12852 fis, clone
Function unknown
2.73




NT2RP2003445


NM_014767
Hs.74583:140
KIAA0275: KIAA0275 gene product
Function unknown
2.72


NM_000699
Hs.75733:129, Hs.278399:100, Hs.274376:1
AMY2A: amylase, alpha 2A; pancreatic
Alpha-amylase, extracellular space, carbohydrate metabolism.
2.71





Pancreatic alpha-amylase 2A (1,4-alpha-D-glucan glucanohydrolase);





cleaves internal a-1,4 bonds between glucose monomers to digest





starch.


AA430348
Hs.288837:40

Homo sapiens cDNA FLJ2927 fis, clone

Function unknown
2.69




NT2RP2004743


X51630
Hs.1145:22, Hs.296851:1
WT1, Wilms tumor 1
Nucleus, transcription factor, transcription regulation. 4 Zn finger
2.58





domains. Functions in kidney and gonad proliferation and differentiation.





Mutations in this gene are associated with the development of Wilms





tumors in the kidney or with abnormalities of the genitourinary tract.


BE393948
Hs.50915:17
KLK5, kallikrein 5
Serine type peptidase, epidermal differentiation, extracellular space.
2.34





Stratum corneum tryptic enzyme (kallikrein-like protein); may function in





epidermal stratum corneum desquamation and turnover. Expression in





prostate cancer negatively correlated with cancer aggressiveness





(Yousef 2002)


NM_002776
Hs.69423:46
KLK10, kallikrein 10
Putative serine protease. Expressed in normal breast tissue and benign
2.24





lesions, with loss of expression during tumor progression (Dhar 2001).





SNPs associated with prostate, breast, testicular, and ovarian cancers





(Bharaj 2002).


NM_000954
Hs.8272:294
PTGDS: prostaglandin D2 synthase (21 kD, brain)
Membrane, prostaglandin-D synthase. Glutathione-independent
2.15





prostaglandin D2 synthase; membrane associated, catalyzes synthesis





of prostaglandin D; member of the lipocalin family of transporters.


AB029000
Hs.70823:109, Hs.297970:48
KIAA1077: sulfatase FP
Function unknown
2.04


AL044315
Hs.173094:70
KIAA1750; KIAA1750 protein
Function unknown
0.95


AA334592
Hs.79914:337
LUM: lumican
Vision, proteoglycan, extracellular matrix, cartilage condensation,
0.93





extracellular matrix glycoprotein. Member of the specialized collagens





and SLRP_family


S79895
Hs.83942:248
CTSK: cathepsin K (pycnodysostosis)
Lysosome, cathepsin K, cysteine-type peptidase, proteolysis and
0.91





peptidolysis. Cathepsin K (cathepsin O), a cysteine (thiol) protease;





involved in bone remodeling and reabsorption


AI091195
Hs.65029:120

Homo sapiens cDNA clone IMAGE: 1566910 3′, mRNA

Function unknown
0.91




sequence


AF026692;
Hs.105700:83, Hs.278611:3
SFRP4: secreted frizzled-related protein 4
Member of the SFRP family that contains a cysteine-rich domain
0.73


NM_003014


homologous to the putative Wnt-binding site of Frizzled proteins. SFRPs





act as soluble modulators of Wnt signaling. The expression of SFRP4 in





ventricular myocardium correlates with apoptosis related gene





expression.


AI683243
Hs.97258:15
ESTs, Moderately similar to S29539 ribosomal protein
Function unknown
−2.96




L13a, cytosolic


AI267700
Hs.111128:7

Homo sapiens, clone IMAGE: 4106329, mRNA

Function unknown
−5.71


AA291377
Hs.50831:23

Homo sapiens Ly-6 antigen/uPA receptor-like domain-

Function unknown
−6.78




containing protein mRNA, complete cds


AI420213
Hs.149722.3
cDNA clone IMAGE: 2094208 3′, mRNA sequence
Function unknown
−8.52


AJ245671
Hs.12844:73
EGFL6, EGF-like-domain; multiple 6
Cell cycle, oncogenesis, integrin ligand, extracellular space. Member of
−9.44





the epidermal growth factor (EGF) repeat superfamily; contains an EGF-





like-domain. Expressed early during development, and its expression





has been detected in lung and meningioma tumors.


AB018305
Hs.5378:149
SPON1, spondin 1, (f-spondin) extracellular matrix
Extracellular matrix protein. Very strongly similar to rat F-spondin
−12.55




protein
(Rn.7546); may have a role in the growth and guidance of axons.


AW872527
Hs.59761:19
ESTs; Weakly similar to DAP1_HUMAN DEATH-
Function unknown
−14.17




ASSOCIATED PROTEIN 1


AF129755
Hs.117772:9, Hs.88474:1

Homo sapiens prostaglandin endoperoxide H

Function unknown
−21.34




synthase-1 mRNA, partial 3′ untranslated region.


AI023799
Hs.163242:5

Homo sapiens cDNA clone IMAGE: 1655725 3′ similar

Function unknown
−41.34




to contains MER20.t2 MER20 repetitive element;,




mRNA sequence
















TABLE 3










Preferred diagnostic and prognostic markers for detecting ovarian cancer or a recurrence thereof


or survival of a subject suffering from ovarian cancer














Unigene



Chromosome
P


Accession Number
Mapping
Gene Name
Function
SEQ ID NO:
Location
value










A. DOWN-REGULATED GENES













AI631024;
Hs.24948:32;
SNCAIP, synuclein, alpha
Cytoplasm, pathogenesis, protein binding. Synphilin-1;
SEQ ID NO: 1 (DNA)
5q23.2
0


NM_005460
Hs.300445:4
interacting protein (synphilin)
promotes formation of cytosolic inclusions in neurons
SEQ ID NO: 2 (PRT)





(SNCAIP). Synuclein alpha interacting protein contains





several protein-protein interaction domains and





interacts with alpha synuclein in neurons. Mutations of





SNCAIP have been linked to Parkinson disease.


NM_002387
Hs.1345:5
MCC, mutated in
Receptor, signal transduction, tumor suppressor.
SEQ ID NO: 3 (DNA)
5q22.2
0




colorectal cancers
Similar to the G protein-coupled m3 muscarinic
SEQ ID NO: 4 (PRT)





acetylcholine receptor. MCC is a candidate for the





putative colorectal tumor suppressor gene. The MCC





gene product are involved in early stages of colorectal





neoplasia in both sporadic and familial tumors.


AI420582;
Hs.136164:23
SE20-4,
Cutaneous T-cell lymphoma-associated tumor antigen
SEQ ID NO: 5 (DNA)
unmapped
0


NM_022117

cutaneous T-cell
se20-4se20-4; differentially expressed nucleolar TGF-beta1 target protein
SEQ ID NO: 6 (PRT)




lymphoma-
(DENTT); also known as CDA1




associated tumor




antigen se20-




4se20-4







B. UP-REGULATED GENES













BC006428;
Hs.15093:210, Hs.290304:1
HSPC195,

Homo sapiens cDNA FLJ10920 fis, clone

SEQ ID NO: 7 (DNA)
5q31.2
0


NM_016463

hypothetical
OVARC1000384-resourcerer.
SEQ ID NO: 8 (PRT)




protein HSPC195


NM_017697
Hs.24743:94
FLJ20171,
contains 3 RNA recognition motifs
SEQ ID NO: 9 (DNA)
8q22.1
0




hypothetical

SEQ ID NO: 10 (PRT)




protein FLJ20171


AW630088;
Hs.76550:164
MAL2
Mal2 T-cell differentiation protein: found thru interaction
SEQ ID NO: 11 (DNA)
8q24.12
0


NM_001306


with TPD52 which is overexpressed in breast cancer; 4
SEQ ID NO: 12 (PRT)





TM are involved in vesicle transport


NM_015238
Hs.21543:36
KIAA0869,
Function unknown
SEQ ID NO: 13 (DNA)
5q34
0.0002




KIAA0869 protein;

SEQ ID NO: 14 (PRT)




KIBRA


AA284679
Hs.25640:264, Hs.5372:2
CLDN3, claudin 3
Integral plasma membrane protein, pathogenesis, tight
SEQ ID NO: 15 (DNA)
7q11.23
0.0004





junction, transmembrane receptor. Member of the
SEQ ID NO: 16 (PRT)





claudin family of integral membrane proteins; receptor





for Clostridium perfringens enterotoxin;


NM_022454
Hs.97984:22
SOX17, SRY (sex
Likely ortholog of mouse SRY-box containing gene 17;
SEQ ID NO: 17 (DNA)
8q11.23
0.0005




determining
alias SOX17
SEQ ID NO: 18 (PRT)




region Y)-box 17


NM_005682
Hs.6527:201
GPR56, G
cell adhesion, cell-cell signalling, G-protein linked
SEQ ID NO: 19 (DNA)
16q13
0.0012




protein-coupled
receptor, integral plasma membrane protein, G-protein
SEQ ID NO: 20 (PRT)




receptor 56
linked receptor protein signalling pathway. Member of





the G protein-coupled receptor family; similar to secretin





and calcitonin receptors. 7 transmembrane domains, a





mucin-like domain and cysteine box in the N-terminal





region. Expressed in range of tissues, highest levels in





thyroid, selectively within the monolayer of cuboidal





epithelial cells of the smaller, more actively secreting





follicles of human thyroid. Differentially expressed in





melanoma cell lines with different metastatic potential





(Zendman et al 1999).


NM_001307
Hs.278562:101
CLDN7, claudin 7
Integral membrane protein, tight junction. Similar to
SEQ ID NO: 21 (DNA)
17p13.1
0.0016





murine Cldn7;
SEQ ID NO: 22 (PRT)


NM_014736
Hs.81892:95
KIAA0101 gene
function unknown; no signficant hits with Superfamily
SEQ ID NO: 23 (DNA)
15q31
0.0025




product

SEQ ID NO: 24 (PRT)


BE184455;
Hs.251754:128, Hs.245742:1
SLPI, secretory
Plasma protein, proteinase inhibitor. Secreted inhibitor
SEQ ID NO: 25 (DNA)
20q13.12
0.0034


NM_003064

leukocyte
which protects epithelial tissues from serine proteases.
SEQ ID NO: 26 (PRT)




protease inhibitor
Found in various secretions including seminal plasma,




(antileukoproteinase)
cervical mucus, and bronchial secretions, has affinity for





trypsin, leukocyte elastase, and cathepsin G. Its





inhibitory effect contributes to the immune response by





protecting epithelial surfaces from attack by





endogenous proteolytic enzymes; the protein is also





thought to have broad-spectrum anti-biotic activity.


NM_013994
Hs.75562:147
DDR1, discoidin
Cell adhesion, integral plasma membrane protein,
SEQ ID NO: 27 (DNA)
6p21.33
0.0055




domain receptor
transmembrane receptor, protein tyrosine kinase.
SEQ ID NO: 28 (PRT)




family, member 1
Epithelial-specific receptor protein tyrosine kinase; are





involved in cell adhesion; has putative discoidin motifs





in extracellular domain. DDR1 (CD167a) is a RTK that





is widely expressed in normal and transformed





epithelial cells and is activated by various types of





collagen.


NM_001067
Hs.156348:184, Hs.270810:2
TOP2A,
DNA binding, DNA topoisomerase (ATP-hydrolyzing),
SEQ ID NO: 29 (DNA)
17q21.2
0.006




topoisomerase
nucleus. DNA topoisomerase II alpha; may relax DNA
SEQ ID NO: 30 (PRT)




(DNA) II alpha
torsion upon replication or transcription. Involved in




(170 kD)
processes such as chromosome condensation,





chromatid separation, and the relief of torsional stress





that occurs during DNA transcription and replication.





Catalyzes the transient breaking and rejoining of two





strands of duplex DNA. The gene encoding this enzyme





functions as the target for several anticancer agents





and a variety of mutations in this gene have been





associated with the development of drug resistance.





Reduced activity of this enzyme may also play a role in





ataxia-telanglectasia.


BE386983;
Hs.343214
CKLFSF7;
chemokine-like factor gene superfamily; transmb 4
SEQ ID NO: 31 (DNA)
3p23
0.0131


NM_138410

chemokine-like
superfamily
SEQ ID NO: 32 (PRT)




factor super family 7


AF098158;
Hs.9329:152
C20orf1,
ATP binding, GTP binding, cell proliferation, mitosis,
SEQ ID NO: 33 (DNA)

0.0183


NM_012112

chromosome 20
nucleus spindle. Proliferation-associated nuclear
SEQ ID NO: 34 (PRT)




open reading
protein; associates with the spindle pole and mitotic




frame 1
spindle during mitosis


NM_001769
Hs.1244:227, Hs.230559:1,
CD9: CD9 antigen
Plasma membrane, integral plasma membrane protein.
SEQ ID NO: 35 (DNA)
12p13.31
0.0006



Hs.242020:1
(p24)
Member of the transmembrane 4 superfamily (TM4SF);
SEQ ID NO: 36 (PRT)





may mediate platelet activation and aggregation. Cell





surface glycoprotein that is known to complex with





integrins and other transmembrane 4 superfamily





proteins.


NM_020859
Hs.278628:52
ShrmL, Shroom-
Amiloride-sensitive sodium channel (weakly similar to
SEQ ID NO: 37 (DNA)

0.0074




related protein

Mus musculus PDZ domain actin binding protein)

SEQ ID NO: 38 (PRT)




(KIAA1481




protein)


NM_004433
Hs.166096:170
ELF3, E74-like
Embryogenesis and morphogenesis, transcription co-
SEQ ID NO: 39 (DNA)
1q32.1
0.0004




factor 3 (ets
activator, transcription factor, transcription from Pol II
SEQ ID NO: 40 (PRT)




domain
promoter. ETS domain transcriptional activator;




transcription
activates expression of epithelial cell specific genes.




factor, epithelial-




specific)


AI791905;
Hs.95549:147, Hs.229556:1
FLJ20273, RNA-
Contains four RNA recognition motifs (RRM, RBD, or
SEQ ID NO: 41 (DNA)

0.0007


NM_019027

binding protein
RNP)
SEQ ID NO: 42 (PRT)


X69699;
Hs.73149:72, Hs.213008:1
PAX8, paired box
Histogenesis and organogenesis, embryogenesis and
SEQ ID NO: 43 (DNA)
2q13
0.0009


NM_013952

gene 8
morphogenesis, thyroid-stimulating hormone receptor,
SEQ ID NO: 44 (PRT)





transcription factor. Member of the paired domain family





of nuclear transcription factors; are involved in the





ribosome assembly, required for normal thyroid





development, PAX genes play critical roles during fetal





development and cancer growth.


AI301558
Hs.290801:35,
EST
Function unknown
SEQ ID NO: 45 (DNA)

0.0044



Hs.356228


NM_018000
Hs.79741:18
FLJ10116,
Function unknown
SEQ ID NO: 46 (DNA)
2q35
0.0051




hypothetical

SEQ ID NO: 47 (PRT)




protein FLJ10116


NM_144724
Hs.124740:18
hypothetical
59% identity to human Zinc finger protein 91
SEQ ID NO: 48 (DNA)
5q13.12
0.0051




protein FLJ30532

SEQ ID NO: 49 (PRT)


AF111856;
Hs.105039:48
SLC34A2, solute
SLC34A2: solute carrier family 34 (sodium phosphate),
SEQ ID NO: 50 (DNA)
4p15.2
0.0121


NM_006424

carrier family 34
member 2; contains 8 predicted TMs and a cysteine-
SEQ ID NO: 51 (PRT)




(sodium
rich N-terminal region. Type 2 sodium-dependent




phosphate),
phosphate transporter. member of the renal type II co-




member 2
transporter family.


AW959311
Hs.87019:8;
EST
probable serine/threonine protein kinase; KIAA0537
SEQ ID NO: 52 (DNA)
1q32.1
0.0251



Hs.172012
DKFZp434J037


AF111713
Hs.286218:64
JAM1, junctional
Cell motility, inflammatory response, intercellular
SEQ ID NO: 53 (DNA)

0.0261




adhesion
junction. Role in the regulation of tight junction
SEQ ID NO: 54 (PRT)




molecule
assembly in epithelia. Ligation of JAM is required for





reovirus-induced activation of NF-kappa-B and





apoptosis. Role in lymphocyte homing.


AU076611;
Hs.154672:123
MTHFD2,
Electron transporter, methenyltetrahydrofolate
SEQ ID NO: 55 (DNA)
2p13.1
0.0342


NM_006636

methylene
cyclohydrolase, mitochondrion, encodes a nuclear-
SEQ ID NO: 56 (PRT)




tetrahydrofolate
encoded mitochondrial bifunctional enzyme with




dehydrogenase
methylenetetrahydrofolate dehydrogenase and




(NAD+
methenyltetrahydrofolate cyclohydrolase activities. may




dependent);
provide formyltetrahydrofolate for formylmethionyl tRNA




methenyltetrahydrofolate
synthesis; involved in initiation of mitochondrial protein




cyclohydrolase
synthesis.







C. UP-REGULATED GENES IN MUCINOUS OVARIAN CANCER ONLY













AA584890;
Hs.5302:132
LGALS4, lectin,
Lectin, cytosol, cell adhesion, plasma membrane. Binds
SEQ ID NO: 57 (DNA)
19q13.2
0.0001


NM_006149

galactoside-
to beta galactoside, involved in cell adhesion, cell
SEQ ID NO: 58 (PRT)




binding, soluble, 4
growth regulation, inflammation, immunomodulation,




(galectin 4)
apoptosis and metastasis; member of a family of





lectins. LGALS4 is an S-type lectin that is strongly





underexpressed in colorectal cancer.



Hs.89436:50
CDH17, cadherin
Cell adhesion, integral plasma membrane protein,
SEQ ID NO: 59 (DNA)
8q22.1
0.0172




17, LI cadherin
membrane fraction, small molecule transport,
SEQ ID NO: 60 (PRT)




(liver-intestine)
transporter. Member of the cadherin family of calcium-





dependent glycoproteins; facilitates uptake of peptide-





based drugs, may mediate cell-cell interactions.





Component of the gastrointestinal tract and pancreatic





ducts, intestinal proton-dependent peptide transporter in





the first step in oral absorption of many medically





important peptide-based drugs.


NM_005588
Hs.179704
MEP1A, meprin A
metalloprotease located apically and secreted by
SEQ ID NO: 61 (DNA)
6p12
0.01




alpha, PABA
epithelial cells in normal colon; degrades broad range of
SEQ ID NO: 62 (PRT)




peptide hydrolase
ECM components in vitro; proposed role in tumour





progression by facilitating migration, intravasation and





metastasis







D. PROGNOSTIC MARKERS FOR SURVIVAL OR RECURRENCE













NM_015092
Hs.278428
DD5; EDD

Homo sapiens progestin induced protein (DD5), mRNA.

SEQ ID NO: 63 (DNA)

0.00





EDD; Soluble fraction, cell proliferation, ubiquitin—
SEQ ID NO: 64 (PRT)





protein ligase, ubiquitin conjugating enzyme, ubiquitin-





dependent protein degradation. Member of the HECT





family of proteins; may function as an E3 ubiquitin-





protein ligase. This gene is localized to chromosome





8q22, a locus disrupted in a variety of cancers. This





gene potentially has a role in regulation of cell





proliferation or differentiation.


BE465867;
Hs.197751:66
DAAM1
dishevelled associated activator of morphogenesis 1
SEQ ID NO: 65 (DNA)
8q22.3
0.04


NM_014992


The protein encoded by this gene contains FH domains
SEQ ID NO: 66 (PRT)





and belongs to a novel FH protein subfamily implicated





in cell polarity, thought to function as a scaffolding





protein.


AA381553;
Hs.198253:21
HLA1QA
major histocompatibility complex, class II, DQ alpha 1
SEQ ID NO: 67 (DNA)
14q23.1
0.00


NM_002122


Pathogenesis, class II major histocompatibility complex
SEQ ID NO: 68 (PRT)





antigen. Alpha 1 chain of HLA-DQ1 class II molecule (la





antigen); complex binds peptides and presents them to





CD4+ T lymphocytes|Proteome


AF026692;
Hs.105700:83, Hs.278611:3
SFRP4: secreted
Member of the SFRP family that contains a cysteine-
SEQ ID NO: 69 (DNA)
6p21.3
0.73


NM_003014

frizzled-related
rich domain homologous to the putative Wnt-binding
SEQ ID NO: 70 (PRT)
7p14




protein 4
site of Frizzled proteins. SFRPs act as soluble





modulators of Wnt signaling. The expression of SFRP4





in ventricular myocardium correlates with apoptosis





related gene expression.


AW015534;
Hs.217493
ANXA2, annexin
Annexin II (lipocortin-2); enhances osteoclast formation
SEQ ID NO: 71 (DNA)
15q21-22
0.00


NM_004039

A2
and bone resorption; member of the annexin protein
SEQ ID NO: 72 (PRT)





family


BE24669;
Hs.345728
SOCS3
STAT induced STAT-inhibitor 3; suppressor of cytokine
SEQ ID NO: 73 (DNA)
17q25.3
0.02


NM_003955


signalling 3; suppression of IL-6 mediated signalling
SEQ ID NO: 74 (PRT)


AI677897;
Hs.76640
RGC32
RGC32, hypothetical protein, unknown function
SEQ ID NO: 75 (DNA)
13q13.3
0.04


NM_014059



SEQ ID NO: 76 (PRT)


AA829286;
Hs.332053
SAA1, serum
Serum amyloid A1; high density lipoapoprotein; role in
SEQ ID NO: 77 (DNA)
11p15.1
0.04


NM_000331

amyloid A1
cholesterol metabolism; inflammatory response
SEQ ID NO: 78 (PRT)


AA243499;
Hs.104800
FLJ10134,
Unknown
SEQ ID NO: 79 (DNA)
3q12.3
0.01


NM_018004

hypothetical

SEQ ID NO: 80 (PRT)




protein


M88849;
Hs.323733
GJB2, gap
Cellular gap junctions; mutations cause some forms of
SEQ ID NO: 81 (DNA)
13q11-12
0.00


NM_004004

junction protein
deafness
SEQ ID NO: 82 (PRT)




beta2; connexin




26


NM_002514
Hs.235935
NOV1;
Role in cell adhesion and migration in endothelial cells;
SEQ ID NO: 83 (DNA)
8q24.1
0.01




Nephroblastoma
promotes cell survival
SEQ ID NO: 84 (PRT)




overexpressed




gene
















TABLE 4










Correlation of expression between normal ovarian surface epithelium


(OSE), non-invasive tumors (borderline, BL) and ovarian cancer


(CA) as determined by ANOVA
















E-

Ep-




CA125
MUC-1
cadherin
CLDN3
CAM
SOX17

















OSE vs IC
<0.0001
<0.0001
0.7251
0.6132
0.1573
0.0854


OSE vs. BL
0.1765
<0.0001
0.0307
0.3633
0.0005
0.2287


OSE vs. CA
0.5443
<0.0001
0.1687
0.0008
<0.0001
0.6900


IC vs. BL
<0.0001
<0.0001
0.1116
0.7849
0.0913
0.2530


IC vs. CA
<0.0001
0.2707
0.4147
0.0071
0.0002
0.0544


BL vs. CA
0.0001
<0.0001
0.0615
<0.0001
0.0011
0.0152
















TABLE 5










Correlation of gene expression with patient outcome (univariate analysis ie., expression


alone without the influence of covariates)


Univariate analysis for clinicopathological variables and CLDN3, Ep-CAM, SOX17,


CA125, MUC1 and E-cadherin immunoreactivity with survival and relapse in 156


patients with epithelial ovarian cancer










Disease Specific Survival
Relapse Free Survival












Univariate

Univariate



Variable
Hazards ratio (95% CI)
p-value
Hazards ratio (95% CI)
p-value














Pathological tumor stage






Stage 1-3bvs. 3c-4b
 5.89 (3.214-10.79)
<0.0001
7.37 (3.26-16.63)
<0.0001


Tumor grade


BL and G1vs. G2 and G3
 5.508 (2.745-11.052)
<0.0001
7.02 (2.76-17.82)
<0.0001


Age


<50 vs. >=50
0.533 (0.288-0.988)
0.0458
0.62 (0.29-1.33) 
0.2221


Residual Disease


RD<1 cm vs. >=1 cm
4.192 (2.671-6.580)
<0.0001
4.17 (2.30-7.55) 
<0.0001


CA125 level at diagnosis


CA125 <500vs. >500 U/ml
1.843 (1.102-3.080)
0.0197
2.292 (1.19-4.40) 
0.0128


Performance Status


PS<1 vs. >1
0.270 (0.133-0.549)
0.0003
0.53 (0.16-1.74) 
0.2965


CLDN3 expression


Membranous Score 0vs. >0
2.794 (1.012-7.718)
0.0474
2.521 (0.908-6.998)
0.0758


Membranous Score <1vs. >1
1.309 (0.763-2.246)
0.3285
1.952 (1.103-3.457)
0.0217


Ep-CAM expression


Membranous Score <1vs. >1
1.460 (0.809-2.634)
0.2093
2.041 (0.997-4.177)
0.0509


Membranous Score <2vs. >2
1.041 (0.634-1.711)
0.873
1.449 (0.845-2.487)
0.1779


SOX17 expression


Nuclear membranous Score 0vs. >0
0.839 (0.514-1.368)
0.481
1.311 (0.728-2.358)
0.3667


Nuclear membranous Score <1vs. >1
1.407 (0.615-3.218)
0.4183
1.037 (0.380-2.829)
0.9437


CA125 expression


Membranous apical Score 0vs. >0
2.581 (1.393-4.781)
0.0026
2.725 (1.218-6.093)
0.0146


Membranous apical Score <1vs. >1
1.637 (1.045-2.564)
0.0313
1.298 (0.731-2.307)
0.3737


MUC1 expression


Membranous apical Score 0vs. >0
 2.479 (0.343-17.898)
0.368
NA


Membranous apical Score <1vs. >1
 3.745 (1.176-11.926)
0.0254
 6.432 (1.562-26.483)
0.0099


Membranous apical Score <2vs. >2
1.814 (0.898-3.664)
0.0969
3.893 (1.552-9.766)
0.0038


E-cadherin expression


Membranous Score 0vs. >0
0.806 (0.493-1.318)
0.3892
0.837 (0.477-1.467)
0.5341


Membranous Score <1vs. >1
1.331 (0.532-3.333)
0.5411
0.847 (0.263-2.731)
0.7814


Membranous Score <2vs. >2
0.593 (0.082-4.284)
0.6041
0.913 (0.125-6.646)
0.9284
















TABLE 6










Correlation of gene expression with patient outcome (multivariate analysis ie looking


at expression incorporating the influence of covariates)


Multivariate analysis for univariate significant clinicopathological variables and CLDN3,


Ep-CAM, SOX17, CA125, MUC1 and E-cadherin immunoreactivity with survival


and relapse in 156 patients with epithelial ovarian cancer










Disease Specific Survival
Relapse Free Survival












Multivariate

Univariate



Variable
Hazards ratio (95% CI)
p-value
Hazards ratio (95% CI)
p-value














Pathological tumor stage






Stage 1-3b vs. 3c-4b
 5.66 (2.467-13.012)
<0.0001
 5.192 (1.860-14.496)
0.0017


Tumor grade


BL and G1 vs. G2 and G3
 4.919 (2.080-11.633)
0.0003
 7.989 (2.385-26.760)
0.0008


Age


<50 vs. >=50
0.951 (0.482-1.877)
0.8853


Residual Disease


RD<1 cm vs. >=1 cm
2.974 (1.783-4.959)
<0.0001
2.779 (1.433-5.393)
0.0025


CA125 level at diagnosis


CA125 <500 vs. >500 U/ml
1.148 (0.625-2.109)
0.6563
1.289 (0.659-2.520)
0.4587


Performance Status


PS<1 vs. >1
0.286 (0.136-0.601)
0.0009


CLDN3 expression


Membranous Score 0 vs. >0
1.165 (0.325-4.183)
0.8145


Membranous Score <1 vs. >1


0.953 (0.473-1.919)
0.8918


CA125 expression


Membranous apical Score 0 vs. >0
0.917 (0.415-2.025)
0.8302
0.693 (0.271-1.768)
0.4427


Membranous apical Score <1 vs. >1
1.664 (0.976-2.837)
0.0612


MUC1 expression


Membranous apical Score 0 vs. >0


Membranous apical Score <1 vs. >1
0.678 (0.255-1.804)
0.4361


Membranous apical Score <2 vs. >2









Claims
  • 1. (canceled)
  • 2. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a modified level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83; (iii) a sequence that is at least about 80% identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 46, 48, 50, 52, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81 and 83; (iv) a sequence that encodes an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 47, 49, 51, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 3. (canceled)
  • 4. The method of claim 2 wherein the hybridization is enhanced in the sample from the subject being tested compared to the hybridization obtained for a sample from a control subject not having ovarian cancer.
  • 5. The method of claim 2 wherein the hybridization is reduced in the sample from the subject being tested compared to the hybridization obtained for a sample from a control subject not having ovarian cancer.
  • 6. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an enhanced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200; (iii) a sequence that is at least about 80% identical to (i) or (ii); (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 7. (canceled)
  • 8. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a reduced level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200; (iii) a sequence that is at least about 80% identical to (i) or (ii); (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 9. (canceled)
  • 10. The method of claim 2 wherein the ovarian cancer that is diagnosed is an epithelial ovarian cancer.
  • 11. The method of claim 2 wherein the ovarian cancer that is diagnosed is selected from the group consisting of serous ovarian cancer, non-invasive ovarian cancer, mixed phenotype ovarian cancer, mucinous ovarian cancer, endometrioid ovarian cancer, clear cell ovarian cancer, papillary serous ovarian cancer, Brenner cell and undifferentiated adenocarcinoma.
  • 12. The method according to claim 11 wherein the ovarian cancer that is diagnosed is selected from the group consisting of serous ovarian cancer, mucinous ovarian cancer, endometrioid ovarian cancer and clear cell ovarian cancer.
  • 13. A method of diagnosing a serous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein a modified level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a serous ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305; (iii) a sequence that is at least about 80% identical to (i) or (ii); (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 14. A method of diagnosing a mucinous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a mucinous ovarian cancer, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—006149, AA315933, U47732, NM—005588, AW503395, NM—004063, AI073913, AI928445, NM—022454, W40460, AA132961 and AF111856; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—006149, AA315933, U47732, NM—005588, AW503395, NM—004063, AI073913, AI928445, NM—022454, W40460, AA 132961 and AF111856; (iii) a sequence that is at least about 80% identical to (i) or (ii); (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—006149, AA315933, U47732, NM 005588, AW503395, NM—004063, AI073913, AI928445, NM—022454, W40460, AA132961 and AF111856; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 15. The method of claim 14 wherein the nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from SEQ ID NO: 57 or 59 or 61; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from SEQ ID NO: 57 or 59 or 61; (iii) a sequence that is at least about 80% identical to SEQ ID NO: 57 or 59 or 61; (iv) a sequence that encodes the amino acid sequence set forth in SEQ ID NO: 58 or 60 or 62; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 16. The method of claim 2 comprising performing a PCR reaction.
  • 17. The method of claim 21 comprising performing a nucleic acid hybridization.
  • 18. (canceled)
  • 19. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a modified level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence having at least about 80% identity to a sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 47, 49, 51, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82 and 84.
  • 20. (canceled)
  • 21. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 1 or 2 other than a nucleic acid having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200.
  • 22. (canceled)
  • 23. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a reduced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has an ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of NM—022117, NM—005460, NM—002387, AI745249 and AI694200.
  • 24. (canceled)
  • 25. The method of claim 19 wherein the ovarian cancer that is diagnosed is an epithelial ovarian cancer.
  • 26. The method of claim 19 wherein the ovarian cancer that is diagnosed is selected from the group consisting of serous ovarian cancer, non-invasive ovarian cancer, mixed phenotype ovarian cancer, mucinous ovarian cancer, endometrioid ovarian cancer, clear cell ovarian cancer, papillary serous ovarian cancer, Brenner cell and undifferentiated adenocarcinoma.
  • 27. (canceled)
  • 28. A method of diagnosing a serous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a modified level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a serous ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 2 or as set forth in Table 1 and having an Accession Number selected from the group consisting of: U62801, D49441, X51630, And AB018305.
  • 29. A method of diagnosing a mucinous ovarian cancer in a human or animal subject being tested said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein a reduced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a mucinous ovarian cancer, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—006149, AA315933, U47732, NM—005588, AW503395, NM—004063, AI073913, AI928445, NM—022454, W40460, AA132961 and AF111856.
  • 30. (canceled)
  • 31. A method of detecting an ovarian cancer-associated antibody in a biological sample the method comprising contacting the biological sample with a polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3, wherein the polypeptide specifically binds to the ovarian cancer-associated antibody.
  • 32. The method according to claim 2 wherein the biological sample is contacted with a plurality of nucleic acid probes.
  • 33. The method of claim 2 wherein the subject being tested is a patient undergoing a therapeutic regimen to treat ovarian cancer.
  • 34. The method of claim 2 wherein the subject being tested is a subject suspected of having ovarian cancer.
  • 35. A method of monitoring the efficacy of a therapeutic treatment of ovarian cancer, the method comprising: (i) providing a biological sample from a patient undergoing the therapeutic treatment; and (ii) determining the level of a ovarian cancer-associated transcript in the biological sample by contacting the biological sample with a polynucleotide that selectively hybridizes to a sequence having at least about 80% identity to a sequence as shown in any one of Tables 1-3, thereby monitoring the efficacy of the therapy.
  • 36. (canceled)
  • 37. A method of monitoring the efficacy of a therapeutic treatment of ovarian cancer, the method comprising: (i) providing a biological sample from a patient undergoing the therapeutic treatment; and (ii) determining the level of a ovarian cancer-associated antibody in the biological sample by contacting the biological sample with a polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3, wherein the polypeptide specifically binds to the ovarian cancer-associated antibody, thereby monitoring the efficacy of the therapy.
  • 38. (canceled)
  • 39. A method of monitoring the efficacy of a therapeutic treatment of ovarian cancer, the method comprising: (i) providing a biological sample from a patient undergoing the therapeutic treatment; and (ii) determining the level of a ovarian cancer-associated polypeptide in the biological sample by contacting the biological sample with an antibody, wherein the antibody specifically binds to a polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3, thereby monitoring the efficacy of the therapy.
  • 40-43. (canceled)
  • 44. A method of determining the likelihood of survival of a subject suffering from an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—003014, AA046217, NM—015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM—014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM—002776, BE261944, NM—006379, AI002238, X81789, NM—002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM—001955, AI680737, AI752666, AA505445, BE246649, and NM—003955; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—003014, AA046217, NM—015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM—014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM—002776, BE261944, NM—006379, AI002238, X81789, NM—002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM—001955, AI680737, AI752666, AA505445, BE246649, and NM—003955; (iii) a sequence that is at least about 80% identical to (i) or (ii); (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—003014, AA046217, NM—015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM—014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM—002776, BE261944, NM—006379, AI002238, X81789, NM—002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM—001955, AI680737, AI752666, AA505445, BE246649, and NM—003955; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 45. (canceled)
  • 46. A method of determining the likelihood of survival of a subject suffering from an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has has a poor probability of survival, and wherein said antibody binds to a polypeptide comprising an amino acid sequence comprising at least about 10 contiguous amino acid residues of a sequence encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: NM—003014, AA046217, NM—015902, T83882, AB040888, AA628980, AI623351, AW614420, AA243499, AF251237, AI970797, AF145713, X78565, T97307, BE243845, AW068302, AL133561, BE313555, X07820, AI973016, AF084545, U41518, Z11894, AW138190, BE086548, W47196, AI796870, X02761, AW968613, AW972565, AF045229, AW953853, U52426, F06700, AI798863, H52761, BE546947, AU076643, U20536, AA581602, AJ245210, X65965, AI806770, BE386490, AW581992, U77534, AL034417, L10343, AW518944, W28729, AI640160, U11862, AW295980, X59135, BE466173, AI354722, M90464, AA829286, AI333771, BE465867, NM—014992, BE616902, AA430373, R27430, BE387335, AW264102, AW952323, AA088177, BE614567, AL079658, NM—002776, BE261944, NM—006379, AI002238, X81789, NM—002122, AB001914, AA311919, AI381750, AA292998, BE439580, AI677897, N72403, BE003054, AL035588, AI080491, AW770994, H24177, AF146761, NM—001955, AI680737, AI752666, AA505445, BE246649, and NM—003955.
  • 47. (canceled)
  • 48. A method of determining the likelihood of survival of a subject suffering from a serous ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid comprising the nucleotide sequence set forth in SEQ ID NO: 71 or 73; (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid comprising the nucleotide sequence set forth in SEQ ID NO: 71 or 73; (iii) a sequence that is at least about 80% identical to (i) or (ii) and encoding an sFRP protein or a SOCS3 protein; (iv) a sequence that encodes a polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 72 or 74; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 49. A method of determining the likelihood of survival of a subject suffering from a serous ovarian cancer, said method comprising contacting a biological sample from said subject being tested with an antibody for a time and under conditions sufficient for an antigen-antibody complex to form and then detecting the complex wherein an enhanced level of the antigen-antibody complex for the subject being tested compared to the amount of the antigen-antibody complex formed for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein said antibody binds to an sFRP polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 72 or a SOCS3 polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 74.
  • 50. A method of determining the likelihood of survival of a subject suffering from a serous ovarian cancer, said method comprising contacting a biological sample from said subject being tested with at least two antibodies for a time and under conditions sufficient for antigen-antibody complexes to form and then detecting the complexes wherein an enhanced level of the antigen-antibody complexes for the subject being tested compared to the amount of the antigen-antibody complexes formed for a control subject not having ovarian cancer indicates that the subject being tested has a poor probability of survival, and wherein one antibody binds to an sFRP polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 72 and wherein one antibody binds to a SOCS3 polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 74.
  • 51-53. (canceled)
  • 54. A method of determining the likelihood that a subject will suffer from a recurrence of an ovarian cancer, said method comprising contacting a biological sample from said subject being tested with a nucleic acid probe for a time and under conditions sufficient for hybridization to occur and then detecting the hybridization wherein an elevated level of hybridization of the probe for the subject being tested compared to the hybridization obtained for a control subject not having ovarian cancer indicates that the subject being tested has a high probability of recurrence, and wherein said nucleic acid probe comprises a sequence selected from the group consisting of: (i) a sequence comprising at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM—012317, NM—000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, AA569756, AW138190, AF126245, L10343, NM—002514, AI863735, NM—005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM—005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM—005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM—000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N51357, N80486, NM—000954, NM—005756, NM—016652, R26584, R31178, W05391, W25005, W45393, W68815, X65965, X76732 and Z45051, (ii) a sequence that hybridizes under at least low stringency hybridization conditions to at least about 20 contiguous nucleotides from a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM—012317, NM—000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, AA569756, AW138190, AF126245, L10343, NM—002514, AI863735, NM—005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM—005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM—005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM—000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N51357, N80486, NM—000954, NM—005756, NM—016652, R26584, R31178, W05391, W25005, W45393, W68815, X65965, X76732 and Z45051; (iii) a sequence that is at least about 80% identical to (i) or (ii); (iv) a sequence that encodes a polypeptide encoded by a nucleic acid set forth in Table 1 and having an Accession Number selected from the group consisting of: M86849, AW963419, BE298665, AK000637, BE077546, T97307, R24601, BE090176, AA393907, W28729, BE313754, AW673081, AA356694, L08239, BE397649, NM—012317, NM—000947, AJ250562, AL040183, BE207573, BE564162, BE439580, AW067800, AA569756, AW138190, AF126245, L10343, NM—002514, AI863735, NM—005397, W26391, H15474, U51166, AA243499, AW408807, AI738719, AB040888, BE313077, AI677897, C14898, AI821730, AF007393, H65423, N46243, AA095971, U20350, NM—005756, D19589, AW957446, AW294647, BE159718, AI888490, AA022569, BE147740, AI798863, BE464341, AL080235, AI557212, X75208, AA628980, BE242587, NM—005512, AW953853, AU076611, AW968613, AL353944, BE614149, AA292998, H12912, AA188763, AK000596, AI970797, AW519204, Z42387, AF145713, AA972412, AK001564, AW959861, BE313555, W25005, AI193356, AF111106, AI130740, AA985190, BE221880, AF084545, R26584, AW247380, AA364261, U25849, AF262992, AW342140, AL133572, AI497778, AI745379, U51712, AW375974, AF251237, NM—000636, AA130986, AA216363, AA628980, AA811657, AA897108, AB040888, AF212225, AI089575, AI282028, AI368826, AI718702, AI827248, AK002039, AL109791, AW090198, AW296454, AW445034, AW452948, AW470411, AW885727, AW970859, AW979189, BE165866, BE175582, BE242587, BE271927, BE439580, BE464016, D63216, F34856, M83822, N33937, N49068, N51357, N80486, NM—000954, NM—005756, NM—016652, R26584, R31178, WO5391, W25005, W45393, W68815, X65965, X76732 and Z45051; and (v) a sequence that is complementary to any one of the sequences set forth in (i) or (ii) or (iii) or (iv).
  • 55-59. (canceled)
  • 60. A method for identifying a compound that modulates an ovarian cancer-associated polypeptide, the method comprising: (i) contacting the compound with a ovarian cancer-associated polypeptide, the polypeptide encoded by a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3; and (ii) determining the functional effect of the compound upon the polypeptide.
  • 61. A method for determining a candidate compound for the treatment of ovarian cancer comprising: (i) administering a test compound to a mammal having ovarian cancer or a cell isolated therefrom; (ii) comparing the level of gene expression of a polynucleotide that selectively hybridizes to a sequence at least 80% identical to a sequence as shown in Tables 1-3 in a treated cell or mammal with the level of gene expression of the polynucleotide in a control cell or mammal, wherein a test compound that modulates the level of expression of the polynucleotide is a candidate for the treatment of ovarian cancer.
  • 62. An assay device for use in the diagnosis or prognosis of ovarian cancer, said device comprising a plurality of polynucleotides immobilized to a solid phase, wherein each of said polynucleotides consists of a gene as listed in any one of Tables 1-3.
  • 63. (canceled)
  • 64. An assay device for use in the diagnosis or prognosis of ovarian cancer, said device comprising a plurality of different antibodies immobilized to a solid phase, wherein each of said antibodies binds to a polypeptide listed in Tables 1-3.
  • 65-69. (canceled)
  • 70. A method of diagnosing an ovarian cancer in a human or animal subject being tested said method comprising determining aberrant methylation in a promoter sequence that regulates expression of a tumor suppressor gene in a biological sample from said subject compared to the methylation of the promoter in nucleic acid obtained for a control subject not having ovarian cancer wherein said aberrant methylation indicates that the subject being tested has an ovarian ovarian cancer.
  • 71-73. (canceled)
  • 74. The method according to claim 19 wherein the biological sample is contacted with a plurality of antibodies.
  • 75. The method of claim 19 wherein the subject being tested is a patient undergoing a therapeutic regimen to treat ovarian cancer.
  • 76. The method of claim 19 wherein the subject being tested is a subject suspected of having ovarian cancer.
Priority Claims (1)
Number Date Country Kind
20022951346 Sep 2002 AU national
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/AU03/01166 9/5/2003 WO 5/17/2006